1
|
Wang C, Chang R, Li J, Li L. TRIM47 silencing inhibits the malignant biological behaviors of prostate cancer cells by regulating MDM2/p53 signaling. Cell Biochem Biophys 2024; 82:1567-1578. [PMID: 38802602 DOI: 10.1007/s12013-024-01308-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024]
Abstract
Prostate cancer (PCa) is a prevalent male malignancy globally. Tripartite motif 47 (TRIM47) has been reported to be associated with PCa. However, how TRIM47 acts on PCa is still incompletely understood. Here, we explored the biological roles of TRIM47 in PCa cells and investigated its potential regulatory mechanism. TRIM47 expression in PCa cells was detected by qRT-PCR and western blot. After TRIM47 silencing, the viability of PCa cells was measured using CCK-8 method. Flow cytometry was employed to estimate cell cycle. Cell apoptotic level was subjected to appraisement with TUNEL assay. Additionally, wound healing- and transwell assays were adopted for evaluation of migration and invasion of PCa cells. Moreover, the Biogrid database and HDOCK SERVER predicated that TRIM47 could interact with mouse double minute 2 (MDM2), which was detected using the Co-immunoprecipitation (co-IP) assay and glutathione S-transferase (GST) pull-down assay. The expression of proteins in MDM2/p53 signaling was detected by western blot analysis. Results indicated that TRIM47 expression was highly expressed in PCa cells. TRIM47 knockdown inhibited PCa proliferation and cell cycle whereas promoted cell apoptosis. Besides, TRIM47 knockdown significantly inhibited the migration and invasion of PCa cells. In addition, TRIM47 was proved to bind to MDM2 and regulated MDM2/p53 expression. Importantly, MDM2 overexpression counteracted the impacts of TRIM47 knockdown on cell viability, cell cycle, apoptosis, migration and invasion by regulating the MDM2/p53 pathway. Collectively, our results suggested that TRIM47 silencing inhibits the malignant biological behaviors of prostate cancer cells by regulating MDM2/p53 signaling, which may provide a novel therapeutic target for PCa treatment.
Collapse
Affiliation(s)
- Chengyong Wang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, 233000, Anhui Province, China.
| | - Rui Chang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, 233000, Anhui Province, China
| | - Jian Li
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, 233000, Anhui Province, China
| | - Liqiang Li
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, 233000, Anhui Province, China
| |
Collapse
|
2
|
Titova E, Shagieva G, Dugina V, Kopnin P. The Role of Aurora B Kinase in Normal and Cancer Cells. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:2054-2062. [PMID: 38462449 DOI: 10.1134/s0006297923120088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/22/2023] [Accepted: 10/28/2023] [Indexed: 03/12/2024]
Abstract
Aurora kinases are essential players in mammalian cell division. These kinases are involved in the regulation of spindle dynamics, microtubule-kinetochore interactions, and chromosome condensation and orientation during mitosis. At least three members of the Aurora family - Aurora kinases A, B, and C - have been identified in mammals. Aurora B is essential for maintaining genomic stability and normal cell division. Mutations and dysregulation of this kinase are implicated in tumor initiation and progression. In this review, we discuss the functions of Aurora B, the relationship between increased Aurora B activity and carcinogenesis, and the prospects for the use of Aurora B kinase inhibitors in antitumor therapy.
