1
|
Rajagopal V, Seiler J, Nasa I, Cantarella S, Theiss J, Herget F, Kaifer B, Schneider M, Helm D, König J, Zarnack K, Diederichs S, Kettenbach AN, Caudron-Herger M. An atlas of RNA-dependent proteins in cell division reveals the riboregulation of mitotic protein-protein interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614981. [PMID: 39386702 PMCID: PMC11463612 DOI: 10.1101/2024.09.25.614981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Ribonucleoprotein complexes are dynamic assemblies of RNA with RNA-binding proteins (RBPs), which can modulate the fate of the RNA molecules from transcription to degradation. Vice versa, RNA can regulate the interactions and functions of the associated proteins. Dysregulation of RBPs is linked to diseases such as cancer and neurological disorders. RNA and RBPs are present in mitotic structures like the centrosomes and spindle microtubules, but their influence on mitotic spindle integrity remains unknown. Thus, we applied the R-DeeP strategy for the proteome-wide identification of RNA-dependent proteins and complexes to cells synchronized in mitosis versus interphase. The resulting atlas of RNA-dependent proteins in cell division can be accessed through the R-DeeP 3.0 database (R-DeeP3.dkfz.de). It revealed key mitotic factors as RNA-dependent such as AURKA, KIFC1 and TPX2 that were linked to RNA despite their lack of canonical RNA-binding domains. KIFC1 was identified as a new interaction partner and phosphorylation substrate of AURKA at S349 and T359. In addition, KIFC1 interacted with both, AURKA and TPX2, in an RNA-dependent manner. Our data suggest a riboregulation of mitotic protein-protein interactions during spindle assembly, offering new perspectives on the control of cell division processes by RNA-protein complexes.
Collapse
Affiliation(s)
- Varshni Rajagopal
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jeanette Seiler
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Isha Nasa
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Simona Cantarella
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jana Theiss
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Franziska Herget
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bianca Kaifer
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Schneider
- Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominic Helm
- Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julian König
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Kathi Zarnack
- Buchmann Institute for Molecular Life Sciences, Frankfurt, Germany
- Department of Bioinformatics, University of Würzburg, Würzburg, Germany
| | - Sven Diederichs
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between DKFZ and University Medical Center Freiburg, Freiburg, Germany
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Maïwen Caudron-Herger
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
2
|
Dawber RS, Gimenez D, Batchelor M, Miles JA, Wright MH, Bayliss R, Wilson AJ. Inhibition of Aurora-A/N-Myc Protein-Protein Interaction Using Peptidomimetics: Understanding the Role of Peptide Cyclization. Chembiochem 2024; 25:e202300649. [PMID: 37907395 PMCID: PMC10962542 DOI: 10.1002/cbic.202300649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/02/2023]
Abstract
Using N-Myc61-89 as a starting template we showcase the systematic use of truncation and maleimide constraining to develop peptidomimetic inhibitors of the N-Myc/Aurora-A protein-protein interaction (PPI); a potential anticancer drug discovery target. The most promising of these - N-Myc73-94-N85C/G89C-mal - is shown to favour a more Aurora-A compliant binding ensemble in comparison to the linear wild-type sequence as observed through fluorescence anisotropy competition assays, circular dichroism (CD) and nuclear magnetic resonance (NMR) experiments. Further in silico investigation of this peptide in its Aurora-A bound state, by molecular dynamics (MD) simulations, imply (i) the bound conformation is more stable as a consequence of the constraint, which likely suppresses dissociation and (ii) the constraint may make further stabilizing interactions with the Aurora-A surface. Taken together this work unveils the first orthosteric N-Myc/Aurora-A inhibitor and provides useful insights on the biophysical properties and thus design of constrained peptides, an attractive therapeutic modality.
Collapse
Affiliation(s)
- Robert S. Dawber
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- School of ChemistryUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- School of Molecular and Cellular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
| | - Diana Gimenez
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- School of ChemistryUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
| | - Matthew Batchelor
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- School of Molecular and Cellular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
| | - Jennifer A. Miles
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- School of Molecular and Cellular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
| | - Megan H. Wright
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- School of ChemistryUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
| | - Richard Bayliss
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- School of Molecular and Cellular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
| | - Andrew J. Wilson
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- School of ChemistryUniversity of LeedsWoodhouse LaneLeedsLS2 9JTUK
- School of ChemistryUniversity of BirminghamEdgbaston, BirminghamB15 2TTUK
| |
Collapse
|
3
|
Bathula S, Sankaranarayanan M, Malgija B, Kaliappan I, Bhandare RR, Shaik AB. 2-Amino Thiazole Derivatives as Prospective Aurora Kinase Inhibitors against Breast Cancer: QSAR, ADMET Prediction, Molecular Docking, and Molecular Dynamic Simulation Studies. ACS OMEGA 2023; 8:44287-44311. [PMID: 38027360 PMCID: PMC10666282 DOI: 10.1021/acsomega.3c07003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/05/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023]
Abstract
The aurora kinase is a key enzyme that is implicated in tumor growth. Research revealed that small molecules that target aurora kinase have beneficial effects as anticancer agents. In the present study, in order to identify potential antibreast cancer agents with aurora kinase inhibitory activity, we employed QSARINS software to perform the quantitative structure-activity relationship (QSAR). The statistical values resulted from the study include R2 = 0.8902, CCCtr = 0.7580, Q2 LOO = 0.7875, Q2LMO = 0.7624, CCCcv = 0.7535, R2ext = 0.8735, and CCCext = 0.8783. Among the four generated models, the two best models encompass five important variables, including PSA, EstateVSA5, MoRSEP3, MATSp5, and RDFC24. The parameters including the atomic volume, atomic charges, and Sanderson's electronegativity played an important role in designing newer lead compounds. Based on the above data, we have designed six series of compounds including 1a-e, 2a-e, 3a-e, 4a-e, 5a-e, and 6a-e. All these compounds were subjected to molecular docking studies by using AutoDock v4.2.6 against the aurora kinase protein (1MQ4). Among the above 30 compounds, the 2-amino thiazole derivatives 1a, 2a, 3e, 4d, 5d, and 6d have excellent binding interactions with the active site of 1MQ4. Compound 1a had the highest docking score (-9.67) and hence was additionally subjected to molecular dynamic simulation investigations for 100 ns. The stable binding of compound 1a with 1MQ4 was verified by RMSD, RMSF, RoG, H-bond, molecular mechanics-generalized Born surface area (MM-GBSA), free binding energy calculations, and solvent-accessible surface area (SASA) analyses. Furthermore, newly designed compound 1a exhibited excellent ADMET properties. Based on the above findings, we propose that the designed compound 1a may be utilized as the best theoretical lead for future experimental research of selective inhibition of aurora kinase, therefore assisting in the creation of new antibreast cancer drugs.
Collapse
Affiliation(s)
- Sivakumar Bathula
- Department
of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM
Institute of Science and Technology, Kattankulathur 603203, Chengalpattu
District, Tamil Nadu, India
| | - Murugesan Sankaranarayanan
- Medicinal
Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science (BITS)
Pilani, Pilani Campus, Pilani 333031, Rajasthan, India
| | - Beutline Malgija
- MCC-MRF
Innovation Park, Madras Christian College, Chennai 600059, Tamil Nadu, India
| | - Ilango Kaliappan
- Department
of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM
Institute of Science and Technology, Kattankulathur 603203, Chengalpattu
District, Tamil Nadu, India
| | - Richie R. Bhandare
- Department
of Pharmaceutical Sciences, College
of Pharmacy and Health Sciences, Ajman University, P.O. Box 346, Ajman 61001, United Arab Emirates
- Centre of
Medical and Bio-allied Health Sciences Research, Ajman University, P.O. Box 346, Ajman 61001, United Arab Emirates
| | - Afzal B. Shaik
- St.
Mary’s College of Pharmacy, St. Mary’s
Group of Institutions Guntur, Affiliated to Jawaharlal Nehru Technological
University Kakinada, Chebrolu, Guntur 522212, Andhra
Pradesh, India
- Center
for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| |
Collapse
|
4
|
D Shankara S, Isloor AM, Jayaswamy PK, Shetty P, Chakraborty D, Venugopal PP. Vetting of New 2,5-Bis (2,2,2-trifluoroethoxy) Phenyl-Linked 1,3-Thiazolidine-4-one Derivatives as AURKA and VEGFR-2 Inhibitor Antiglioma Agents Assisted with In Vitro and In Silico Studies. ACS OMEGA 2023; 8:43596-43609. [PMID: 38027362 PMCID: PMC10666141 DOI: 10.1021/acsomega.3c04662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/29/2023] [Indexed: 12/01/2023]
Abstract
The bioactivity of 1,3-thiazolidin-4-one derivatives with a 2,5-bis (2,2,2-trifluoroethoxy) phenyl moiety was computationally developed and evaluated. All of the synthesized thiazolidin-4-one derivatives have their chemical structures characterized using a variety of methods, including nuclear magnetic resonance (NMR) (1H and 13C), high-resolution mass spectrometry (HRMS), and Fourier transform infrared (FTIR) radiation. A human glioblastoma cancer cell line (LN229) was used to investigate the purified derivatives' antiglioma cancer efficacy. By using the MTT, colony formation, and tunnel tests, respectively, the in vitro cytotoxic and apoptotic effects of these compounds were assessed. Thiazolidin-4-one derivatives 5b, 5c, and 5e were discovered to have the best efficacy against glioblastoma cells out of all of these compounds. The derivatives 5b, 5c, and 5e were determined to have respective IC50 values of 9.48, 12.16, and 6.43 g/mL. Computation results showed that the bioactivity evaluations of the compounds were quite significant. The bridging -NH group forms a hydrogen bond with Glu 260 of synthesized derivatives 5b, 5c, 5d, 5e, and 5h. The vast majority of freshly developed compounds obeyed Lipinski's rule of five, which is in line with the results that the ADMET model predicted. Additionally, molecular docking evaluation and molecular dynamics simulation investigations against the proteins AURKA and VEGFR-2 were conducted for the synthesized compounds to incorporate both in silico and in vitro data. The findings revealed that almost all of the compounds had considerable binding to AURKA and VEGFR-2 residues, with binding affinities ranging from -9.8 to -7.9 kcal/mol. Consequently, the results of the biological investigations and the docking scores demonstrated that thiazolidinone molecule 5e containing 4-chlorophenyl substituent may be considered as a potential moiety for glioblastoma cancer treatments.
Collapse
Affiliation(s)
- Sathyanarayana D Shankara
- Medicinal Chemistry
Laboratory, Department of Chemistry, National
Institute of Technology Karnataka, Surathkal, Mangalore 575 025, India
| | - Arun M. Isloor
- Medicinal Chemistry
Laboratory, Department of Chemistry, National
Institute of Technology Karnataka, Surathkal, Mangalore 575 025, India
| | - Pavan K. Jayaswamy
- Central Research Laboratory, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore 575018, Karnataka, India
| | - Praveenkumar Shetty
- Central Research Laboratory, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore 575018, Karnataka, India
- Department of Biochemistry, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore575018, Karnataka, India
| | - Debashree Chakraborty
- Biophysical and Computational Chemistry
Laboratory, Department of Chemistry, National
Institute of Technology Karnataka, Surathkal, Mangalore 575025, India
| | - Pushyaraga P. Venugopal
- Biophysical and Computational Chemistry
Laboratory, Department of Chemistry, National
Institute of Technology Karnataka, Surathkal, Mangalore 575025, India
| |
Collapse
|
5
|
Mao X, Cai Y, Long S, Perez-Losada J, Mao JH, Chang H. Pan-cancer evaluation of clinical value of mitotic network activity index (MNAI) and its predictive value for immunotherapy. Front Oncol 2023; 13:1178568. [PMID: 37456231 PMCID: PMC10349373 DOI: 10.3389/fonc.2023.1178568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Increased mitotic activity is associated with the genesis and aggressiveness of many cancers. To assess the clinical value of mitotic activity as prognostic biomarker, we performed a pan-cancer study on the mitotic network activity index (MNAI) constructed based on 54-gene mitotic apparatus network. Our pan-cancer assessment on TCGA (33 tumor types, 10,061 patients) and validation on other publicly available cohorts (23 tumor types, 9,209 patients) confirmed the significant association of MNAI with overall survival, progression-free survival, and other prognostic endpoints in multiple cancer types, including lower-grade gliomas (LGG), breast invasive carcinoma (BRCA), as well as many others. We also showed significant association between MNAI and genetic instability, which provides a biological explanation of its prognostic impact at pan-cancer landscape. Our association analysis revealed that patients with high MNAI benefitted more from anti-PD-1 and Anti-CTLA-4 treatment. In addition, we demonstrated that multimodal integration of MNAI and the AI-empowered Cellular Morphometric Subtypes (CMS) significantly improved the predictive power of prognosis compared to using MNAI and CMS alone. Our results suggest that MNAI can be used as a potential prognostic biomarker for different tumor types toward different clinical endpoints, and multimodal integration of MNAI and CMS exceeds individual biomarker for precision prognosis.
