1
|
Muto S, Homma MK, Kiko Y, Ozaki Y, Watanabe M, Okabe N, Hamada K, Hashimoto Y, Suzuki H. Nucleolar casein kinase 2 alpha as a prognostic factor in patients with surgically resected early‑stage lung adenocarcinoma. Oncol Rep 2025; 53:4. [PMID: 39513582 DOI: 10.3892/or.2024.8837] [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: 05/21/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024] Open
Abstract
Lung cancer remains a leading cause of global cancer‑related deaths, therefore the identification of prognostic factors for lung cancer is critical. Casein kinase 2 alpha (CK2α) is one of the driver kinases in various cancers, and it was previously demonstrated that CK2α localization was associated with a poor prognosis in invasive breast cancer. In the present study, the importance of CK2α in the nucleolus was explored as a potential prognostic marker for surgically resected early‑stage lung adenocarcinoma. The present study included 118 patients who underwent pulmonary lobectomy between 2014 and 2018 in Fukushima Medical University Hospital (Fukushima, Japan), and in whom CK2α localization in tumor samples was assessed by immunohistochemistry. Patient and tumor characteristics, including pathological stage, histological type and histological grade, were analyzed. Recurrence‑free survival (RFS) and overall survival were evaluated in relation to nucleolar CK2α staining. CK2α staining in the nucleoli was observed in 50.8% of lung adenocarcinoma tumors. Positive nucleolar CK2α staining was independent of pathological stage, histological type and histological grade. Patients with positive nucleolar CK2α staining exhibited significantly worse RFS compared with patients with negative staining. Multivariate analysis identified nucleolar CK2α staining and lymph node metastasis as independent poor prognostic factors. The results of the present study suggested that nucleolar CK2α staining is a novel and independent prognostic factor in surgically resected early‑stage lung adenocarcinoma. These findings indicated the potential of nucleolar CK2α as a predictive biomarker for future recurrence, and a guide to treatment decisions. Further research is required, particularly in understanding the molecular mechanisms linking nucleolar CK2α to recurrence.
Collapse
Affiliation(s)
- Satoshi Muto
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Miwako Kato Homma
- Department of Biomolecular Sciences, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Yuichiro Kiko
- Department of Diagnostic Pathology, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Yuki Ozaki
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Masayuki Watanabe
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Naoyuki Okabe
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Kazuyuki Hamada
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Yuko Hashimoto
- Department of Diagnostic Pathology, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Hiroyuki Suzuki
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| |
Collapse
|
2
|
Greco FA, Krämer A, Wahl L, Elson L, Ehret TAL, Gerninghaus J, Möckel J, Müller S, Hanke T, Knapp S. Synthesis and evaluation of chemical linchpins for highly selective CK2α targeting. Eur J Med Chem 2024; 276:116672. [PMID: 39067440 DOI: 10.1016/j.ejmech.2024.116672] [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: 05/27/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/30/2024]
Abstract
Casein kinase-2 (CK2) are serine/threonine kinases with dual co-factor (ATP and GTP) specificity, that are involved in the regulation of a wide variety of cellular functions. Small molecules targeting CK2 have been described in the literature targeting different binding pockets of the kinase with a focus on type I inhibitors such as the recently published chemical probe SGC-CK2-1. In this study, we investigated whether known allosteric inhibitors binding to a pocket adjacent to helix αD could be combined with ATP mimetic moieties defining a novel class of ATP competitive compounds with a unique binding mode. Linking both binding sites requires a chemical linking moiety that would introduce a 90-degree angle between the ATP mimetic ring system and the αD targeting moiety, which was realized using a sulfonamide. The synthesized inhibitors were highly selective for CK2 with binding constants in the nM range and low micromolar activity. While these inhibitors need to be further improved, the present work provides a structure-based design strategy for highly selective CK2 inhibitors.
Collapse
Affiliation(s)
- Francesco A Greco
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Andreas Krämer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), DKTK Site Frankfurt-Mainz, 69120 Heidelberg, Germany
| | - Laurenz Wahl
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Lewis Elson
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Theresa A L Ehret
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Joshua Gerninghaus
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Janina Möckel
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Susanne Müller
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Thomas Hanke
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany.
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), DKTK Site Frankfurt-Mainz, 69120 Heidelberg, Germany.
| |
Collapse
|
3
|
Liu ZD, Shi YH, Xu QC, Zhao GY, Zhu YQ, Li FX, Ma MJ, Ye JY, Huang XT, Wang XY, Xu X, Wang JQ, Zhao W, Yin XY. CSNK2A1 confers gemcitabine resistance to pancreatic ductal adenocarcinoma via inducing autophagy. Cancer Lett 2024; 585:216640. [PMID: 38290659 DOI: 10.1016/j.canlet.2024.216640] [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: 08/28/2023] [Revised: 12/07/2023] [Accepted: 01/11/2024] [Indexed: 02/01/2024]
Abstract
Gemcitabine, a pivotal chemotherapeutic agent for pancreatic ductal adenocarcinoma (PDAC), frequently encounters drug resistance, posing a significant clinical challenge with implications for PDAC patient prognosis. In this study, employing an integrated approach involving bioinformatic analyses from multiple databases, we unveil CSNK2A1 as a key regulatory factor. The patient-derived xenograft (PDX) model further substantiates the critical role of CSNK2A1 in gemcitabine resistance within the context of PDAC. Additionally, targeted silencing of CSNK2A1 expression significantly enhances sensitivity of PDAC cells to gemcitabine treatment. Mechanistically, CSNK2A1's transcriptional regulation is mediated by H3K27 acetylation in PDAC. Moreover, we identify CSNK2A1 as a pivotal activator of autophagy, and enhanced autophagy drives gemcitabine resistance. Silmitasertib, an established CSNK2A1 inhibitor, can effectively inhibit autophagy. Notably, the combinatorial treatment of Silmitasertib with gemcitabine demonstrates remarkable efficacy in treating PDAC. In summary, our study reveals CSNK2A1 as a potent predictive factor for gemcitabine resistance in PDAC. Moreover, targeted CSNK2A1 inhibition by Silmitasertib represents a promising therapeutic strategy to restore gemcitabine sensitivity in PDAC, offering hope for improved clinical outcomes.