Collapse
Affiliation(s)
- Ekaterina Titova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | - Galina Shagieva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Vera Dugina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Pavel Kopnin
- Institute of Carcinogenesis, Blokhin National Medical Research Centre of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| |
Collapse
|
3
|
Mascarenhas J, Kremyanskaya M, Patriarca A, Palandri F, Devos T, Passamonti F, Rampal RK, Mead AJ, Hobbs G, Scandura JM, Talpaz M, Granacher N, Somervaille TCP, Hoffman R, Wondergem MJ, Salama ME, Colak G, Cui J, Kiladjian JJ, Vannucchi AM, Verstovsek S, Curto-García N, Harrison C, Gupta V. MANIFEST: Pelabresib in Combination With Ruxolitinib for Janus Kinase Inhibitor Treatment-Naïve Myelofibrosis. J Clin Oncol 2023; 41:4993-5004. [PMID: 36881782 PMCID: PMC10642902 DOI: 10.1200/jco.22.01972] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/04/2022] [Accepted: 12/23/2022] [Indexed: 03/09/2023] Open
Abstract
PURPOSE Standard therapy for myelofibrosis comprises Janus kinase inhibitors (JAKis), yet spleen response rates of 30%-40%, high discontinuation rates, and a lack of disease modification highlight an unmet need. Pelabresib (CPI-0610) is an investigational, selective oral bromodomain and extraterminal domain inhibitor (BETi). METHODS MANIFEST (ClinicalTrails.gov identifier: NCT02158858), a global, open-label, nonrandomized, multicohort, phase II study, includes a cohort of JAKi-naïve patients with myelofibrosis treated with pelabresib and ruxolitinib. The primary end point is a spleen volume reduction of ≥ 35% (SVR35) at 24 weeks. RESULTS Eighty-four patients received ≥ 1 dose of pelabresib and ruxolitinib. The median age was 68 (range, 37-85) years; 24% of patients were intermediate-1 risk, 61% were intermediate-2 risk, and 16% were high risk as per the Dynamic International Prognostic Scoring System; 66% (55 of 84) of patients had a hemoglobin level of < 10 g/dL at baseline. At 24 weeks, 68% (57 of 84) achieved SVR35, and 56% (46 of 82) achieved a total symptom score reduction of ≥ 50% (TSS50). Additional benefits at week 24 included 36% (29 of 84) of patients with improved hemoglobin levels (mean, 1.3 g/dL; median, 0.8 g/dL), 28% (16 of 57) with ≥ 1 grade improvement in fibrosis, and 29.5% (13 of 44) with > 25% reduction in JAK2V617F-mutant allele fraction, which was associated with SVR35 response (P = .018, Fisher's exact test). At 48 weeks, 60% (47 of 79) of patients had SVR35 response. Grade 3 or 4 toxicities seen in ≥ 10% patients were thrombocytopenia (12%) and anemia (35%), leading to treatment discontinuation in three patients. 95% (80 of 84) of the study participants continued combination therapy beyond 24 weeks. CONCLUSION The rational combination of the BETi pelabresib and ruxolitinib in JAKi-naïve patients with myelofibrosis was well tolerated and showed durable improvements in spleen and symptom burden, with associated biomarker findings of potential disease-modifying activity.
Collapse
Affiliation(s)
- John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Marina Kremyanskaya
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Andrea Patriarca
- Hematology Unit, Department of Translational Medicine, University of Eastern Piedmont and AOU Maggiore della Carità, Novara, Italy
| | - Francesca Palandri
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Institute of Hematology “Seràgnoli”, Bologna, Italy
| | - Timothy Devos
- Department of Hematology, University Hospitals Leuven and Department of Microbiology and Immunology, Laboratory of Molecular Immunology (Rega Institute), KU Leuven, Leuven, Belgium
| | | | | | - Adam J. Mead
- NIHR Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Gabriella Hobbs
- Division of Hematology/Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | - Moshe Talpaz
- University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | | | - Tim C. P. Somervaille
- The Christie NHS Foundation Trust & Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | | - Gozde Colak
- Constellation Pharmaceuticals Inc, a MorphoSys Company, Boston, MA
| | - Jike Cui
- Constellation Pharmaceuticals Inc, a MorphoSys Company, Boston, MA
| | | | | | - Srdan Verstovsek
- Leukemia Department, University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Claire Harrison
- Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Vikas Gupta
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Wang PC, Yang ZS, Gu XW. Effect of Aurora kinase B on polyploidy and decidualization in mouse uterus. Am J Reprod Immunol 2023; 90:e13793. [PMID: 37881124 DOI: 10.1111/aji.13793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/18/2023] [Accepted: 10/09/2023] [Indexed: 10/27/2023] Open
Abstract
RESEARCH QUESTION Decidualization is critical to the establishment of mouse normal pregnancy. The fibroblast-like stromal cells in the process form polyploid multinucleated cells. Aurora kinase B (Aurora B) has previously been shown to regulate polyploidy in various cells. However, whether Aurora B regulates the formation of decidual cell polyploidization and its regulatory mechanisms remain poorly understood. DESIGN Establish decidualization model of mouse primary endometrial stromal cells in vitro. Construct pseudopregnancy mouse models and delayed-activation mouse models. Detect Aurora B and polyploidization related genes in mouse uteri treated by Aurora B specific inhibitor Barasertib and CPT. RESULTS In this study, we found that Aurora B was strongly expressed in endometrial stromal cells after implantation. Additionally, Aurora B was remarkably up regulated in the stromal cells of oil-induced deciduomoa and in vitro decidualization. As an Aurora B specific inhibitor, Barasertib significantly inhibits the mRNA expression of Prl8a2, a marker of mouse decidualization. Furthermore, the protein levels of p-Plk1, Survivin and p-Cdk1 were inhibited by Barasertib. CPT-induced DNA damage suppressed Aurkb (encodes Aurora B) expression, thus resulting in polyploidization. CONCLUSION Our data shows that Aurora B is expressed in decidual stromal cells of implantation sites and plays a key role for mouse decidualization. The protein of Plk1, Survivn, and Cdk1 may participate in formation of decidual cell polyploidization during mouse decidualization.