Collapse
Affiliation(s)
- Xuanyu Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Yimeng Cai
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, United States
| | - Sarah Long
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Jesus Perez-Losada
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Hang Chang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| |
Collapse
|
6
|
Shi H, Guo N, Zhao Z, Liu L, Ni T, Zhang J, Lu Y. Comparison of the second-line treatments for patients with small cell lung cancer sensitive to previous platinum-based chemotherapy: A systematic review and Bayesian network analysis. Front Oncol 2023; 13:1154685. [PMID: 37007093 PMCID: PMC10061131 DOI: 10.3389/fonc.2023.1154685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
ObjectiveIt remains unclear what the best second-line treatment is for patients with small-cell lung cancer sensitive to previous platinum-based chemotherapy.MethodsWe systematically screened randomized controlled trials from several online databases. The primary outcome was objective response rate (ORR), and the secondary outcomes were disease control rate (DCR), overall survival (OS), progression-free survival (PFS), and hematological complications graded 3 to 5. The efficacy of included treatments was ranked by surface under the cumulative ranking curve (SUCRA) value.ResultsWe included eleven trials involving 1560 patients in quantitative analysis. Triple chemotherapy containing platinum (TP, combination of cisplatin, etoposide, and irinotecan) was associated with favorable ORR (intravenous topotecan vs TP; odds ratio: 0.13, 95% CI:0.03-0.63; SUCRA, 0.94) and PFS (vs intravenous topotecan; hazard ratio, 0.5; 95% CI: 0.25-0.99; SUCRA, 0.90). Belotecan ranked highest for OS (SUCRA, 0.90), while intravenous topotecan plus Ziv-aflibercept ranked highest for DCR (SUCRA, 0.75). TP was more likely to cause anemia and thrombocytopenia while intravenous topotecan plus Ziv-aflibercept resulted in most neutrocytopenia.ConclusionTP is the first recommendation for the second-line treatment of sensitive relapsed SCLC. TP achieved priority in ORR and PFS with the most frequent adverse effects in anemia and thrombocytopenia. For patients who cannot tolerate the hematological adverse effects of triple chemotherapy, amrubicin is an optional option. Amrubicin had relatively good ORR and PFS, accompanied by fewer hematological complications. The rechallenge of the platinum doublet is inferior to amrubicin in ORR, DCR, and PFS. Oral topotecan has a similar effect compared with IV topotecan, but oral topotecan was associated with slightly higher safety and less stress in nursing. Belotecan contributed to the best PFS with slightly better safety but was not ideal in other outcomes.Systematic review registrationhttps://www.crd.york.ac.uk/PROSPERO/, identifier CRD42022358256.
Collapse
Affiliation(s)
- Hekai Shi
- Department of Thoracic Surgery, Fudan University Affiliated Huadong Hospital, Shanghai, China
| | - Nuojin Guo
- Shanghai East Hospital, Tongji University, Shanghai, China
| | - Zeming Zhao
- Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ligang Liu
- Institute of Therapeutic Innovations and Outcomes, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Tianyi Ni
- Department of Thoracic Surgery, Fudan University Affiliated Huadong Hospital, Shanghai, China
| | - Jinye Zhang
- First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yingjie Lu
- Department of Thoracic Surgery, Fudan University Affiliated Huadong Hospital, Shanghai, China
- *Correspondence: Yingjie Lu,
| |
Collapse
|
7
|
Boi D, Rubini E, Breccia S, Guarguaglini G, Paiardini A. When Just One Phosphate Is One Too Many: The Multifaceted Interplay between Myc and Kinases. Int J Mol Sci 2023; 24:4746. [PMID: 36902175 PMCID: PMC10003727 DOI: 10.3390/ijms24054746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
Myc transcription factors are key regulators of many cellular processes, with Myc target genes crucially implicated in the management of cell proliferation and stem pluripotency, energy metabolism, protein synthesis, angiogenesis, DNA damage response, and apoptosis. Given the wide involvement of Myc in cellular dynamics, it is not surprising that its overexpression is frequently associated with cancer. Noteworthy, in cancer cells where high Myc levels are maintained, the overexpression of Myc-associated kinases is often observed and required to foster tumour cells' proliferation. A mutual interplay exists between Myc and kinases: the latter, which are Myc transcriptional targets, phosphorylate Myc, allowing its transcriptional activity, highlighting a clear regulatory loop. At the protein level, Myc activity and turnover is also tightly regulated by kinases, with a finely tuned balance between translation and rapid protein degradation. In this perspective, we focus on the cross-regulation of Myc and its associated protein kinases underlying similar and redundant mechanisms of regulation at different levels, from transcriptional to post-translational events. Furthermore, a review of the indirect effects of known kinase inhibitors on Myc provides an opportunity to identify alternative and combined therapeutic approaches for cancer treatment.
Collapse
Affiliation(s)
- Dalila Boi
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Elisabetta Rubini
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Sara Breccia
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Giulia Guarguaglini
- Institute of Molecular Biology and Pathology, National Research Council of Italy, Sapienza University of Rome, 00185 Rome, Italy
| | - Alessandro Paiardini
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
8
|
Lee IG, Lee BJ. Aurora Kinase A Regulation by Cysteine Oxidative Modification. Antioxidants (Basel) 2023; 12:antiox12020531. [PMID: 36830089 PMCID: PMC9952272 DOI: 10.3390/antiox12020531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Aurora kinase A (AURKA), which is a member of serine/threonine kinase family, plays a critical role in regulating mitosis. AURKA has drawn much attention as its dysregulation is critically associated with various cancers, leading to the development of AURKA inhibitors, a new class of anticancer drugs. As the spatiotemporal activity of AURKA critically depends on diverse intra- and inter-molecular factors, including its interaction with various protein cofactors and post-translational modifications, each of these pathways should be exploited for the development of a novel class of AURKA inhibitors other than ATP-competitive inhibitors. Several lines of evidence have recently shown that redox-active molecules can modify the cysteine residues located on the kinase domain of AURKA, thereby regulating its activity. In this review, we present the current understanding of how oxidative modifications of cysteine residues of AURKA, induced by redox-active molecules, structurally and functionally regulate AURKA and discuss their implications in the discovery of novel AURKA inhibitors.
Collapse
Affiliation(s)
- In-Gyun Lee
- Biomedical Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Bong-Jin Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- Correspondence:
| |
Collapse
|
9
|
Lv G, Shi Q, Zhang T, Li J, Long Y, Zhang W, Choudhry N, Yang K, Li H, Kalashova J, Yang C, Zhou X, Reddy MC, Anantoju KK, Zhang S, Zhang J, Allen TD, Liu H, Nimishetti N, Yang D. Integrating a phenotypic screening with a structural simplification strategy to identify 4-phenoxy-quinoline derivatives to potently disrupt the mitotic localization of Aurora kinase B. Bioorg Med Chem 2023; 80:117173. [PMID: 36696874 DOI: 10.1016/j.bmc.2023.117173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/31/2022] [Accepted: 01/09/2023] [Indexed: 01/18/2023]
Abstract
We combined a mechanism-informed phenotypic screening (MIPS) assay with a structural simplification strategy to guide the discovery of compounds that disrupt the localization of the mitotic regulator, Aurora kinase B (AURKB), rather than inhibiting its catalytic activity. An initial hit 4-(4-methylthiophen-2-yl)-N-(4-(quinolin-4-yloxy)phenyl)phthalazin-1-amine was identified after screening an in-house library of small molecules and phenocopied the loss of function mutations in AURKB without inhibiting its catalytic activity. We isolated this hit compound activity to its 4-phenoxy-quinoline moiety. The fragment was further optimized into a class of new chemical entities that potently disrupt the mitotic localization of AURKB at low nanomolar concentrations and consequently elicit severe growth inhibition in diverse human cancer cell lines. A lead compound, N-(3-methoxy-5-(6-methoxyquinolin-4-yl)oxy)phenyl)acetamide possessed desirable pharmacokinetic properties such as AUC0-∞: 227.15 [ng∙h/mL/(mg/kg)]; Cmax: 3378.52 ng/mL T1/2: 3.52 h; and F%: 42 % and produced the AURKB-inhibitory phenotypes in a mouse xenograft model. A lead compound is a powerful tool for interrogating the regulation of AURKB and has the potential to be further developed as a first-in-class oncology therapeutic.
Collapse
Affiliation(s)
- Gang Lv
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China.
| | - Qiong Shi
- J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China
| | - Ting Zhang
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China
| | - Jinhua Li
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China
| | - Yan Long
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China
| | - Wenhui Zhang
- Chengdu Anticancer Bioscience, Chengdu 610000, China
| | - Namrta Choudhry
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China
| | - Kevin Yang
- Chengdu Anticancer Bioscience, Chengdu 610000, China; Trinity College of Arts and Sciences, Duke University, Durham, NC 27708, USA
| | - Hongmei Li
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China
| | - Julia Kalashova
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China
| | - Chenglu Yang
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China
| | - Xiaohu Zhou
- Chengdu Anticancer Bioscience, Chengdu 610000, China
| | | | | | - Shenqiu Zhang
- J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China
| | - Jing Zhang
- J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China
| | | | - Hong Liu
- Anticancer Bioscience (US), South San Francisco, CA 94080, USA
| | - Naganna Nimishetti
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China.
| | - Dun Yang
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China.
| |
Collapse
|
10
|
STYK1/NOK affects cell cycle late mitosis and directly interacts with anaphase-promoting complex activator CDH1. Heliyon 2022; 8:e12058. [PMID: 36506394 PMCID: PMC9732331 DOI: 10.1016/j.heliyon.2022.e12058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/25/2022] [Accepted: 11/25/2022] [Indexed: 12/07/2022] Open
Abstract
The novel oncogene STYK1/NOK plays critical roles in cancer development. However, its regulation during cell division is less defined. In this paper, we show that over-expression of STYK1/NOK caused mitotic arrest and cytokinesis defects. The protein level of STYK/NOK fluctuated during the cell cycle, with a peak at mitosis and a quick reduction upon mitotic exit. The cell cycle-related expression pattern of STYK1/NOK resembled the one of aurora kinases and polo-like kinase 1. Depletion of APC3 led to accumulation of STYK1/NOK and to the G2/M arrest. Co-immunoprecipitation experiment demonstrated the direct interaction of STYK1/NOK with CDH1. Overexpression of CDH1 shortened the half-life of STYK1/NOK. The kinase domain, but not the five D boxes, of STYK1/NOK was responsible for the interaction with CDH1. Altogether, our data demonstrated for the first time that STYK1/NOK could affect cell division, probably by directly targeting key components of APC/C such as CDH1 at late mitosis. Current study may provide a vital mechanistic clue for understanding the roles of STYK1/NOK in mitosis and cytokinesis during STYK1NOK mediated genomic instability and oncogenesis.
Collapse
|
11
|
Bhosle MR, Palke A, Bondle GM, Sarkate AP, Azad R, Burra PVLS. Efficient Synthesis of Densely Functionalized Pyrido[2,3-d]Pyrimidines via Three-component One-pot Domino Knoevenagel aza-Diels Alder Reaction and Induces Apoptosis in Human Cancer Cell Lines via Inhibiting Aurora A and B Kinases. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2022.2143538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Manisha R. Bhosle
- Department of Chemistry, Dr. Babasaheb Ambedkar, Marathwada University, Aurangabad, India
| | - Amruta Palke
- Department of Chemistry, Dr. Babasaheb Ambedkar, Marathwada University, Aurangabad, India
| | - Giribala M. Bondle
- Department of Chemistry, Dr. Babasaheb Ambedkar, Marathwada University, Aurangabad, India
| | - Aniket P. Sarkate
- Department of Chemical Technology, Dr. Babasaheb Ambedkar, Marathwada University, Aurangabad, India
| | - Rajaram Azad
- Department of Animal Biology, University of Hyderabad, Hyderabad, India
| | | |
Collapse
|
12
|
de Oliveira FM, Jamur VR, Merfort LW, Pozzo AR, Mai S. Three-dimensional nuclear telomere architecture and differential expression of aurora kinase genes in chronic myeloid leukemia to measure cell transformation. BMC Cancer 2022; 22:1024. [PMID: 36175852 PMCID: PMC9520804 DOI: 10.1186/s12885-022-10094-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 09/14/2022] [Indexed: 11/10/2022] Open
Abstract
Background Telomere dysfunction results in aneuploidy, and ongoing chromosomal abnormalities. The three-dimensional (3D) nuclear organization of telomeres allows for a distinction between normal and tumor cells. On the other hand, aurora kinase genes (AURKA and AURKB) play an important role regulating the cell cycle. A correlation between overexpression of aurora kinase genes and clinical aggressiveness has been demonstrated in different types of neoplasias. To better understand cellular and molecular mechanisms of CML evolution, it was examined telomere dysfunction (alterations in the 3D nuclear telomere architecture), and the expression levels of AURKA and AURKB genes in two clinical distinct subgroups of CML samples, from the same patient. Methods Eighteen CML patients, in total, 36 bone marrow samples (18 patients, chronic vs. accelerated/blast phase) were eligible for 3D telomeric investigations. Quantitative 3D imaging, cytologic diagnosis and cytogenetic determination of additional chromosomal abnormalities were assessed according to standard protocols. Results Using TeloView software, two CML subgroups were defined based on their 3D telomeric profiles, reflecting the different stages of the disease (chronic vs. accelerated/blast phase). Statistical analyses showed significant differences between the CML subgroups (p < 0.001). We also found that AURKA and AURKB mRNA were expressed at significantly higher levels in both CML subgroups, when compared with healthy donors. Our findings suggest that the evolution of CML progresses from a low to a high level of telomere dysfunction, that is, from an early stage to a more aggressive stage, followed by disease transformation, as demonstrated by telomere, additional chromosomal abnormalities, and gene expression profile dynamics. Conclusions Thus, we demonstrated that 3D telomere organization, in accordance with the genomic instability observed in CML samples were able to distinguish subgroup CML patients. Classifying CML patients based on these characteristics might represent an important strategy to define better therapeutic strategies.
Collapse
Affiliation(s)
- Fábio Morato de Oliveira
- Laboratory of Medical Genetics, Câmpus Jatobá - Cidade Universitária, Federal University of Jataí, BR 364, km 195, n° 3800, Jataí, CEP 75801-615, Brazil.
| | - Valderez Ravaglio Jamur
- Complexo Hospital das Clínicas, Universidade Federal do Paraná, Curitiba, Paraná, 80060-240, Brazil
| | - Lismeri Wuicik Merfort
- Complexo Hospital das Clínicas, Universidade Federal do Paraná, Curitiba, Paraná, 80060-240, Brazil
| | - Aline Rangel Pozzo
- Research Institute in Oncology and Hematology CancerCare Manitoba, The Genomic Centre for Cancer Research and Diagnosis, The University of Manitoba, Winnipeg, MB, R3E 0V9, Canada
| | - Sabine Mai
- Research Institute in Oncology and Hematology CancerCare Manitoba, The Genomic Centre for Cancer Research and Diagnosis, The University of Manitoba, Winnipeg, MB, R3E 0V9, Canada.
| |
Collapse
|
13
|
Exploring Potential Biomarkers, Ferroptosis Mechanisms, and Therapeutic Targets Associated with Cutaneous Squamous Cell Carcinoma via Integrated Transcriptomic Analysis. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:3524022. [PMID: 36247089 PMCID: PMC9553755 DOI: 10.1155/2022/3524022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/17/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022]
Abstract
Background Cutaneous squamous cell carcinoma (cSCC) is the leading cause of death in patients with nonmelanoma skin cancers (NMSC). However, the unclear pathogenesis of cSCC limits the application of molecular targeted therapy. Methods Three microarray datasets (GSE2503, GSE45164, and GSE66359) were downloaded from the Gene Expression Omnibus (GEO). After identifying the differentially expressed genes (DEGs) in tumor and nontumor tissues, five kinds of analyses, namely, functional annotation, protein-protein interaction (PPI) network, hub gene selection, TF-miRNA-mRNA regulatory network analysis, and ferroptosis mechanism, were performed. Results A total of 146 DEGs were identified with significant differences, including 113 upregulated genes and 33 downregulated genes. The enriched functions and pathways of the DEGs included microtubule-based movement, ATP binding, cell cycle, P53 signaling pathway, oocyte meiosis, and PLK1 signaling events. Nine hub genes were identified (CDK1, AURKA, RRM2, CENPE, CCNB1, KIAA0101, ZWINT, TOP2A, and ASPM). Finally, RRM2, AURKA, and SAT1 were identified as significant ferroptosis-related genes in cSCC. The differential expression of these genes has been verified in two other independent datasets. Conclusions By integrated bioinformatic analysis, the hub genes identified in this study elucidated the molecular mechanism of the pathogenesis and progression of cSCC and are expected to become future biomarkers or therapeutic targets.
Collapse
|
14
|
Balahmar RM, Ranganathan B, Ebegboni V, Alamir J, Rajakumar A, Deepak V, Sivasubramaniam S. Analyses of selected tumour-associated factors expression in normotensive and preeclamptic placenta. Pregnancy Hypertens 2022; 29:36-45. [PMID: 35717832 DOI: 10.1016/j.preghy.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Human placenta is often considered a controlled-tumour because of shared properties such as invasion and angiogenesis. We assessed the status of a few selected tumour-associated factors (TAFs) in late onset pre-eclamptic (PE) and normotensive (NT) placentae, to understand their involvement in trophoblast invasion. These molecules include aldehyde dehydrogenase (ALDH3A1), aurora kinases (AURK-A/C), platelet derived growth factor receptor-α (PDGFRα), jagged-1 (JAG1) and twist related protein-1 (TWIST1). METHODS The expression of TAF was compared in 13 NT and 11 PE (late onset) placentae using immunoblotting/immunohistochemistry. We then used a novel spheroidal cell model developed from transformed human first trimester trophoblast cell lines HTR8/SVneo and TEV-1 to determine the expression and localization of these six factors during invasion. We also compared the expression of these TAFs during migration and invasion. RESULTS Our results suggest that expressions of ALDH3A1, AURK-A, PDGFRα, and TWIST1 are significantly upregulated in PE placentae (p < 0.05) when compared to NT placentae, whereas AURK-C and JAG1 are down-regulated (p < 0.05). The protein expression pattern of all the six factors were found to be similar in spheroids in comparison to their parental counterparts. The invasive potential of the spheroids was also enhanced when compared with the parental cells. DISCUSSION Collectively, data from our present study suggests that these TAFs are involved in placental invasion and their altered expressions may be regarded as a compensatory mechanism against reduced invasion.
Collapse
Affiliation(s)
- Reham M Balahmar
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - Bhuvaneshwari Ranganathan
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - Vernon Ebegboni
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - Jumanah Alamir
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - Augustine Rajakumar
- Department of Gynecology & Obstetrics(3), Emory University School of Medicine, Atlanta, GA 30033, USA
| | - Venkataraman Deepak
- School of Human Sciences, College of Life and Natural Sciences, University of Derby, Kedleston Road, Derby DE22 1GB, United Kingdom.
| | - Shiva Sivasubramaniam
- School of Human Sciences, College of Life and Natural Sciences, University of Derby, Kedleston Road, Derby DE22 1GB, United Kingdom.
| |
Collapse
|
15
|
Lee JY, Yang H, Kim D, Kyaw KZ, Hu R, Fan Y, Lee SK. Antiproliferative Activity of a New Quinazolin-4(3H)-One Derivative via Targeting Aurora Kinase A in Non-Small Cell Lung Cancer. Pharmaceuticals (Basel) 2022; 15:ph15060698. [PMID: 35745617 PMCID: PMC9228987 DOI: 10.3390/ph15060698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/24/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common lung cancer subtype. Although chemotherapy and targeted therapy are used for the treatment of patients with NSCLC, the survival rate remains very low. Recent findings suggested that aurora kinase A (AKA), a cell cycle regulator, is a potential target for NSCLC therapy. Previously, we reported that a chemical entity of quinazolin-4(3H)-one represents a new template for AKA inhibitors, with antiproliferative activity against cancer cells. A quinazolin-4(3H)-one derivative was further designed and synthesized in order to improve the pharmacokinetic properties and antiproliferation activity against NSCLC cell lines. The derivative, BIQO-19 (Ethyl 6-(4-oxo-3-(pyrimidin-2-ylmethyl)-3,4-dihydroquinazolin-6-yl)imidazo [1,2-a]pyridine-2-carboxylate), exhibited improved solubility and antiproliferative activity in NSCLC cells, including epidermal growth factor receptor–tyrosine kinase inhibitor (EGFR-TKI)-resistant NSCLC cells. BIQO-19 effectively inhibited the growth of the EGFR-TKI-resistant H1975 NSCLC cells, with the suppression of activated AKA (p-AKA) expression in these cells. The inhibition of AKA by BIQO-19 significantly induced G2/M phase arrest and subsequently evoked apoptosis in H1975 cells. In addition, the combination of gefitinib and BIQO-19 exhibited synergistic antiproliferative activity in NSCLC cells. These findings suggest the potential of BIQO-19 as a novel therapeutic agent for restoring the sensitivity of gefitinib in EGFR-TKI-resistant NSCLC cells.
Collapse
Affiliation(s)
- Ji Yun Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (J.Y.L.); (D.K.); (K.Z.K.); (R.H.)
| | - Huarong Yang
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China;
| | - Donghwa Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (J.Y.L.); (D.K.); (K.Z.K.); (R.H.)
| | - Kay Zin Kyaw
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (J.Y.L.); (D.K.); (K.Z.K.); (R.H.)
| | - Ruoci Hu
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (J.Y.L.); (D.K.); (K.Z.K.); (R.H.)
| | - Yanhua Fan
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province, Chinese Academy of Sciences, Guiyang 550014, China;
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Correspondence: (Y.F.); (S.K.L.); Tel.: +82-2-880-2475 (S.K.L.)
| | - Sang Kook Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (J.Y.L.); (D.K.); (K.Z.K.); (R.H.)
- Correspondence: (Y.F.); (S.K.L.); Tel.: +82-2-880-2475 (S.K.L.)
| |
Collapse
|
16
|
Wharton’s Jelly-Derived Mesenchymal Stem Cells with High Aurora Kinase A Expression Show Improved Proliferation, Migration, and Therapeutic Potential. Stem Cells Int 2022; 2022:4711499. [PMID: 35450345 PMCID: PMC9017458 DOI: 10.1155/2022/4711499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/11/2022] [Indexed: 11/25/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are effective therapeutic agents that contribute to tissue repair and regeneration by secreting various factors. However, donor-dependent variations in MSC proliferation and therapeutic potentials result in variable production yields and clinical outcomes, thereby impeding MSC-based therapies. Hence, selection of MSCs with high proliferation and therapeutic potentials would be important for effective clinical application of MSCs. This study is aimed at identifying the upregulated genes in human Wharton's jelly-derived MSCs (WJ-MSCs) with high proliferation potential using mRNA sequencing. Aurora kinase A (AURKA) and dedicator of cytokinesis 2 (DOCK2) were selected as the upregulated genes, and their effects on proliferation, migration, and colony formation of the WJ-MSCs were verified using small interfering RNA (siRNA) techniques. mRNA expression levels of both the genes were positively correlated with the proliferation capacity of WJ-MSCs. Moreover, AURKA from human WJ-MSCs regulated the antiapoptotic effect of skeletal muscle cells by upregulating the chemokine (C motif) ligand (XCL1); this was further confirmed in the mdx mouse model. Taken together, the results indicated that AURKA and DOCK2 can be used as potential biomarkers for proliferation and migration of human WJ-MSCs. In particular, human WJ-MSCs with high expression of AURKA might have therapeutic efficacy against muscle diseases, such as Duchenne muscular dystrophy (DMD).
Collapse
|
17
|
Guo J, Li W, Cheng L, Gao X. Identification and Validation of Hub Genes with Poor Prognosis in Hepatocellular Carcinoma by Integrated Bioinformatical Analysis. Int J Gen Med 2022; 15:3933-3941. [PMID: 35431572 PMCID: PMC9012340 DOI: 10.2147/ijgm.s353708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/01/2022] [Indexed: 12/24/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the reason for the world’s second largest cancer-related death. It is clinically valuable to study the molecular mechanisms of HCC occurrence and development for formulating more effective diagnosis and treatment strategies. Methods The five microarray data sets GSE45267, GSE101685, GSE84402, GSE62232 and GSE45267 were downloaded from Gene Expression Omnibus (GEO) database, including 165 HCC tissues and 73 normal tissues. Differential expressed genes (DEGs) between HCC tissues and normal tissues were determined by GEO2R. Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and the protein–protein interaction network (PPI) network analysis were employed to identify DEGs and to evaluate the clinical significance in prognosis of HCC. Results A total of 152 genes differentially expressed in HCC tissues and normal tissues were identified. GO and KEGG functional enrichment analysis revealed that 39 up-regulated genes were mainly enriched in mitosis, cell cycle and oocyte meiosis, while those down-regulated genes (113) were concentrated in exogenous drug catabolism and the metabolism of cytochrome P450 on exogenous drugs. Totally, 19 hub genes were chosen by PPI network and module analysis and verified by The Cancer Genome Atlas (TCGA) database. Finally, 8 hub genes were selected, including CDK1, CYP2C8, CCNB1, AURKA, CYP2C9, BUB1B, MAD2L1 and TTK, which were associated with the overall survival rate of HCC patients. Conclusion This study presented eight target genes connected to the prognosis of HCC patients. Those mainly exists in cell cycle and drug catabolism, which may be latent targets for clinical treatment.
Collapse
Affiliation(s)
- Jiang Guo
- Department of Interventional Oncology, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
| | - Wei Li
- Center of Liver Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
| | - Long Cheng
- Department of Interventional Oncology, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
| | - Xuesong Gao
- Department of General Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China
- Correspondence: Xuesong Gao, Department of General Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China, Tel +86 13718689825, Fax +861084322146, Email
| |
Collapse
|
18
|
The Higher Expression of CDCA2 Associated with Poor Prognosis in Glioma. DISEASE MARKERS 2022; 2022:2184867. [PMID: 35386230 PMCID: PMC8977301 DOI: 10.1155/2022/2184867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 11/17/2022]
Abstract
Glioma is the most common primary intracranial tumor and is related to poor clinical outcomes. The developments of sensitive markers can be applied to reveal the mechanisms involved in the progression of glioma. This study examined CDCA2 expression in glioma samples and its significance in predicting glioma patient outcome. GEPIA and GEO datasets were used to explore the expression of CDCA2 in glioma. Kaplan-Meier and multivariate assays were applied to delve into the prognostic values of CDCA2 expression in glioma patients using CGGA datasets. Our group also determined the associations between CDCA2 and clinical characteristics. Coexpression analysis was performed. In this research, we observed that CDCA2 expression was distinctly upregulated in glioma specimens compared with nontumor specimens. The prognosis of glioma with high CDCA2 expression was distinctly worse compared with that of glioma with low CDCA2 expression. Additionally, multivariate Cox regression analysis revealed that high CDCA2 expression was an independent poor prognostic indicator for glioma patients. High expression of CDCA2 was positively associated with advanced clinical progression. Coexpression analysis revealed that CDCA2 could be positively related to ASPM, SKA1, DLGAP5, NCAPG, and CDCA8 and was negatively associated with ETNPPL, LDHD, MRVI1, CBX7, and CENPJ. Overall, our findings revealed that CDCA2 might serve as an independent prognosis indicator for glioma.
Collapse
|
19
|
Targetable Pathways in the Treatment of Retroperitoneal Liposarcoma. Cancers (Basel) 2022; 14:cancers14061362. [PMID: 35326514 PMCID: PMC8946646 DOI: 10.3390/cancers14061362] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 02/04/2023] Open
Abstract
Liposarcoma (LPS) is the most prevalent soft tissue sarcoma histological subtype. When it occurs in the abdomen the overall survival rate is as low as 10% at 10 years and is fraught with high rates of recurrence, particularly for the more aggressive dedifferentiated subtype. Surgery remains the mainstay of treatment. Systemic therapies for the treatment of metastatic or unresectable disease have low response rates. Deep understanding of well-differentiated and de-differentiated LPS (WDLPS and DDLPS, respectively) oncologic drivers is necessary for the development of new efficacious targeted therapies for the management of this disease. This review discusses the current treatments under evaluation for retroperitoneal DDLPS and the potential targetable pathways in DDLPS.