Collapse
Affiliation(s)
- Peng-Chao Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Zhen-Shan Yang
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Xiao-Wei Gu
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| |
Collapse
|
5
|
Fan Y, Zhang F, Xiong L, Su M, Luo F, Li M, Li Q, Zhong T, Yuan M, Xu Y, Mu S, Yang H. Design, synthesis, and biological evaluation of 6-(imidazo[1,2-a] pyridin-6-yl) quinazolin-4(3H)-one derivatives as potent anticancer agents by dual targeting Aurora kinase and ROR1. Bioorg Chem 2023; 135:106484. [PMID: 36963371 DOI: 10.1016/j.bioorg.2023.106484] [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: 12/31/2022] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/26/2023]
Abstract
ROR1 and Aurora kinase were overexpressed in various cancers and essential for cell proliferation, survive and metastasis. Pharmaceutical inhibition of ROR1 and Aurora kinase abrogated the activation of downstream signaling and induced cancer cell apoptosis. Hence, ROR1 and Aurora kinase considered as attractive therapeutic targets for the development of anticancer drugs. In the present work, three series of novel 6-(imidazo[1,2-a] pyridin-6-yl)-quinazolin-4(3H)-one derivatives were designed and synthesized via bioisosterism and scaffold-hopping strategies guided by FLF-13, an Aurora kinase inhibitor we discovered earlier. Most of compounds in series 2 and series 3 showed submicromolar to nanomolar inhibitory activity against multiple cancer cell lines. More importantly, compounds 12d and 12f in series 3 showed nanomolar inhibitory activity against all test cancer cells. The most promising compound 12d exhibited potent inhibitory activity against Aurora A and Aurora B with IC50 values of 84.41 nM and 14.09 nM, respectively. Accordingly, compounds 12d induced G2/M phase cell cycle arrest at 24 h and polyploidy at 48 h. It's worth noting that 12d also displayed inhibitory activity against ROR1 and induce cell apoptosis. Furthermore, 12d could significantly inhibit the tumor growth in SH-SY5Y xenograft model with tumor growth inhibitory rate (IR) up to 46.31 % at 10 mg/kg and 52.66 % at 20 mg/kg. Overall, our data suggested that 12d might serve as a promising candidate for the development of therapeutic agents for cancers with aberrant expression of ROR1 and Aurora kinases by simultaneously targeting ROR1 and Aurora kinase.
Collapse
Affiliation(s)
- Yanhua Fan
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China.
| | - Feng Zhang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Liang Xiong
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Mingzhi Su
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Fang Luo
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Mei Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Qing Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Ting Zhong
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Meitao Yuan
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Yongnan Xu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuzhen Mu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China.
| | - Huarong Yang
- Chongqing Liangping District People's Hospital, Chongqing 405200, China.