Collapse
|
20
|
Zhang Y, Wang Y, Xue J, Liang W, Zhang Z, Yang X, Qiao Z, Jiang Y, Wang J, Cao X, Chen P. Co-treatment with miR-21-5p inhibitor and Aurora kinase inhibitor reversine suppresses breast cancer progression by targeting sprouty RTK signaling antagonist 2. Bioengineered 2021; 13:455-468. [PMID: 34967265 PMCID: PMC8805969 DOI: 10.1080/21655979.2021.2009410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Numerous studies have reported the regulatory effects of miR-21-5p and reversine in human breast cancer (HBC). However, the mechanism of reversine and miR-21-5p has not been fully investigated in HBC. The aim of the current study was to assess the mechanism of action of reversine, with or without miR-21-5p, in HBC progression. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot results confirmed the upregulation of miR-21-5p and downregulation of sprouty RTK signaling antagonist 2 (SPRY2) in HBC. Bioinformatics analysis and luciferase assay identified the correlation between miR-21-5p and SPRY2. Cell function experiment results indicated a decrease in migration, proliferation, and invasion of HBC cells treated with miR-21-5p inhibitor and reversine; however, an increase in apoptosis was observed in these cells. Apoptotic ability was more enhanced and migration, proliferation, and invasion were more impaired in HBC cells treated with both miR-21-5p inhibitor and reversine than in those treated individually with either inhibitors. SPRY2, downstream of miR-21-5p, participated in HBC progression with reversine. Overall, our study proved that combining the miR-21-5p inhibitor with reversine produced a synergistic effect by regulating SPRY2, thereby limiting HBC progression. This knowledge might offer insights into the clinical therapy of HBC.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China.,Lung Cancer Diagnosis and Treatment Center, Tianjin, China.,Department of Mammography Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yaoyi Wang
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Key Laboratory of Functional Imaging, Tianjin, China
| | - Jun Xue
- Department of Mammography Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Wanping Liang
- Department of Mammography Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Zhisheng Zhang
- Department of Mammography Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Xiuming Yang
- Department of Mammography Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Zhifei Qiao
- Department of Mammography Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yang Jiang
- Department of Mammography Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Junping Wang
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuchen Cao
- National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China.,Lung Cancer Diagnosis and Treatment Center, Tianjin, China.,The First Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Peng Chen
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China.,Lung Cancer Diagnosis and Treatment Center, Tianjin, China
| |
Collapse
|
21
|
González-Borja I, Alors-Pérez E, Amat I, Alonso L, Viyuela-García C, Goñi S, Reyes JC, Ceballos-Chávez M, Hernández-García I, Sánchez-Frías ME, Santamaría E, Razquin S, Arjona-Sánchez Á, Arrazubi V, Pérez-Sanz J, Vera R, Fernández-Irigoyen J, Castaño JP, Viúdez A. Deciphering CHFR Role in Pancreatic Ductal Adenocarcinoma. Front Med (Lausanne) 2021; 8:720128. [PMID: 34869418 PMCID: PMC8639583 DOI: 10.3389/fmed.2021.720128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/04/2021] [Indexed: 12/09/2022] Open
Abstract
Checkpoint with forkhead-associated and ring finger domains (CHFR) has been proposed as a predictive and prognosis biomarker for different tumor types, but its role in pancreatic ductal adenocarcinoma (PDAC) remains unknown. The aim of this study was two-pronged: to review the role of CHFR in PDAC and evaluating CHFR as a potential predictive biomarker in this disease. For this purpose, we first explored the CHFR messenger (m)RNA expression and promoter methylation through the TCGA database. Secondly, the CHFR expression and promoter methylation were prospectively evaluated in a cohort of patients diagnosed with borderline (n = 19) or resectable (n = 16) PDAC by immunohistochemistry (IHC), methylation specific-PCR (MSP), and pyrosequencing. The results from the TCGA database showed significant differences in terms of progression-free survival (PFS) and overall survival (OS) based on the CHFR mRNA expression, which was likely independent from the promoter methylation. Importantly, our results showed that in primarily resected patients and also the entire cohort, a higher CHFR expression as indicated by the higher IHC staining intensity might identify patients with longer disease-free survival (DFS) and OS, respectively. Similarly, in the same cohorts, patients with lower methylation levels by pyrosequencing showed significantly longer OS than patients without this pattern. Both, the CHFR expression intensity and its promoter methylation were established as independent prognostic factors for PFS and OS in the entire cohort. In contrast, no significant differences were found between different methylation patterns for CHFR and the response to taxane-based neoadjuvant treatment. These results suggest the potential role of the higher expression of CHFR and the methylation pattern of its promoter as potential prognostic biomarkers in PDAC, thus warranting further comprehensive studies to extend and confirm our preliminary findings.
Collapse
Affiliation(s)
- Iranzu González-Borja
- OncobionaTras Lab, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Emilia Alors-Pérez
- Hormones and Cancer Group, Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Irene Amat
- Pathology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Laura Alonso
- Pathology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Cristina Viyuela-García
- Hormones and Cancer Group, Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,Surgery Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Saioa Goñi
- OncobionaTras Lab, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - José C Reyes
- Centro Andaluz de Biología Molecular y Medicina Regenerativa, Consejo Superior de Investigaciones Científicas-Universidad de Sevilla-Universidad Pablo de Olavide, Seville, Spain
| | - María Ceballos-Chávez
- Centro Andaluz de Biología Molecular y Medicina Regenerativa, Consejo Superior de Investigaciones Científicas-Universidad de Sevilla-Universidad Pablo de Olavide, Seville, Spain
| | | | - Marina E Sánchez-Frías
- Hormones and Cancer Group, Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,Pathology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Enrique Santamaría
- Proteomics Platform, Clinical Neuroproteomics Unit, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Socorro Razquin
- Pathology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Álvaro Arjona-Sánchez
- Hormones and Cancer Group, Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,Surgery Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Virginia Arrazubi
- Medical Oncology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Jairo Pérez-Sanz
- OncobionaTras Lab, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Ruth Vera
- Medical Oncology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Proteomics Platform, Clinical Neuroproteomics Unit, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Justo P Castaño
- Hormones and Cancer Group, Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain.,Centro de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | - Antonio Viúdez
- OncobionaTras Lab, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain.,Medical Oncology Department, Complejo Hospitalario de Navarra, Pamplona, Spain.,Medical Affairs Services, ICON plc, North Wales, PA, United States
| |
Collapse
|
22
|
Li S, Qin C, Chen Y, Wei D, Tan Z, Meng J. Implications of cell division cycle associated 4 on the Wilm's tumor cells viability via AKT/mTOR signaling pathway. Ren Fail 2021; 43:1470-1478. [PMID: 34723730 PMCID: PMC8567894 DOI: 10.1080/0886022x.2021.1994994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Objective The aim of present report was to elucidate the effect of cell division cycle associated 4 (CDCA4) on the proliferation and apoptosis of Wilm’s tumor cells, and to further evaluate its underlying mechanism. Methods The expression profiles of CDCA4 and clinical information of Wilm’s tumor patients were obtained from public Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database portal. Real-time qPCR and western blot analyses were utilized to determine the expression levels of CDCA4. Gain- and loss-of-function of CDCA4 assays were conducted with transfection technology to investigate the biological role of CDCA4 in Wilm’s tumor cells. Cell counting kit 8 and flow cytometer assays were employed to examine the effect of CDCA4 on the cells proliferation and apoptosis. Protein expression levels of indicated markers in each group of Wilm’s tumor cells were measured by western blot. Results The transcriptional expression of CDCA4 was drastically upregulated in Wilm’s tumor tissues according to the public TARGET database and in Wilm’s tumor cells. The cells viability was remarkably reduced whereas the cells apoptosis was increased in CDCA4-knockdown group compared with negative control group. However, CDCA4-overexpression group promoted the cells proliferation and suppressed the cells apoptosis. Furthermore, the protein expression levels of p-AKT, p-mTOR, and Cyclin D1 were significantly reduced after depletion of CDCA4, whereas overexpression of CDCA4 dramatically elevated these markers’ expression levels. Conclusions CDCA4 is highly expressed in Wilm’s tumor and promoted the proliferation whereas inhibited the apoptosis of Wilm’s tumor cells through activating the AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Suqing Li
- Department of Pediatrics, Guigang City People's Hospital, the Eight Affiliated Hospital of Guangxi Medical University, Guigang, P.R. China
| | - Cong Qin
- Department of Pediatrics, Guigang City People's Hospital, the Eight Affiliated Hospital of Guangxi Medical University, Guigang, P.R. China
| | - Yike Chen
- Department of Pediatrics, Guigang City People's Hospital, the Eight Affiliated Hospital of Guangxi Medical University, Guigang, P.R. China
| | - Dan Wei
- Department of Pediatrics, the First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Zhijun Tan
- Department of Pediatrics, Guigang City People's Hospital, the Eight Affiliated Hospital of Guangxi Medical University, Guigang, P.R. China
| | - Jiadong Meng
- Department of Pediatrics, Guigang City People's Hospital, the Eight Affiliated Hospital of Guangxi Medical University, Guigang, P.R. China
| |
Collapse
|
23
|
Wang PC, Chen ST, Yang ZM. Effects of Aurora kinase A on mouse decidualization via Stat3-plk1-cdk1 pathway. Reprod Biol Endocrinol 2021; 19:162. [PMID: 34715887 PMCID: PMC8557062 DOI: 10.1186/s12958-021-00847-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Decidualization is essential to the successful pregnancy in mice. The molecular mechanisms and effects of Aurora kinase A (Aurora A) remain poorly understood during pregnancy. This study is the first to investigate the expression and role of Aurora A during mouse decidualization. METHODS Quantitative real time polymerase chain reaction, western blotting and in situ hybridization were used to determine the expression of Aurora A in mouse uteri. Aurora A activity was inhibited by Aurora A inhibitor to explore the role of Aurora A on decidualization via regulating the Aurora A/Stat3/Plk1/Cdk1 signaling pathway. RESULTS Aurora A was strongly expressed at implantation sites compared with inter-implantation sites. Furthermore, Aurora A was also significantly increased in oil-induced deciduoma compared with control. Both Aurora A mRNA and protein were significantly increased under in vitro decidualization. Under in vitro decidualization, Prl8a2, a marker of mouse decidualization, was significantly decreased by TC-S 7010, an Aurora A inhibitor. Additionally, Prl8a2 was reduced by Stat3 inhibitor, Plk1 inhibitor and Cdk1 inhibitor, respectively. Moreover, the protein levels of p-Stat3, p-Plk1 and p-Cdk1 were suppressed by TC-S 7010. The protein levels of p-Stat3, p-Plk1 and p-Cdk1 were also suppressed by S3I-201, a Stat3 inhibitor). SBE 13 HCl (Plk1 inhibitor) could reduce the protein levels of p-Plk1 and p-Cdk1. Collectively, Aurora A could regulate Stat3/Plk1/Cdk1 signaling pathway. CONCLUSION Our study shows that Aurora A is expressed in decidual cells and should be important for mouse decidualization. Aurora A/Stat3/Plk1/Cdk1 signaling pathway may be involved in mouse decidualization.
Collapse
Affiliation(s)
- Peng-Chao Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801 China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Si-Ting Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Zeng-Ming Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| |
Collapse
|
24
|
Chen X, Xia Z, Wan Y, Huang P. Identification of hub genes and candidate drugs in hepatocellular carcinoma by integrated bioinformatics analysis. Medicine (Baltimore) 2021; 100:e27117. [PMID: 34596112 PMCID: PMC8483840 DOI: 10.1097/md.0000000000027117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 08/14/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the third cancer-related cause of death in the world. Until now, the involved mechanisms during the development of HCC are largely unknown. This study aims to explore the driven genes and potential drugs in HCC. METHODS Three mRNA expression datasets were used to analyze the differentially expressed genes (DEGs) in HCC. The bioinformatics approaches include identification of DEGs and hub genes, Gene Ontology terms analysis and Kyoto encyclopedia of genes and genomes enrichment analysis, construction of protein-protein interaction network. The expression levels of hub genes were validated based on The Cancer Genome Atlas, Gene Expression Profiling Interactive Analysis, and the Human Protein Atlas. Moreover, overall survival and disease-free survival analysis of HCC patients were further conducted by Kaplan-Meier plotter and Gene Expression Profiling Interactive Analysis. DGIdb database was performed to search the candidate drugs for HCC. RESULTS A total of 197 DEGs were identified. The protein-protein interaction network was constructed using Search Tool for the Retrieval of Interacting Genes software, 10 genes were selected by Cytoscape plugin cytoHubba and served as hub genes. These 10 genes were all closely related to the survival of HCC patients. DGIdb database predicted 29 small molecules as the possible drugs for treating HCC. CONCLUSION Our study provides some new insights into HCC pathogenesis and treatments. The candidate drugs may improve the efficiency of HCC therapy in the future.