| |
Collapse
|
6
|
Javed A, Özduman G, Altun S, Duran D, Yerli D, Özar T, Şimşek F, Korkmaz KS. Mitotic Kinase Inhibitors as Therapeutic Interventions for Prostate Cancer: Evidence from In Vitro Studies. Endocr Metab Immune Disord Drug Targets 2023; 23:1699-1712. [PMID: 36872354 DOI: 10.2174/1871530323666230303092243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 03/07/2023]
Abstract
Prostate cancer is one of the devastating diseases characterized by genetic changes leading to uncontrolled growth and metastasis of the cells of the prostate gland and affects men worldwide. Conventional hormonal and chemotherapeutic agents are effective in mitigating the disease if diagnosed at an early stage. All dividing eukaryotic cells require mitotic progression for the maintenance of genomic integrity in progeny populations. The protein kinases, upon activation and de-activation in an ordered fashion, lead to spatial and temporal regulation of the cell division process. The entry into mitosis along with the progression into sub-phases of mitosis is ensured due to the activity of mitotic kinases. These kinases include Polo-Like-Kinase 1 (PLK1), Aurora kinases, and Cyclin-Dependent- Kinase 1 (CDK1), among others. The mitotic kinases, among others, are usually overexpressed in many cancers and can be targeted using small molecule inhibitors to reduce the effects of these regulators on mechanisms, such as regulation of genomic integrity and mitotic fidelity. In this review, we attempted to discuss the appropriate functions of mitotic kinases revealed through cell culture studies and the impact of their respective inhibitors derived in pre-clinical studies. The review is designed to elucidate the growing field of small molecule inhibitors and their functional screening or mode of action at the cellular and molecular level in the context of Prostate Cancer. Therefore, studies performed specifically on cells of Prostatic-origin are narrated in this review, culminating in a comprehensive view of the specific field of mitotic kinases that can be targeted for therapy of Prostate cancer.
Collapse
Affiliation(s)
- Aadil Javed
- Department of Bioengineering, Faculty of Engineering, Cancer Biology Laboratory, Ege University, Bornova, Izmir, Turkey
| | - Gülseren Özduman
- Department of Bioengineering, Faculty of Engineering, Cancer Biology Laboratory, Ege University, Bornova, Izmir, Turkey
| | - Sevda Altun
- Department of Bioengineering, Faculty of Engineering, Cancer Biology Laboratory, Ege University, Bornova, Izmir, Turkey
| | - Doğan Duran
- Department of Bioengineering, Faculty of Engineering, Cancer Biology Laboratory, Ege University, Bornova, Izmir, Turkey
| | - Dilan Yerli
- Department of Bioengineering, Faculty of Engineering, Cancer Biology Laboratory, Ege University, Bornova, Izmir, Turkey
| | - Tilbe Özar
- Department of Bioengineering, Faculty of Engineering, Cancer Biology Laboratory, Ege University, Bornova, Izmir, Turkey
| | - Faruk Şimşek
- Department of Bioengineering, Faculty of Engineering, Cancer Biology Laboratory, Ege University, Bornova, Izmir, Turkey
| | - Kemal Sami Korkmaz
- Department of Bioengineering, Faculty of Engineering, Cancer Biology Laboratory, Ege University, Bornova, Izmir, Turkey
| |
Collapse
|
7
|
Jiao Y, Xu P, Luan S, Wang X, Gao Y, Zhao C, Fu P. Molecular imaging and treatment of PSMA-positive prostate cancer with 99mTc radiolabeled aptamer-siRNA chimeras. Nucl Med Biol 2021; 104-105:28-37. [PMID: 34847481 DOI: 10.1016/j.nucmedbio.2021.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/08/2021] [Accepted: 11/16/2021] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Prostate-specific membrane antigen (PSMA) is highly expressed in prostate cancer (PCa). The aptamer (Apt) A10-3.2 can be used as a specific ligand for the early diagnosis and targeted treatment of PCa. siRNA-Apt has been used to therapeutically target PSMA-positive PCa. We aimed to synthesize a new type of molecular probe to facilitate the integration of diagnosis and treatment for PSMA-positive PCa. METHODS Chimeras were obtained by covalent linking PSMA Apt-A10-3.2 and the MDM2 siRNA. SHNH, a bifunctional chelating agent, was used to couple 99mTc with chimeras to synthesize a new molecular probe. Labeling efficiency, radiochemical purity, and stability were confirmed using a γ-well counter and Whatman paper No.1. SPECT imaging and biodistribution studies were performed on BALB/c mice bearing 22Rv1 or PC-3 xenografts. Tumor inhibition and cytotoxicity of Chimeras were evaluated. LNCaP, 22RV1, and PC-3 PCa cell lines were used for in vitro and in vivo experiments. RESULTS [99mTc]Tc-chimeras showed high labeling efficiency (61.47% ± 2.85%, n = 3), radiochemical purity (>95%), and stability. Biodistribution studies and SPECT imaging with 99mTc-chimeras in mice bearing 22Rv1 xenografts demonstrated a high T/M ratio (4.63 ± 0.68, n = 3) and a high T/B ratio (3.61 ± 0.7, n = 3) at 2 h post-injection. 99mTc-chimeras showed rapid renal clearance. Compared with the PBS group, tumor growth in the chimera group was significantly inhibited (P < 0.01, n = 4), but there was no significant difference in body weight (p > 0.05, n = 4). H&E staining showed no obvious liver or kidney damage. CONCLUSIONS Our study proved that [99mTc]Tc-Aptamer-siRNA chimeras could be used to diagnose and treat PSMA-positive PCa in vivo.
Collapse
Affiliation(s)
- Yuying Jiao
- Department of Nuclear Medicine, 1st Hospital of Harbin Medical University, Harbin, 150000, China
| | - Peng Xu
- Department of Nuclear Medicine, 1st Hospital of Harbin Medical University, Harbin, 150000, China
| | - Sha Luan
- Department of Nuclear Medicine, 4th Hospital of Harbin Medical University, Harbin, 150000, China
| | - Xinyu Wang
- Department of Nuclear Medicine, 4th Hospital of Harbin Medical University, Harbin, 150000, China
| | - Yue Gao
- Department of Nuclear Medicine, 4th Hospital of Harbin Medical University, Harbin, 150000, China
| | - Changjiu Zhao
- Department of Nuclear Medicine, 1st Hospital of Harbin Medical University, Harbin, 150000, China.
| | - Peng Fu
- Department of Nuclear Medicine, 1st Hospital of Harbin Medical University, Harbin, 150000, China.
| |
Collapse
|
8
|
Effect of the HDAC Inhibitor on Histone Acetylation and Methyltransferases in A2780 Ovarian Cancer Cells. ACTA ACUST UNITED AC 2021; 57:medicina57050456. [PMID: 34066975 PMCID: PMC8151761 DOI: 10.3390/medicina57050456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 11/30/2022]
Abstract
Background andObjective: Epigenetic modifications are believed to play a significant role in the development of cancer progression, growth, differentiation, and cell death. One of the most popular histone deacetylases inhibitors (HDACIs), suberoylanilide hydroxamic acid (SAHA), also known as Vorinostat, can directly activate p21WAF1/CIP1 gene transcription through hyperacetylation of histones by a p53 independent mechanism. In the present investigation, we evaluated the correlation between histone modifications and DNA methyltransferase enzyme levels following SAHA treatments in A2780 ovarian cancer cells. Materials and Methods: Acetylation of histones and methyltransferases levels were analyzed using RT2 profiler PCR array, immunoblotting, and immunofluorescence methods in 2D and 3D cell culture systems. Results: The inhibition of histone deacetylases (HDAC) activities by SAHA can reduce DNA methyl transferases / histone methyl transferases (DNMTs/HMTs) levels through induction of hyperacetylation of histones. Immunofluorescence analysis of cells growing in monolayers and spheroids revealed significant up-regulation of histone acetylation preceding the above-described changes. Conclusions: Our results depict an interesting interplay between histone hyperacetylation and a decrease in methyltransferase levels in ovarian cancer cells, which may have a positive impact on the overall outcomes of cancer treatment.
Collapse
|
9
|
Aurora Kinase B Inhibition: A Potential Therapeutic Strategy for Cancer. Molecules 2021; 26:molecules26071981. [PMID: 33915740 PMCID: PMC8037052 DOI: 10.3390/molecules26071981] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/23/2022] Open
Abstract
Aurora kinase B (AURKB) is a mitotic serine/threonine protein kinase that belongs to the aurora kinase family along with aurora kinase A (AURKA) and aurora kinase C (AURKC). AURKB is a member of the chromosomal passenger protein complex and plays a role in cell cycle progression. Deregulation of AURKB is observed in several tumors and its overexpression is frequently linked to tumor cell invasion, metastasis and drug resistance. AURKB has emerged as an attractive drug target leading to the development of small molecule inhibitors. This review summarizes recent findings pertaining to the role of AURKB in tumor development, therapy related drug resistance, and its inhibition as a potential therapeutic strategy for cancer. We discuss AURKB inhibitors that are in preclinical and clinical development and combination studies of AURKB inhibition with other therapeutic strategies.