Collapse
Affiliation(s)
- Xiaolong Chen
- National Key Clinical Department, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhixiong Xia
- Department of Pathology, The Center Hospital of Wuhan, Hubei, China
| | - Yafeng Wan
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Ping Huang
- National Key Clinical Department, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
25
|
Tariq MU, Furqan M, Parveen H, Ullah R, Muddassar M, Saleem RSZ, Bavetsias V, Linardopoulos S, Faisal A. CCT245718, a dual FLT3/Aurora A inhibitor overcomes D835Y-mediated resistance to FLT3 inhibitors in acute myeloid leukaemia cells. Br J Cancer 2021; 125:966-974. [PMID: 34446858 DOI: 10.1038/s41416-021-01527-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/17/2021] [Accepted: 08/11/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Activating mutations in the Fms-like tyrosine kinase 3 (FLT3) are among the most prevalent oncogenic mutations in acute myeloid leukaemia. Inhibitors selectively targeting FLT3 kinase have shown promising clinical activity; their success in the clinic, however, has been limited due to the emergence of acquired resistance. METHODS CCT245718 was identified and characterised as a dual Aurora A/FLT3 inhibitor through cell-based and biochemical assays. The ability of CCT245718 to overcome TKD-mediated resistance was evaluated in a cell line-based model of drug resistance to FLT3 inhibitors. RESULTS CCT245718 exhibits potent antiproliferative activity towards FLT3-ITD + AML cell lines and strongly binds to FLT3-ITD and TKD (D835Y) mutants in vitro. Activities of both FLT3-ITD and Aurora A are also inhibited in cells. Inhibition of FLT3 results in reduced phosphorylation of STAT5, downregulation of survivin and induction of apoptotic cell death. Moreover, CCT245718 overcomes TKD-mediated resistance in a MOLM-13-derived cell line containing FLT3 with both ITD and D835Y mutations. It also inhibits FLT3 signalling in both parental and resistant cell lines compared to FLT3-specific inhibitor MLN518, which is only active in the parental cell line. CONCLUSIONS Our results demonstrate that CCT245718 is a potent dual FLT3/Aurora A inhibitor that can overcome TKD-mediated acquired resistance.
Collapse
Affiliation(s)
- Muhammad Usama Tariq
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Muhammad Furqan
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Hira Parveen
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Rahim Ullah
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Muhammad Muddassar
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Rahman Shah Zaib Saleem
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Vassilios Bavetsias
- Cancer Research UK, Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Spiros Linardopoulos
- Cancer Research UK, Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK.,Breast Cancer Now, Division of Breast Cancer Research, The Institute of Cancer Research, London, UK.,AstraZeneca, Cambridge, UK
| | - Amir Faisal
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan.
| |
Collapse
|
26
|
Liu L, Qu J, Dai Y, Qi T, Teng X, Li G, Qu Q. An interactive nomogram based on clinical and molecular signatures to predict prognosis in multiple myeloma patients. Aging (Albany NY) 2021; 13:18442-18463. [PMID: 34260414 PMCID: PMC8351694 DOI: 10.18632/aging.203294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 06/23/2021] [Indexed: 12/29/2022]
Abstract
Although novel drugs and treatments have been developed and improved, multiple myeloma (MM) is still recurrent and difficult to cure. In the present study, the magenta module containing 400 hub genes was determined from the training dataset of GSE24080 through weighted gene co-expression network analysis (WGCNA). Then, using the least absolute shrinkage and selection operator (Lasso) analysis, a fifteen-gene signature was firstly selected and the predictive performance for overall survival (OS) was favorable, which was identified by Receiver Operating Characteristic (ROC) curves. The risk score model was constructed based on survival-associated fifteen genes from the Lasso model, which classified MM patients into high-risk and low-risk groups. Areas under the curve (AUC) of ROC curve and log-rank test showed that the high-risk group was correlated to the dismal survival outcome of MM patients, which was also identified in testing dataset of GSE9782. The calibration plot, the AUC value of the ROC curve and Concordance-index showed that the interactive nomogram with risk score could favorably predict the probability of multi-year OS of MM patients. Therefore, it may help clinicians make a precise therapeutic decision based on the easy-to-use tool of the nomogram.
Collapse
Affiliation(s)
- Linxin Liu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Qu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Yuxin Dai
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, China
| | - Tingting Qi
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xinqi Teng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Guohua Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Qiang Qu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
27
|
Machado CB, DA Silva EL, Dias Nogueira BM, DA Silva JBS, DE Moraes Filho MO, Montenegro RC, DE Moraes MEA, Moreira-Nunes CA. The Relevance of Aurora Kinase Inhibition in Hematological Malignancies. CANCER DIAGNOSIS & PROGNOSIS 2021; 1:111-126. [PMID: 35399305 DOI: 10.21873/cdp.10016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/27/2021] [Indexed: 12/26/2022]
Abstract
Aurora kinases are a family of serine/threonine protein kinases that play a central role in eukaryotic cell division. Overexpression of aurora kinases in cancer and their role as major regulators of the cell cycle quickly inspired the idea that their inhibition might be a potential pathway when treating oncologic patients. Over the past couple of decades, the search for designing and testing of molecules capable of inhibiting aurora activities fueled many pre-clinical and clinical studies. In this study, data from the past 10 years of in vitro and in vivo investigations, as well as clinical trials, utilizing aurora kinase inhibitors as therapeutics for hematological malignancies were compiled and discussed, aiming to highlight potential uses of these inhibitors as a novel monotherapy model or alongside conventional chemotherapies. While there is still much to be elucidated, it is clear that these kinases play a key role in oncogenesis, and their manageable toxicity and potentially synergistic effects still render them a focus of interest for future investigations in combinatorial clinical trials.
Collapse
Affiliation(s)
- Caio Bezerra Machado
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Emerson Lucena DA Silva
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Beatriz Maria Dias Nogueira
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Jean Breno Silveira DA Silva
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Manoel Odorico DE Moraes Filho
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Raquel Carvalho Montenegro
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Caroline Aquino Moreira-Nunes
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
28
|
Arslanhan MD, Rauniyar N, Yates JR, Firat-Karalar EN. Aurora Kinase A proximity map reveals centriolar satellites as regulators of its ciliary function. EMBO Rep 2021; 22:e51902. [PMID: 34169630 PMCID: PMC8339716 DOI: 10.15252/embr.202051902] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 05/19/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022] Open
Abstract
Aurora kinase A (AURKA) is a conserved kinase that plays crucial roles in numerous cellular processes. Although AURKA overexpression is frequent in human cancers, its pleiotropic functions and multifaceted regulation present challenges in its therapeutic targeting. Key to overcoming these challenges is to identify and characterize the full range of AURKA interactors, which are often weak and transient. Previous proteomic studies were limited in monitoring dynamic and non-mitotic AURKA interactions. Here, we generate the proximity interactome of AURKA in asynchronous cells, which consists of 440 proteins involving multiple biological processes and cellular compartments. Importantly, AURKA has extensive proximate and physical interactions to centriolar satellites, key regulators of the primary cilium. Loss-of-function experiments identify satellites as negative regulators of AURKA activity, abundance, and localization in quiescent cells. Notably, loss of satellites activates AURKA at the basal body, decreases centrosomal IFT88 levels, and causes ciliogenesis defects. Collectively, our results provide a resource for dissecting spatiotemporal regulation of AURKA and uncover its proteostatic regulation by satellites as a new mechanism for its ciliary functions.
Collapse
Affiliation(s)
- Melis D Arslanhan
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Navin Rauniyar
- Department of Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - John R Yates
- Department of Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | | |
Collapse
|
29
|
Zangouei AS, Moghbeli M. MicroRNAs as the critical regulators of cisplatin resistance in gastric tumor cells. Genes Environ 2021; 43:21. [PMID: 34099061 PMCID: PMC8182944 DOI: 10.1186/s41021-021-00192-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/24/2021] [Indexed: 12/13/2022] Open
Abstract
Combined chemotherapeutic treatment is the method of choice for advanced and metastatic gastric tumors. However, resistance to chemotherapeutic agents is one of the main challenges for the efficient gastric cancer (GC) treatment. Cisplatin (CDDP) is used as an important regimen of chemotherapy for GC which induces cytotoxicity by interfering with DNA replication in cancer cells and inducing their apoptosis. Majority of patients experience cisplatin-resistance which is correlated with tumor metastasis and relapse. Moreover, prolonged and high-dose cisplatin administrations cause serious side effects such as nephrotoxicity, ototoxicity, and anemia. Since, there is a high rate of recurrence after CDDP treatment in GC patients; it is required to clarify the molecular mechanisms associated with CDDP resistance to introduce novel therapeutic methods. There are various cell and molecular processes associated with multidrug resistance (MDR) including drug efflux, detoxification, DNA repair ability, apoptosis alteration, signaling pathways, and epithelial-mesenchymal transition (EMT). MicroRNAs are a class of endogenous non-coding RNAs involved in chemo resistance of GC cells through regulation of all of the MDR mechanisms. In present review we have summarized all of the miRNAs associated with cisplatin resistance based on their target genes and molecular mechanisms in gastric tumor cells. This review paves the way of introducing a miRNA-based panel of prognostic markers to improve the efficacy of chemotherapy and clinical outcomes in GC patients. It was observed that miRNAs are mainly involved in cisplatin response of gastric tumor cells via regulation of signaling pathways, autophagy, and apoptosis.
Collapse
Affiliation(s)
- Amir Sadra Zangouei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
30
|
Miura A, Sootome H, Fujita N, Suzuki T, Fukushima H, Mizuarai S, Masuko N, Ito K, Hashimoto A, Uto Y, Sugimoto T, Takahashi H, Mitsuya M, Hirai H. TAS-119, a novel selective Aurora A and TRK inhibitor, exhibits antitumor efficacy in preclinical models with deregulated activation of the Myc, β-Catenin, and TRK pathways. Invest New Drugs 2021; 39:724-735. [PMID: 33409897 DOI: 10.1007/s10637-020-01019-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022]
Abstract
Aurora kinase A, a mitotic kinase that is overexpressed in various cancers, is a promising cancer drug target. Here, we performed preclinical characterization of TAS-119, a novel, orally active, and highly selective inhibitor of Aurora A. TAS-119 showed strong inhibitory effect against Aurora A, with an IC50 value of 1.04 nmol/L. The compound was highly selective for Aurora A compared with 301 other protein kinases, including Aurora kinase B. TAS-119 induced the inhibition of Aurora A and accumulation of mitotic cells in vitro and in vivo. It suppressed the growth of various cancer cell lines harboring MYC family amplification and CTNNB1 mutation in vitro. In a xenograft model of human lung cancer cells harboring MYC amplification and CTNNB1 mutation, TAS-119 showed a strong antitumor activity at well-tolerated doses. TAS-119 induced N-Myc degradation and inhibited downstream transcriptional targets in MYCN-amplified neuroblastoma cell lines. It also demonstrated inhibitory effect against tropomyosin receptor kinase (TRK)A, TRKB, and TRKC, with an IC50 value of 1.46, 1.53, and 1.47 nmol/L, respectively. TAS-119 inhibited TRK-fusion protein activity and exhibited robust growth inhibition of tumor cells via a deregulated TRK pathway in vitro and in vivo. Our study indicates the potential of TAS-119 as an anticancer drug, especially for patients harboring MYC amplification, CTNNB1 mutation, and NTRK fusion.
Collapse
Affiliation(s)
- Akihiro Miura
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, 3 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 770-8506, 2-1 Minamijosanjima-cho, Tokushima, Japan
| | - Hiroshi Sootome
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, 3 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Naoya Fujita
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, 3 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Takamasa Suzuki
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, 3 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Hiroto Fukushima
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, 3 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Shinji Mizuarai
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, 3 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Norio Masuko
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, 3 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Kimihiro Ito
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, 3 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Akihiro Hashimoto
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, 3 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Yoshihiro Uto
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 770-8506, 2-1 Minamijosanjima-cho, Tokushima, Japan
| | - Tetsuya Sugimoto
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, 3 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Hidekazu Takahashi
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, 3 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Morihiro Mitsuya
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, 3 Okubo, Tsukuba, Ibaraki, 300-2611, Japan
| | - Hiroshi Hirai
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, 3 Okubo, Tsukuba, Ibaraki, 300-2611, Japan.
| |
Collapse
|
31
|
Elie J, Feizbakhsh O, Desban N, Josselin B, Baratte B, Bescond A, Duez J, Fant X, Bach S, Marie D, Place M, Ben Salah S, Chartier A, Berteina-Raboin S, Chaikuad A, Knapp S, Carles F, Bonnet P, Buron F, Routier S, Ruchaud S. Design of new disubstituted imidazo[1,2- b]pyridazine derivatives as selective Haspin inhibitors. Synthesis, binding mode and anticancer biological evaluation. J Enzyme Inhib Med Chem 2021; 35:1840-1853. [PMID: 33040634 PMCID: PMC7580722 DOI: 10.1080/14756366.2020.1825408] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Haspin is a mitotic protein kinase required for proper cell division by modulating Aurora B kinase localisation and activity as well as histone phosphorylation. Here a series of imidazopyridazines based on the CHR-6494 and Structure Activity Relationship was established. An assessment of the inhibitory activity of the lead structures on human Haspin and several other protein kinases is presented. The lead structure was rapidly optimised using a combination of crystal structures and effective docking models, with the best inhibitors exhibiting potent inhibitory activity on Haspin with IC50 between 6 and 100 nM in vitro. The developed inhibitors displayed anti-proliferative properties against various human cancer cell lines in 2D and spheroid cultures and significantly inhibited the migration ability of osteosarcoma U-2 OS cells. Notably, we show that our lead compounds are powerful Haspin inhibitors in human cells, and did not block G2/M cell cycle transition due to improved selectivity against CDK1/CyclinB.
Collapse
Affiliation(s)
- Jonathan Elie
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans Cedex 2, France
| | - Omid Feizbakhsh
- Sorbonne Université/CNRS UMR8227, Station Biologique, Roscoff cedex, France
| | - Nathalie Desban
- Sorbonne Université/CNRS UMR8227, Station Biologique, Roscoff cedex, France
| | - Béatrice Josselin
- Sorbonne Université/CNRS UMR8227, Station Biologique, Roscoff cedex, France.,Sorbonne Université/CNRS FR2424, Plateforme de criblage KISSf (Kinase Inhibitor Specialized Screening facility) Station Biologique, Roscoff cedex, France
| | - Blandine Baratte
- Sorbonne Université/CNRS UMR8227, Station Biologique, Roscoff cedex, France.,Sorbonne Université/CNRS FR2424, Plateforme de criblage KISSf (Kinase Inhibitor Specialized Screening facility) Station Biologique, Roscoff cedex, France
| | - Amandine Bescond
- Sorbonne Université/CNRS UMR8227, Station Biologique, Roscoff cedex, France
| | - Julien Duez
- Sorbonne Université/CNRS UMR8227, Station Biologique, Roscoff cedex, France
| | - Xavier Fant
- Sorbonne Université/CNRS UMR8227, Station Biologique, Roscoff cedex, France
| | - Stéphane Bach
- Sorbonne Université/CNRS UMR8227, Station Biologique, Roscoff cedex, France.,Sorbonne Université/CNRS FR2424, Plateforme de criblage KISSf (Kinase Inhibitor Specialized Screening facility) Station Biologique, Roscoff cedex, France
| | - Dominique Marie
- Sorbonne Université/CNRS UMR7144, Station Biologique, Roscoff cedex, France
| | - Matthieu Place
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans Cedex 2, France
| | - Sami Ben Salah
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans Cedex 2, France
| | - Agnes Chartier
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans Cedex 2, France
| | - Sabine Berteina-Raboin
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans Cedex 2, France
| | - Apirat Chaikuad
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Frankfurt am Main, Germany.,Structure Genomics Consortium, Johann Wolfgang Goethe University, Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany
| | - Stefan Knapp
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Frankfurt am Main, Germany.,Structure Genomics Consortium, Johann Wolfgang Goethe University, Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany
| | - Fabrice Carles
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans Cedex 2, France
| | - Pascal Bonnet
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans Cedex 2, France
| | - Frédéric Buron
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans Cedex 2, France
| | - Sylvain Routier
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans Cedex 2, France
| | - Sandrine Ruchaud
- Sorbonne Université/CNRS UMR8227, Station Biologique, Roscoff cedex, France
| |
Collapse
|
32
|
Fragliasso V, Tameni A, Inghirami G, Mularoni V, Ciarrocchi A. Cytoskeleton Dynamics in Peripheral T Cell Lymphomas: An Intricate Network Sustaining Lymphomagenesis. Front Oncol 2021; 11:643620. [PMID: 33928032 PMCID: PMC8076600 DOI: 10.3389/fonc.2021.643620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/17/2021] [Indexed: 12/04/2022] Open
Abstract
Defects in cytoskeleton functions support tumorigenesis fostering an aberrant proliferation and promoting inappropriate migratory and invasive features. The link between cytoskeleton and tumor features has been extensively investigated in solid tumors. However, the emerging genetic and molecular landscape of peripheral T cell lymphomas (PTCL) has unveiled several alterations targeting structure and function of the cytoskeleton, highlighting its role in cell shape changes and the aberrant cell division of malignant T cells. In this review, we summarize the most recent evidence about the role of cytoskeleton in PTCLs development and progression. We also discuss how aberrant signaling pathways, like JAK/STAT3, NPM-ALK, RhoGTPase, and Aurora Kinase, can contribute to lymphomagenesis by modifying the structure and the signaling properties of cytoskeleton.
Collapse
Affiliation(s)
- Valentina Fragliasso
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Annalisa Tameni
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Valentina Mularoni
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
33
|
Abstract
Introduction: Aurora kinases are a family of serine/threonine kinases, and promote mitotic spindle assembly by regulating centrosome duplication and separation. Aurora kinases are overexpressed in a variety of tumor cell lines, thus, the use of Aurora kinase small-molecule inhibitors has become a potential treatment option for cancer.Areas covered: As a continuing review of Aurora kinase inhibitors and their patents published in 2009, 2011 and 2014. Herein, we updated the information for Aurora kinase inhibitors in clinical trials and the patents filed from 2014 to 2020. PubMed, Scopus, SciFinder, and www.clinicaltrials.gov databases were used for searching the clinical information and patents of Aurora kinase inhibitors.Expert opinion: Even though Aurora A or B selective as well as pan inhibitors show preclinical and clinical efficacy, so far, no Aurora kinase inhibitor has been approved for clinical use. Preliminary evidence suggested that highly selective Aurora kinase or multi-target inhibitors as a single agent as well as in combination therapy are still the current main development trend of Aurora kinase inhibitors.
Collapse
Affiliation(s)
- Xue-Li Jing
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Shi-Wu Chen
- School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
34
|
Karatas M, Chaikuad A, Berger B, Kubbutat MHG, Totzke F, Knapp S, Kunick C. 7-(2-Anilinopyrimidin-4-yl)-1-benzazepin-2-ones Designed by a "Cut and Glue" Strategy Are Dual Aurora A/VEGF-R Kinase Inhibitors. Molecules 2021; 26:molecules26061611. [PMID: 33799460 PMCID: PMC7998669 DOI: 10.3390/molecules26061611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/06/2021] [Accepted: 03/07/2021] [Indexed: 11/16/2022] Open
Abstract
Although overexpression and hyperactivity of protein kinases are causative for a wide range of human cancers, protein kinase inhibitors currently approved as cancer drugs address only a limited number of these enzymes. To identify new chemotypes addressing alternative protein kinases, the basic structure of a known PLK1/VEGF-R2 inhibitor class was formally dissected and reassembled. The resulting 7-(2-anilinopyrimidin-4-yl)-1-benzazepin-2-ones were synthesized and proved to be dual inhibitors of Aurora A kinase and VEGF receptor kinases. Crystal structures of two representatives of the new chemotype in complex with Aurora A showed the ligand orientation in the ATP binding pocket and provided the basis for rational structural modifications. Congeners with attached sulfamide substituents retained Aurora A inhibitory activity. In vitro screening of two members of the new kinase inhibitor family against the cancer cell line panel of the National Cancer Institute (NCI) showed antiproliferative activity in the single-digit micromolar concentration range in the majority of the cell lines.
Collapse
Affiliation(s)
- Mehmet Karatas
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany; (M.K.); (B.B.)
- Zentrum für Pharmaverfahrenstechnik (PVZ), Technische Universität Braunschweig, Franz-Liszt-Straße 35A, 38106 Braunschweig, Germany
| | - Apirat Chaikuad
- Structural Genomics Consortium, BMLS, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany; (A.C.); (S.K.)
- Institut für Pharmazeutische Chemie, Johann Wolfgang-Goethe-Universität, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Bianca Berger
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany; (M.K.); (B.B.)
| | | | - Frank Totzke
- Reaction Biology Europe GmbH, 79108 Freiburg, Germany; (M.H.G.K.); (F.T.)
| | - Stefan Knapp
- Structural Genomics Consortium, BMLS, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany; (A.C.); (S.K.)
- Institut für Pharmazeutische Chemie, Johann Wolfgang-Goethe-Universität, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Conrad Kunick
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany; (M.K.); (B.B.)
- Zentrum für Pharmaverfahrenstechnik (PVZ), Technische Universität Braunschweig, Franz-Liszt-Straße 35A, 38106 Braunschweig, Germany
- Correspondence: ; Tel.: +49-531-391-2754
| |
Collapse
|
35
|
Mittal K, Kaur J, Jaczko M, Wei G, Toss MS, Rakha EA, Janssen EAM, Søiland H, Kucuk O, Reid MD, Gupta MV, Aneja R. Centrosome amplification: a quantifiable cancer cell trait with prognostic value in solid malignancies. Cancer Metastasis Rev 2021; 40:319-339. [PMID: 33106971 PMCID: PMC7897259 DOI: 10.1007/s10555-020-09937-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
Numerical and/or structural centrosome amplification (CA) is a hallmark of cancers that is often associated with the aberrant tumor karyotypes and poor clinical outcomes. Mechanistically, CA compromises mitotic fidelity and leads to chromosome instability (CIN), which underlies tumor initiation and progression. Recent technological advances in microscopy and image analysis platforms have enabled better-than-ever detection and quantification of centrosomal aberrancies in cancer. Numerous studies have thenceforth correlated the presence and the degree of CA with indicators of poor prognosis such as higher tumor grade and ability to recur and metastasize. We have pioneered a novel semi-automated pipeline that integrates immunofluorescence confocal microscopy with digital image analysis to yield a quantitative centrosome amplification score (CAS), which is a summation of the severity and frequency of structural and numerical centrosome aberrations in tumor samples. Recent studies in breast cancer show that CA increases across the disease progression continuum, while normal breast tissue exhibited the lowest CA, followed by cancer-adjacent apparently normal, ductal carcinoma in situ and invasive tumors, which showed the highest CA. This finding strengthens the notion that CA could be evolutionarily favored and can promote tumor progression and metastasis. In this review, we discuss the prevalence, extent, and severity of CA in various solid cancer types, the utility of quantifying amplified centrosomes as an independent prognostic marker. We also highlight the clinical feasibility of a CA-based risk score for predicting recurrence, metastasis, and overall prognosis in patients with solid cancers.
Collapse
Affiliation(s)
- Karuna Mittal
- Department of Biology, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA
| | - Jaspreet Kaur
- Department of Biology, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA
| | - Meghan Jaczko
- Department of Biology, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA
| | - Guanhao Wei
- Department of Biology, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA
| | - Michael S Toss
- Department of Pathology, University of Nottingham and Nottingham University Hospitals, Nottingham, UK
| | - Emad A Rakha
- Department of Pathology, University of Nottingham and Nottingham University Hospitals, Nottingham, UK
| | | | - Håvard Søiland
- Department of Breast and Endocrine Surgery, Stavanger University Hospital, Stavanger, Norway
| | - Omer Kucuk
- Winship Cancer Institute, Department of Hematology and Medical Oncology, Emory University Hospital, Atlanta, GA, USA
| | | | | | - Ritu Aneja
- Department of Biology, Georgia State University, 100 Piedmont Ave, Atlanta, GA, 30303, USA.
| |
Collapse
|
36
|
Inhibition of Aurora Kinase B activity disrupts development and differentiation of salivary glands. Cell Death Discov 2021; 7:16. [PMID: 33462217 PMCID: PMC7814035 DOI: 10.1038/s41420-020-00393-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/24/2020] [Accepted: 12/14/2020] [Indexed: 11/13/2022] Open
Abstract
Little is known about the key molecules that regulate cell division during organogenesis. Here we determine the role of the cell cycle promoter aurora kinase B (AURKB) during development, using embryonic salivary glands (E-SGs) as a model. AURKB is a serine/threonine kinase that regulates key events in mitosis, which makes it an attractive target for tailored anticancer therapy. Many reports have elaborated on the role of AURKB in neoplasia and cancer; however, no previous study has shown its role during organ development. Our previous experiments have highlighted the essential requirement for AURKB during adult exocrine regeneration. To investigate if AURKB is similarly required for progression during embryonic development, we pharmacologically inhibited AURKB in developing submandibular glands (SMGs) at embryonic day (E)13.5 and E16.5, using the highly potent and selective drug Barasertib. Inhibition of AURKB interfered with the expansion of the embryonic buds. Interestingly, this effect on SMG development was also seen when the mature explants (E16.5) were incubated for 24 h with another cell cycle inhibitor Aphidicolin. Barasertib prompted apoptosis, DNA damage and senescence, the markers of which (cleaved caspase 3, γH2AX, SA-βgal and p21, respectively), were predominantly seen in the developing buds. In addition to a reduction in cell cycling and proliferation of the epithelial cells in response to AURKB inhibition, Barasertib treatment led to an excessive generation of reactive oxygen species (ROS) that resulted in downregulation of the acinar differentiation marker Mist1. Importantly, inhibition of ROS was able to rescue this loss of identity, with Mist1 expression maintained despite loss of AURKB. Together, these data identify AURKB as a key molecule in supporting embryonic development and differentiation, while inhibiting senescence-inducing signals during organogenesis.
Collapse
|
37
|
Juillet C, Ermolenko L, Boyarskaya D, Baratte B, Josselin B, Nedev H, Bach S, Iorga BI, Bignon J, Ruchaud S, Al-Mourabit A. From Synthetic Simplified Marine Metabolite Analogues to New Selective Allosteric Inhibitor of Aurora B Kinase. J Med Chem 2021; 64:1197-1219. [PMID: 33417773 DOI: 10.1021/acs.jmedchem.0c02064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significant inhibition of Aurora B was achieved by the synthesis of simplified fragments of benzosceptrins and oroidin belonging to the marine pyrrole-2-aminoimidazoles metabolites isolated from sponges. Evaluation of kinase inhibition enabled the discovery of a synthetically accessible rigid acetylenic structural analogue EL-228 (1), whose structure could be optimized into the potent CJ2-150 (37). Here we present the synthesis of new inhibitors of Aurora B kinase, which is an important target for cancer therapy through mitosis regulation. The biologically oriented synthesis yielded several nanomolar inhibitors. The optimized compound CJ2-150 (37) showed a non-ATP competitive allosteric mode of action in a mixed-type inhibition for Aurora B kinase. Molecular docking identified a probable binding mode in the allosteric site "F" and highlighted the key interactions with the protein. We describe the improvement of the inhibitory potency and specificity of the novel scaffold as well as the characterization of the mechanism of action.