Collapse
|
10
|
Borah NA, Sradhanjali S, Barik MR, Jha A, Tripathy D, Kaliki S, Rath S, Raghav SK, Patnaik S, Mittal R, Reddy MM. Aurora Kinase B Expression, Its Regulation and Therapeutic Targeting in Human Retinoblastoma. Invest Ophthalmol Vis Sci 2021; 62:16. [PMID: 33704359 PMCID: PMC7960835 DOI: 10.1167/iovs.62.3.16] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/05/2021] [Indexed: 12/22/2022] Open
Abstract
Purpose Aurora kinase B (AURKB) plays a pivotal role in the regulation of mitosis and is gaining prominence as a therapeutic target in cancers; however, the role of AURKB in retinoblastoma (RB) has not been studied. The purpose of this study was to determine if AURKB plays a role in RB, how its expression is regulated, and whether it could be specifically targeted. Methods The protein expression of AURKB was determined using immunohistochemistry in human RB patient specimens and immunoblotting in cell lines. Pharmacological inhibition and shRNA-mediated knockdown were used to understand the role of AURKB in cell viability, apoptosis, and cell cycle distribution. Cell viability in response to AURKB inhibition was also assessed in enucleated RB specimens. Immunoblotting was employed to determine the protein levels of phospho-histone H3, p53, p21, and MYCN. Chromatin immunoprecipitation-qPCR was performed to verify the binding of MYCN on the promoter region of AURKB. Results The expression of AURKB was found to be markedly elevated in human RB tissues, and the overexpression significantly correlated with optic nerve and anterior chamber invasion. Targeting AURKB with small-molecule inhibitors and shRNAs resulted in reduced cell survival and increased apoptosis and cell cycle arrest at the G2/M phase. More importantly, primary RB specimens showed decreased cell viability in response to pharmacological AURKB inhibition. Additional studies have demonstrated that the MYCN oncogene regulates the expression of AURKB in RB. Conclusions AURKB is overexpressed in RB, and targeting it could serve as a novel therapeutic strategy to restrict tumor cell growth.
Collapse
Affiliation(s)
- Naheed Arfin Borah
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Bhubaneswar, India
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Swatishree Sradhanjali
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Bhubaneswar, India
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Manas Ranjan Barik
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Bhubaneswar, India
| | - Atimukta Jha
- Immuno-Genomics and Systems Biology Laboratory, Institute of Life Sciences, Bhubaneswar, India
- Manipal Academy of Higher Education, Manipal, India
| | - Devjyoti Tripathy
- Ophthalmic Plastics, Orbit and Ocular Oncology Service, LV Prasad Eye Institute, Bhubaneswar, India
| | - Swathi Kaliki
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Hyderabad, India
| | - Suryasnata Rath
- Ophthalmic Plastics, Orbit and Ocular Oncology Service, LV Prasad Eye Institute, Bhubaneswar, India
| | - Sunil K. Raghav
- Immuno-Genomics and Systems Biology Laboratory, Institute of Life Sciences, Bhubaneswar, India
| | | | - Ruchi Mittal
- Kanupriya Dalmia Ophthalmic Pathology Laboratory, LV Prasad Eye Institute, Bhubaneswar, India
- Department of Pathology, Kalinga Institute of Medical Sciences, Bhubaneswar, India
| | - Mamatha M. Reddy
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Bhubaneswar, India
- School of Biotechnology, KIIT University, Bhubaneswar, India
| |
Collapse
|
11
|
Li M, Liu H, Zhao Q, Han S, Zhou L, Liu W, Li W, Gao F. Targeting Aurora B kinase with Tanshinone IIA suppresses tumor growth and overcomes radioresistance. Cell Death Dis 2021; 12:152. [PMID: 33542222 PMCID: PMC7862432 DOI: 10.1038/s41419-021-03434-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/13/2022]
Abstract
Aurora B kinase is aberrantly overexpressed in various tumors and shown to be a promising target for anti-cancer therapy. In human oral squamous cell carcinoma (OSCC), the high protein level of Aurora B is required for maintaining of malignant phenotypes, including in vitro cell growth, colony formation, and in vivo tumor development. By molecular modeling screening of 74 commercially available natural products, we identified that Tanshinone IIA (Tan IIA), as a potential Aurora B kinase inhibitor. The in silico docking study indicates that Tan IIA docks into the ATP-binding pocket of Aurora B, which is further confirmed by in vitro kinase assay, ex vivo pull-down, and ATP competitive binding assay. Tan IIA exhibited a significant anti-tumor effect on OSCC cells both in vitro and in vivo, including reduction of Aurora B and histone H3 phosphorylation, induction of G2/M cell cycle arrest, increase the population of polyploid cells, and promotion of apoptosis. The in vivo mouse model revealed that Tan IIA delayed tumor growth of OSCC cells. Tan IIA alone or in combination with radiation overcame radioresistance in OSCC xenograft tumors. Taken together, our data indicate that Tan IIA is an Aurora B kinase inhibitor with therapeutic potentials for cancer treatment.