Collapse
Affiliation(s)
- Charlotte Juillet
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| | - Ludmila Ermolenko
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| | - Dina Boyarskaya
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| | - Blandine Baratte
- Plateforme de Criblage KISSf, Station Biologique de Roscoff, Sorbonne Université, CNRS, FR 2424, Roscoff, 29680, France
| | - Béatrice Josselin
- Plateforme de Criblage KISSf, Station Biologique de Roscoff, Sorbonne Université, CNRS, FR 2424, Roscoff, 29680, France
| | - Hristo Nedev
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| | - Stéphane Bach
- Plateforme de Criblage KISSf, Station Biologique de Roscoff, Sorbonne Université, CNRS, FR 2424, Roscoff, 29680, France.,Integrative Biology of Marine Models Laboratory (LBI2M), Station Biologique de Roscoff, Sorbonne Université, CNRS, UMR 8227, Roscoff, 29680, France
| | - Bogdan I Iorga
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| | - Jérôme Bignon
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| | - Sandrine Ruchaud
- Integrative Biology of Marine Models Laboratory (LBI2M), Station Biologique de Roscoff, Sorbonne Université, CNRS, UMR 8227, Roscoff, 29680, France
| | - Ali Al-Mourabit
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| |
Collapse
|
38
|
Nikzamir A, Rezaei-Tavirani M, Razzaghi Z, Rostami-Nejad M, Hamdieh M, Arjmand B. Gene Activation as a Cell Protection Mechanism Against Gamma-Ray radiation. J Lasers Med Sci 2020; 11:S80-S84. [PMID: 33995974 DOI: 10.34172/jlms.2020.s13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Introduction: Gamma radiation is accompanied by prominent biological effects and damages. Cell proliferation and tumorigenesis are highlighted as the main resulted effects of gamma radiation on cultured cells. This study aims to assess the dysregulated mode of gene function after gamma radiation in human Jurkat cells. Methods: Six gene expression profiles from Gene Expression Omnibus (GEO) were analyzed by GEO2R to find the significant differentially expressed genes (DEGs) via gamma radiation. Action map analysis was applied to screen the query DEGs. Results: Among 108 study genes, 20 critical DEGs including AURKA, AURKB, BORA, CCNB1, CCNB2, CCNF, CDC20, CDCA8, CENPA, CENPE, CENPF, KIF18A, KIF20A, KIF23, BUB1, DLGAP5, ECT2, PLK1, SGO2, and TPX2 were introduced as down-regulated genes by the gamma ray. Conclusion: Activators of the introduced critical genes may be the cell protector against gamma radiation.
Collapse
Affiliation(s)
- Abdolrahim Nikzamir
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Razzaghi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Rostami-Nejad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Hamdieh
- Department of Psychosomatic, Taleghani Hospital, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Ekebergh A, Mårtensson J, Ekebergh CL. Cyclopenta[ b]indole Derivative Inhibits Aurora B in Primary Cells. ACS OMEGA 2020; 5:33455-33460. [PMID: 33403307 PMCID: PMC7774273 DOI: 10.1021/acsomega.0c05491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 11/27/2020] [Indexed: 06/12/2023]
Abstract
The Aurora family of kinases is closely involved in regulating cell division. Inhibition of Aurora A and B with small molecules is currently being investigated in clinical trials for the treatment of different cancers. It has also been evaluated as a treatment option against different autoimmune diseases in preclinical studies. Here, we present a cyclopenta[b]indole derivative capable of inhibiting Aurora B selectively in kinase assays. To evaluate the Aurora B inhibition capacity of the compound, we used a kinase IC50 assay as well as a suppression assay of proliferating primary cells. In addition, we examined if the cells had gained a phenotype characteristic for Aurora B inhibition after treatment with the compound. We found that the compound selectively inhibited Aurora B (IC50 = 1.4 μM) over Aurora A (IC50 > 30 μM). Moreover, the compound inhibited proliferating PBMCs with an IC50 = 4.2 μM, and the cells displayed reduced phosphorylation of histone H3 as well as tetraploidy, consistent with Aurora B inhibition.
Collapse
Affiliation(s)
- Andreas Ekebergh
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Jerker Mårtensson
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Christine Lingblom Ekebergh
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 46, Sweden
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg 413 46, Sweden
| |
Collapse
|
40
|
Malami I, Bunza AM, Alhassan AM, Muhammad A, Abubakar IB, Yunusa A, Waziri PM, Etti IC. Dihydroartemisinin as a potential drug candidate for cancer therapy: a structural-based virtual screening for multitarget profiling. J Biomol Struct Dyn 2020; 40:1347-1362. [PMID: 32964804 DOI: 10.1080/07391102.2020.1824811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cancer is a rapidly growing non-communicable disease worldwide that is responsible for high mortality rates, which account for 9.6 million death in 2018. Dihydroartemisinin (DHA) is an active metabolite of artemisinin, an active principle present in the Chinese medicinal plant Artemisia annua used for malaria treatment. Dihydroartemisinin possesses remarkable and selective anticancer properties however the underlying mechanism of the antitumor effects of DHA from the structural point of view is still not yet elucidated. In the present study, we employed molecular docking simulation techniques using Autodock suits to access the binding properties of dihydroartemisinin to multiple protein targets implicated in cancer pathogenesis. Its potential targets with comprehensive pharmacophore were predicted using a PharmMapper database. The co-crystallised structures of the protein were obtained from a Protein Data Bank and prepared for molecular docking simulation. Out of the 24 selected protein targets, DHA has shown about 29% excellent binding to the targets compared to their co-crystallised ligand. Additionally, 75% of the targets identified for dihydroartemisinin binding are protein kinases, and 25% are non-protein kinases. Hydroxyl functional group of dihydroartemisinin contributed to 58.5% of the total hydrogen interactions, while pyran (12.2%), endoperoxide (9.8%), and oxepane (19.5%) contributed to the remaining hydrogen bonding. The present findings have elucidated the possible antitumor properties of dihydroartemisinin through the structural-based virtual studies, which provides a lead to a safe and effective anticancer agent useful for cancer therapy.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ibrahim Malami
- Department of Pharmacognosy and Ethnopharmacy, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria.,Centre for Advanced Medical Research and Training (CAMRET), Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Aisha Muktar Bunza
- Department of Pharmacognosy and Ethnopharmacy, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Alhassan Muhammad Alhassan
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Aliyu Muhammad
- Department of Biochemistry, Faculty of life Sciences, Ahmadu Bello University, Zaria, Nigeria
| | | | - Abdulmajeed Yunusa
- Department of Pharmacology and Therapeutics, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Peter M Waziri
- Department of Biochemistry, Kaduna State University, Kaduna, Nigeria
| | - Imaobong C Etti
- Department of Pharmacology and Toxicology, University of Uyo, Uyo, Nigeria
| |
Collapse
|
41
|
AURKA Increase the Chemosensitivity of Colon Cancer Cells to Oxaliplatin by Inhibiting the TP53-Mediated DNA Damage Response Genes. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8916729. [PMID: 32851091 PMCID: PMC7439175 DOI: 10.1155/2020/8916729] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 04/04/2020] [Accepted: 07/07/2020] [Indexed: 12/03/2022]
Abstract
AURKA, a cell cycle-regulated kinase, is associated with malignant transformation and progression in many cancer types. We analyzed the expression change of AURKA in pan-cancer and its effect on the prognosis of cancer patients using the TCGA dataset. We revealed that AURKA was extensively elevated and predicted a poor prognosis in most of the detected cancer types, with an exception in colon cancer. AURKA was elevated in colon cancer, but the upregulation of AURKA indicated better outcomes of colon cancer patients. Then we revealed that undermethylation of the AURKA gene and several transcription factors contributed to the upregulation of AURKA in colon cancer. Moreover, we demonstrated that AURKA overexpression promoted the death of colon cancer cells induced by Oxaliplatin, whereas knockdown of AURKA significantly weakened the chemosensitivity of colon cancer cells to Oxaliplatin. Mechanistically, AURKA inhibited DNA damage response by suppressing the expression of various DNA damage repair genes in a TP53-dependent manner, which can partly explain that ARUKA is associated with a beneficial outcome of colon cancer. This study provided a possibility to use AURKA as a biomarker to predict the chemosensitivity of colon cancer to platinum in the clinic.
Collapse
|
42
|
A Synthetic Pan-Aurora Kinase Inhibitor, 5-Methoxy-2-(2-methoxynaphthalen-1-yl)-4H-chromen-4-one, Triggers Reactive Oxygen Species-Mediated Apoptosis in HCT116 Colon Cancer Cells. J CHEM-NY 2020. [DOI: 10.1155/2020/3025281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aurora kinases are Ser/Thr kinases that function as mitotic regulators. 5-Methoxy-2-(2-methoxynaphthalen-1-yl)-4H-chromen-4-one (DK1913) is a synthetic pan-Aurora kinase inhibitor. However, the mode of action of DK1913 concerning the induction of apoptosis is unclear. Here, we report that DK1913 triggered apoptosis, as revealed by flow cytometry and Annexin V staining. DK1913 enhanced the intracellular levels of reactive oxygen species (ROS) and stimulated the endoplasmic reticulum (ER) and genotoxic stress responses. We also found that DK1913 induced the loss of mitochondrial membrane potential, leading to the activation of caspase-9, caspase-7, and caspase-3. In addition, the antioxidant, butylated hydroxyanisole (BHA), abrogated DK1913-induced loss of mitochondrial membrane potential and activation of the caspase cascade. These findings demonstrate that pan-Aurora kinase inhibitor DK1913 triggers apoptosis through ROS-mediated ER and genotoxic stress responses.
Collapse
|
43
|
Jang J, Engleka KA, Liu F, Li L, Song G, Epstein JA, Li D. An Engineered Mouse to Identify Proliferating Cells and Their Derivatives. Front Cell Dev Biol 2020; 8:388. [PMID: 32523954 PMCID: PMC7261916 DOI: 10.3389/fcell.2020.00388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/29/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Cell proliferation is a fundamental event during development, disease, and regeneration. Effectively tracking and quantifying proliferating cells and their derivatives is critical for addressing many research questions. Cell cycle expression such as for Ki67, proliferating cell nuclear antigen (PCNA), or aurora kinase B (Aurkb), or measurement of 5-bromo-2'-deoxyuridine (BrdU) or 3H-thymidine incorporation have been widely used to assess and quantify cell proliferation. These are powerful tools for detecting actively proliferating cells, but they do not identify cell populations derived from proliferating progenitors over time. AIMS We developed a new mouse tool for lineage tracing of proliferating cells by targeting the Aurkb allele. RESULTS In quiescent cells or cells arrested at G1/S, little or no Aurkb mRNA is detectable. In cycling cells, Aurkb transcripts are detectable at G2 and become undetectable by telophase. These findings suggest that Aurkb transcription is restricted to proliferating cells and is tightly coupled to cell proliferation. Accordingly, we generated an Aurkb ER Cre/+ mouse by targeting a tamoxifen inducible Cre cassette into the start codon of Aurkb. We find that the Aurkb ER Cre/+ mouse faithfully labels proliferating cells in developing embryos and regenerative adult tissues such as intestine but does not label quiescent cells such as post-mitotic neurons. CONCLUSION The Aurkb ER Cre/+ mouse faithfully labels proliferating cells and their derivatives in developing embryos and regenerative adult tissues. This new mouse tool provides a novel genetic tracing capability for studying tissue proliferation and regeneration.
Collapse
Affiliation(s)
- Jihyun Jang
- Department of Surgery, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Kurt A. Engleka
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Feiyan Liu
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Li Li
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Guang Song
- Department of Surgery, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jonathan A. Epstein
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Deqiang Li
- Department of Surgery, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
44
|
Zhang X, Wang F, Wang Z, Yang X, Yu H, Si S, Lu J, Zhou Z, Lu Q, Wang Z, Yang H. ALKBH5 promotes the proliferation of renal cell carcinoma by regulating AURKB expression in an m 6A-dependent manner. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:646. [PMID: 32566583 PMCID: PMC7290639 DOI: 10.21037/atm-20-3079] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Background The modification and regulation of N6-methyladenosine (m6A) at mRNA level can affect the development and progression in various tumors. ALKBH5, as an m6A demethylase, plays different roles in tumors by regulating the m6A modification of mRNA. However, its role in renal cell carcinoma (RCC) remains unclear. Methods First, levels of ALKBH5 in RCC tissues and cell lines were verified by qRT-PCR and western blot. We analyzed the relationship between ALKBH5 and the clinicopathological characteristics of RCC patients and the influence of ALKBH5 on the prognosis of patients. Then we generated ALBKH5-overexpression, ALBKH5-knockdown stable RCC cell lines and their control cell lines. Through cell proliferation assay, colony formation assay, cell invasion and tumor migration assay, cell cycle assay and xenograft studies, we studied the ALKBH5 roles in RCC cell lines. AURKB was predicted to be its potential target based on TCGA database analysis and verified by western blot. The role of AURKB in RCC was verified by TCGA database and Kaplan-Meier analysis with TMA immunohistochemical analysis. Finally, the specific molecular mechanism of ALKBH5 targeting AURKB was explored by dual-luciferase reporter assay, RNA immunoprecipitation (RIP), m6A dot-blot assay, m6A RNA Immunoprecipitation (MeRIP) assay, and mRNA stability assay. Results We found that ALKBH5 was highly expressed in both RCC tumor tissues and cell lines. Clinicopathological analysis showed that high ALKBH5 expression was associated with larger tumor volume (P=0.017) and higher TNM staging (P=0.006), and worse prognosis (log rank: P=0.0199). The cellular functional assays showed that stably overexpression ALKBH5 could promote the cell proliferation, colony formation, cell migration and cell invasion of renal cell carcinoma cells in vitro and promote tumor growth in vivo. In contrast, ALKBH5 knocked down inhibited cell proliferation, colony formation, migration and invasion of renal cell carcinoma cells in vitro. Based on TCGA database analysis, AURKB was predicted highly expressed in RCC and a potential target of ALKBH5. Both database prediction and TMA immunohistochemical analysis supported that AURKB could affect the prognosis of RCC patients (P values of 5.5e-08 and 0.0004, respectively) and was regulated by ALKBH5 expression level. Subsequent mechanism experiments showed that ALKBH5 regulated the expression of AURKB by regulating the stability of AURKB mRNA in the m6A-dependent manner, and finally promoted cell proliferation. Furthermore, we found that hypoxia-induced HIF could up-regulate both expressions of AURKB and ALKBH5. Conclusions Our findings suggest that ALKBH5 may play a carcinogenic role in renal cell carcinoma by stabilizing AURKB mRNA in a m6A-dependent manner. These data suggest that ALKBH5 may play a key role in RCC and targeting the ALKBH5 signaling pathway may be a promising strategy for the treatment of RCC.