Collapse
Affiliation(s)
- Ming Li
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, People's Republic of China
- Changsha Stomatological Hospital, Changsha, 410004, Hunan, People's Republic of China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, People's Republic of China
- Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, 410000, Hunan, People's Republic of China
| | - Haidan Liu
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, People's Republic of China
- Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Qin Zhao
- Changsha Stomatological Hospital, Changsha, 410004, Hunan, People's Republic of China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, People's Republic of China
| | - Shuangze Han
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, People's Republic of China
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Li Zhou
- Department of Pathology, Xiangya Hospital, Changsha, 410008, Hunan, People's Republic of China
| | - Wenbin Liu
- Department of Pathology, Hunan Cancer Hospital, Changsha, 410013, Hunan, People's Republic of China
| | - Wei Li
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, People's Republic of China.
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China.
| | - Feng Gao
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, People's Republic of China.
- Department of Ultrasonography, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China.
| |
Collapse
|
12
|
Natarajan U, Venkatesan T, Dhandayuthapani S, Dondapatti P, Rathinavelu A. Differential mechanisms involved in RG-7388 and Nutlin-3 induced cell death in SJSA-1 osteosarcoma cells. Cell Signal 2020; 75:109742. [PMID: 32827690 DOI: 10.1016/j.cellsig.2020.109742] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 11/17/2022]
Abstract
Targeted therapy is becoming the mainstay of cancer treatment due to reduced side effects and enhanced tumor attack. In the last few decades, Murine Double Minute 2 (MDM2) protein has become one of the targets for developing cancer therapies. Blocking MDM2-p53 interaction has long been considered to offer a broad range of advantages during cancer treatment. In this study, we are reporting the differential mechanism of cell death induced by the two small-molecule inhibitors, named RG-7388 and Nutlin-3, that are specific for MDM2 in SJSA-1 Osteosarcoma cells (OS). Mechanistically, RG-7388 was able to enhance the phosphorylation of Mcl-1, which appears to significantly enhance its degradation, thereby relieving the pro-apoptotic protein Bak to execute the apoptosis mechanism. It was noted that the untreated SJSA-1 cells showed an accumulation of Mcl-1 levels, which was decreased following RG-7388 and to a lesser extent by Nutlin-3 and GSK-3β (glycogen synthase kinase 3β) inhibitor treatments. Additionally, we noted that CHIR-99021 (GSK-3β inhibitor) blocked the cytotoxicity exerted by RG-7388 on SJSA-1 cells by decreasing Bak levels. Since Bak is an important pro-apoptotic protein, we hypothesized that phosphorylation of Mcl-1 by GSK-3β could negatively impact the Mcl-1/Bak dimerization and relieve Bak to trigger the loss of mitochondrial membrane potential and thereby initiates apoptosis. We also observed that inhibition of GSK-3β mediated reduction in Bak levels had a protective effect on the mitochondrial membrane integrity, and thus, caused a significant inhibition of the caspase-3 activity and PARP cleavage. Nutlin-3, on the other hand, appears to increase the levels of Bax, leading to the inactivation of Bcl-2, consequently loss of mitochondrial membrane potential and release of Cytochrome c (Cyt c) and elevation of Apaf-1 triggering apoptosis. Thus, to the best of our knowledge, this is the first study that delineates the differences in the molecular mechanism involving two MDM2 inhibitors triggering apoptosis through parallel pathways in SJSA-1 cells. This study further opens new avenues for the use of RG-7388 in treating osteosarcomas that often becomes resistant to chemotherapy due to Bcl-2 overexpression.
Collapse
Affiliation(s)
- Umamaheswari Natarajan
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Ft. Lauderdale, FL 33314, USA
| | - Thiagarajan Venkatesan
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Ft. Lauderdale, FL 33314, USA
| | - Sivanesan Dhandayuthapani
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Ft. Lauderdale, FL 33314, USA
| | - Priya Dondapatti
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Ft. Lauderdale, FL 33314, USA
| | - Appu Rathinavelu
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Ft. Lauderdale, FL 33314, USA; College of Pharmacy, Health Professions Division, Nova Southeastern University, Ft. Lauderdale, FL 33314, USA.
| |
Collapse
|
13
|
Aschacher T, Wolf B, Aschacher O, Enzmann F, Laszlo V, Messner B, Türkcan A, Weis S, Spiegl-Kreinecker S, Holzmann K, Laufer G, Ehrlich M, Bergmann M. Long interspersed element-1 ribonucleoprotein particles protect telomeric ends in alternative lengthening of telomeres dependent cells. Neoplasia 2019; 22:61-75. [PMID: 31846834 PMCID: PMC6920197 DOI: 10.1016/j.neo.2019.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 12/21/2022] Open
Abstract
Malignant cells ensure telomere maintenance by the alternative lengthening of telomeres (ALT) in the absence of telomerase activity (TA). The retrotransposons "long interspersed nuclear element-1" (LINE-1, L1) are expressed in malignant cells and are primarily known to contribute to complex karyotypes. Here we demonstrate that LINE-1 ribonucleoprotein particles (L1-RNPs) expression is significantly higher in ALT+- versus in TA+-human glioma. Analyzing a role of L1-RNP in ALT, we show that L1-RNPs bind to telomeric repeat containing RNA (TERRA), which is critical for telomere stabilization and which is overexpressed in ALT+ cells. In turn, L1-RNP knockdown (KD) abrogated the nuclear retention of TERRA, resulted in increased telomeric DNA damage, decreased cell growth and reduced expression of ALT characteristics such as c-circles and PML-bodies. L1-RNP KD also decreased the expression of Shelterin- and the ALT-regulating protein Topoisomerase IIIα (TopoIIIα) indicating a more general role of L1-RNPs in supporting telomeric integrity in ALT. Our findings suggest an impact of L1-RNP on telomere stability in ALT+ dependent tumor cells. As L1-RNPs are rarely expressed in normal adult human tissue those elements might serve as a novel target for tumor ablative therapy.
Collapse
Affiliation(s)
- Thomas Aschacher
- Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Brigitte Wolf
- Surgical Research Laboratories, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Olivia Aschacher
- Department of Plastic and Reconstructive Surgery, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Florian Enzmann
- Department of Vascular and Endovascular Surgery, Paracelsus Medical University Salzburg, Muellner Hauptstraße 48, 5020 Salzburg, Austria
| | - Viktoria Laszlo
- Surgical Research Laboratories, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Barbara Messner
- Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Adrian Türkcan
- Surgical Research Laboratories, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Serge Weis
- Division of Neuropathology, Neuromed Campus, Kepler University Hospital, 4020 Linz, Austria
| | - Sabine Spiegl-Kreinecker
- University Clinic for Neurosurgery, Neuromed Campus, Kepler University Hospital, Johannes Kepler University, Linz, Austria
| | - Klaus Holzmann
- Department of Cancer Research, Borschkegasse 8a, 1090 Vienna, Austria; Comprehensive Cancer Centre, Medical University of Vienna, Austria
| | - Günther Laufer
- Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Marek Ehrlich
- Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Michael Bergmann
- Surgical Research Laboratories, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Comprehensive Cancer Centre, Medical University of Vienna, Austria.
| |
Collapse
|