Collapse
Affiliation(s)
- Xiaolei Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Feng Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zijie Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiao Yang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hao Yu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shuhui Si
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jiancheng Lu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zijian Zhou
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qiang Lu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zengjun Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Haiwei Yang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
45
|
Chen C, Chen S, Luo M, Yan H, Pang L, Zhu C, Tan W, Zhao Q, Lai J, Li H. The role of the CDCA gene family in ovarian cancer. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:190. [PMID: 32309337 PMCID: PMC7154490 DOI: 10.21037/atm.2020.01.99] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Ovarian cancer is a frequently-occurring reproductive system malignancy in females, which leads to an annual of over 100 thousand deaths worldwide. Methods The electronic databases, including GEPIA, ONCOMINE, Metascape, and Kaplan-Meier Plotter, were used to examine both survival and transcriptional data regarding the cell division cycle associated (CDCA) gene family among ovarian cancer patients. Results All CDCA genes expression levels were up-regulated in ovarian cancer tissues relative to those in non-carcinoma ovarian counterparts. Besides, CDCA5/7 expression levels were related to the late tumor stage. In addition, the Kaplan-Meier Plotter database was employed to carry out survival analysis, which suggested that ovarian cancer patients with increased CDCA2/3/5/7 expression levels had poor overall survival (OS) (P<0.05). Moreover, ovarian cancer patients that had up-regulated mRNA expression levels of CDCA2/5/8 had markedly reduced progression-free survival (PFS) (P<0.05); and up-regulated CDCA4 expression showed remarkable association with reduced post-progression survival (PPS) (P<0.05). Additionally, the following processes were affected by CDCA genes alterations, including R-HAS-2500257: resolution of sister chromatid cohesion; GO:0051301: cell division; CORUM: 1118: Chromosomal passenger complex (CPC, including CDCA8, INCENP, AURKB and BIRC5); CORUM: 127: NDC80 kinetochore complex; M129: PID PLK1 pathway; and GO: 0007080: mitotic metaphase plate congression, all of which were subjected to marked regulation since the alterations affected CDCA genes. Conclusions Up-regulated CDCA gene expression in ovarian cancer tissues probably played a crucial part in the occurrence of ovarian cancer. The up-regulated CDCA2/3/5/7 expression levels were used as the potential prognostic markers to improve the poor ovarian cancer survival and prognostic accuracy. Moreover, CDCA genes probably exerted their functions in tumorigenesis through the PLK1 pathway.
Collapse
Affiliation(s)
- Chongxiang Chen
- Guangzhou Institute of Respiratory Diseases, State Key Laboratory of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.,Department of Intensive Care Unit, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Siliang Chen
- Department of Hematology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Ma Luo
- Department of Interventional Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Honghong Yan
- Department of Intensive Care Unit, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Lanlan Pang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Chaoyang Zhu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Weiyan Tan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Qingyu Zhao
- Department of Intensive Care Unit, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Jielan Lai
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Huan Li
- Department of Intensive Care Unit, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| |
Collapse
|
46
|
Bertolin G, Tramier M. Insights into the non-mitotic functions of Aurora kinase A: more than just cell division. Cell Mol Life Sci 2020; 77:1031-1047. [PMID: 31562563 PMCID: PMC11104877 DOI: 10.1007/s00018-019-03310-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/13/2019] [Accepted: 09/18/2019] [Indexed: 02/02/2023]
Abstract
AURKA is a serine/threonine kinase overexpressed in several cancers. Originally identified as a protein with multifaceted roles during mitosis, improvements in quantitative microscopy uncovered several non-mitotic roles as well. In physiological conditions, AURKA regulates cilia disassembly, neurite extension, cell motility, DNA replication and senescence programs. In cancer-like contexts, AURKA actively promotes DNA repair, it acts as a transcription factor, promotes cell migration and invasion, and it localises at mitochondria to regulate mitochondrial dynamics and ATP production. Here we review the non-mitotic roles of AURKA, and its partners outside of cell division. In addition, we give an insight into how structural data and quantitative fluorescence microscopy allowed to understand AURKA activation and its interaction with new substrates, highlighting future developments in fluorescence microscopy needed to better understand AURKA functions in vivo. Last, we will recapitulate the most significant AURKA inhibitors currently in clinical trials, and we will explore how the non-mitotic roles of the kinase may provide new insights to ameliorate current pharmacological strategies against AURKA overexpression.
Collapse
Affiliation(s)
- Giulia Bertolin
- Univ Rennes, CNRS, IGDR (Genetics and Development Institute of Rennes), UMR 6290, F-35000, Rennes, France.
| | - Marc Tramier
- Univ Rennes, CNRS, IGDR (Genetics and Development Institute of Rennes), UMR 6290, F-35000, Rennes, France.
| |
Collapse
|
47
|
Cui P, Abbasi B, Lin D, Rui R, Ju S. Aurora A inhibition disrupts chromosome condensation and spindle assembly during the first embryonic division in pigs. Reprod Domest Anim 2020; 55:584-593. [PMID: 32053743 DOI: 10.1111/rda.13655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/10/2020] [Indexed: 11/29/2022]
Abstract
As common overexpression of Aurora A in various tumours, much attention has focused on its function in inducing cancer, and its value in cancer therapeutics, considerably less is known regarding its role in the first cleavage division of mammalian embryos. Here, we highlight an indispensable role of Aurora A during the first mitotic division progression of pig embryos just after meiosis. The expression and spatiotemporal localization of Aurora A were initially assessed in pig embryos during the first mitotic division by Western blot analysis and indirect immunofluorescent staining. Then, the potential role of Aurora A was further evaluated using a highly selective Aurora A inhibitor, MLN8054, during this mitotic progression in pig embryos. Aurora A was found to express and exhibit a specific dynamic intracellular localization pattern during the first mitotic division in pig embryos. Aurora A was diffused in the cytoplasm at the prophase stage, and then exhibited a dynamic intracellular localization which was tightly associated with the chromosome and spindle dynamics throughout subsequent mitotic phases. Inhibition of Aurora A by MLN8054 treatment led to the failure of the first cleavage, with the majority of embryos being arrested in prophase of the mitotic division. Further subcellular structure examination showed that Aurora A inhibition not only led to the failure of spindle microtubule assembly, but also resulted in severe defects in chromosome condensation, accompanied by an obvious decrease in p-TACC3(S558) expression during the prophase of the first mitosis. Together, these results illustrated that Aurora A is crucial for both spindle assembly and chromosome condensation during the first mitotic division in pig embryos, and that the regulation of Aurora A may be associated with its effects on p-TACC3(S558) expression.
Collapse
Affiliation(s)
- Panpan Cui
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Benazir Abbasi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Defeng Lin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Rong Rui
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Shiqiang Ju
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
48
|
Wimbish RT, DeLuca JG. Hec1/Ndc80 Tail Domain Function at the Kinetochore-Microtubule Interface. Front Cell Dev Biol 2020; 8:43. [PMID: 32161753 PMCID: PMC7054225 DOI: 10.3389/fcell.2020.00043] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/17/2020] [Indexed: 12/28/2022] Open
Abstract
Successful mitotic cell division is critically dependent on the formation of correct attachments between chromosomes and spindle microtubules. Microtubule attachments are mediated by kinetochores, which are large proteinaceous structures assembled on centromeric chromatin of mitotic chromosomes. These attachments must be sufficiently stable to transduce force; however, the strength of these attachments are also tightly regulated to ensure timely, error-free progression through mitosis. The highly conserved, kinetochore-associated NDC80 complex is a core component of the kinetochore-microtubule attachment machinery in eukaryotic cells. A small, disordered region within the Hec1 subunit of the NDC80 complex – the N-terminal “tail” domain – has been actively investigated during the last decade due to its roles in generating and regulating kinetochore-microtubule attachments. In this review, we discuss the role of the NDC80 complex, and specifically the Hec1 tail domain, at the kinetochore-microtubule interface, and how recent studies provide a more unified view of Hec1 tail domain function.
Collapse
Affiliation(s)
- Robert T Wimbish
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Jennifer G DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
49
|
Analysis of the Expression of Cell Division Cycle-Associated Genes and Its Prognostic Significance in Human Lung Carcinoma: A Review of the Literature Databases. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6412593. [PMID: 32104702 PMCID: PMC7037569 DOI: 10.1155/2020/6412593] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 12/30/2019] [Indexed: 12/21/2022]
Abstract
Background Lung cancer (LC) has become the top cause responsible for cancer-related deaths. Cell division cycle-associated (CDCA) genes exert an important role in the life process. Dysregulation in the process of cell division may lead to malignancy. Methods Transcriptional data on CDCA gene family and patient survival data were examined for lung cancer (LC) patients from the GEPIA, Oncomine, cBioPortal, and Kaplan-Meier Plotter databases. Results CDCA1/2/3/4/5/7/8 expression levels were higher in lung adenocarcinoma tissues, and the CDCA1/2/3/4/5/6/7/8 expression levels were increased in squamous cell LC tissues compared with those in noncarcinoma lung tissues. The expression levels of CDCA1/2/3/4/5/8 showed correlation with tumor classification. The Kaplan-Meier Plotter database was employed to carry out survival analysis, indicating that increased CDCA1/2/3/4/5/6/7/8 expression levels were increased in squamous cell LC tissues compared with those in noncarcinoma lung tissues. The expression levels of P < 0.05). Only LC patients with increased CDCA3/4/5/8 expression were significantly related to lower post-progression survival (PPS) (P < 0.05). Only LC patients with increased CDCA gene family and patient survival data were examined for lung cancer (LC) patients from the GEPIA, Oncomine, cBioPortal, and Kaplan-Meier Plotter databases. CDCA8, INCENP, AURKB, and BIRC5); CORUM: 127: NDC80 kinetochore complex; M129: the PID PLK1 pathway; and GO: 0007080: mitotic metaphase plate congression, all of which were remarkably modulated since the alterations affected CDCA gene family and patient survival data were examined for lung cancer (LC) patients from the GEPIA, Oncomine, cBioPortal, and Kaplan-Meier Plotter databases. Conclusions Upregulated CDCA genes' expression levels in LC tissues probably play a crucial part in LC oncogenesis. The upregulated CDCA genes' expression levels are used as the potential prognostic markers to improve patient survival and the LC prognostic accuracy. CDCA genes probably exert their functions in tumorigenesis through the PLK1 pathway.CDCA gene family and patient survival data were examined for lung cancer (LC) patients from the GEPIA, Oncomine, cBioPortal, and Kaplan-Meier Plotter databases. CDCA gene family and patient survival data were examined for lung cancer (LC) patients from the GEPIA, Oncomine, cBioPortal, and Kaplan-Meier Plotter databases. CDCA gene family and patient survival data were examined for lung cancer (LC) patients from the GEPIA, Oncomine, cBioPortal, and Kaplan-Meier Plotter databases.
Collapse
|
50
|
Dong S, Liang J, Zhai W, Yu Z. Common and distinct features of potentially predictive biomarkers in small cell lung carcinoma and large cell neuroendocrine carcinoma of the lung by systematic and integrated analysis. Mol Genet Genomic Med 2020; 8:e1126. [PMID: 31981472 PMCID: PMC7057089 DOI: 10.1002/mgg3.1126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/10/2019] [Accepted: 01/02/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Large-cell neuroendocrine carcinoma of the lung (LCNEC) and small-cell lung carcinoma (SCLC) are neuroendocrine neoplasms. However, the underlying mechanisms of common and distinct genetic characteristics between LCNEC and SCLC are currently unclear. Herein, protein expression profiles and possible interactions with miRNAs were provided by integrated bioinformatics analysis, in order to explore core genes associated with tumorigenesis and prognosis in SCLC and LCNEC. METHODS GSE1037 gene expression profiles were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) in LCNEC and SCLC, as compared with normal lung tissues, were selected using the GEO2R online analyzer and Venn diagram software. Gene ontology (GO) analysis was performed using Database for Annotation, Visualization and Integrated Discovery. The biological pathway analysis was performed using the FunRich database. Subsequently, a protein-protein interaction (PPI) network of DEGs was generated using Search Tool for the Retrieval of Interacting Genes and displayed via Cytoscape software. The PPI network was analyzed by the Molecular Complex Detection app from Cytoscape, and 16 upregulated hub genes were selected. The Oncomine database was used to detect expression patterns of hub genes for validation. Furthermore, the biological pathways of these 16 hub genes were re-analyzed, and potential interactions between these genes and miRNAs were explored via FunRich. RESULTS A total of 384 DEGs were identified. A Venn diagram determined 88 common DEGs. The PPI network was constructed with 48 nodes and 221 protein pairs. Among them, 16 hub genes were extracted, 14 of which were upregulated in SCLC samples, as compared with normal lung specimens, and 10 were correlated with the cell cycle pathway. Furthermore, 57 target miRNAs for 8 hub genes were identified, among which 31 miRNAs were correlated with the progression of carcinoma, drug-resistance, radio-sensitivity, or autophagy in lung cancer. CONCLUSION This study provided effective biomarkers and novel therapeutic targets for diagnosis and prognosis of SCLC and LCNEC.
Collapse
Affiliation(s)
- Shenghua Dong
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jun Liang
- Department of Oncology, Peking University International Hospital, Beijing, China
| | - Wenxin Zhai
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhuang Yu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|