Collapse
Affiliation(s)
- Zhi-De Liu
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Yin-Hao Shi
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Qiong-Cong Xu
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Guang-Yin Zhao
- Department of Animal Experiment Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Ying-Qin Zhu
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Fu-Xi Li
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, China
| | - Ming-Jian Ma
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jing-Yuan Ye
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xi-Tai Huang
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xi-Yu Wang
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xiang Xu
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jie-Qin Wang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Wei Zhao
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, China.
| | - Xiao-Yu Yin
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
4
|
Homma MK, Nakato R, Niida A, Bando M, Fujiki K, Yokota N, Yamamoto S, Shibata T, Takagi M, Yamaki J, Kozuka-Hata H, Oyama M, Shirahige K, Homma Y. Cell cycle-dependent gene networks for cell proliferation activated by nuclear CK2α complexes. Life Sci Alliance 2024; 7:e202302077. [PMID: 37907238 PMCID: PMC10618106 DOI: 10.26508/lsa.202302077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/02/2023] Open
Abstract
Nuclear expression of protein kinase CK2α is reportedly elevated in human carcinomas, but mechanisms underlying its variable localization in cells are poorly understood. This study demonstrates a functional connection between nuclear CK2 and gene expression in relation to cell proliferation. Growth stimulation of quiescent human normal fibroblasts and phospho-proteomic analysis identified a pool of CK2α that is highly phosphorylated at serine 7. Phosphorylated CK2α translocates into the nucleus, and this phosphorylation appears essential for nuclear localization and catalytic activity. Protein signatures associated with nuclear CK2 complexes reveal enrichment of apparently unique transcription factors and chromatin remodelers during progression through the G1 phase of the cell cycle. Chromatin immunoprecipitation-sequencing profiling demonstrated recruitment of CK2α to active gene loci, more abundantly in late G1 phase than in early G1, notably at transcriptional start sites of core histone genes, growth stimulus-associated genes, and ribosomal RNAs. Our findings reveal that nuclear CK2α complexes may be essential to facilitate progression of the cell cycle, by activating histone genes and triggering ribosomal biogenesis, specified in association with nuclear and nucleolar transcriptional regulators.
Collapse
Affiliation(s)
- Miwako Kato Homma
- Department of Biomolecular Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Ryuichiro Nakato
- Laboratory of Computational Genomics, Institute for Quantitative Biosciences, University of Tokyo, Bunkyo, Japan
| | - Atsushi Niida
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Minato, Japan
| | - Masashige Bando
- Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo, Japan
| | - Katsunori Fujiki
- Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo, Japan
| | - Naoko Yokota
- Laboratory of Computational Genomics, Institute for Quantitative Biosciences, University of Tokyo, Bunkyo, Japan
| | - So Yamamoto
- Department of Biomolecular Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | | | - Motoki Takagi
- Translational Research Center, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Junko Yamaki
- Department of Biomolecular Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hiroko Kozuka-Hata
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Minato, Japan
| | - Masaaki Oyama
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Minato, Japan
| | - Katsuhiko Shirahige
- Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo, Japan
- Department of Biosciences and Nutrition, Karolinska Institutet, Biomedicum, Stockholm, Sweden
- Department of Cell and Molecular Biology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - Yoshimi Homma
- Department of Biomolecular Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
5
|
Zhai F, Wang J, Luo X, Ye M, Jin X. Roles of NOLC1 in cancers and viral infection. J Cancer Res Clin Oncol 2023; 149:10593-10608. [PMID: 37296317 DOI: 10.1007/s00432-023-04934-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND The nucleolus is considered the center of metabolic control and an important organelle for the biogenesis of ribosomal RNA (rRNA). Nucleolar and coiled-body phosphoprotein 1(NOLC1), which was originally identified as a nuclear localization signal-binding protein is a nucleolar protein responsible for nucleolus construction and rRNA synthesis, as well as chaperone shuttling between the nucleolus and cytoplasm. NOLC1 plays an important role in a variety of cellular life activities, including ribosome biosynthesis, DNA replication, transcription regulation, RNA processing, cell cycle regulation, apoptosis, and cell regeneration. PURPOSE In this review, we introduce the structure and function of NOLC1. Then we elaborate its upstream post-translational modification and downstream regulation. Meanwhile, we describe its role in cancer development and viral infection which provide a direction for future clinical applications. METHODS The relevant literatures from PubMed have been reviewed for this article. CONCLUSION NOLC1 plays an important role in the progression of multiple cancers and viral infection. In-depth study of NOLC1 provides a new perspective for accurate diagnosis of patients and selection of therapeutic targets.
Collapse
Affiliation(s)
- Fengguang Zhai
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
- The Affiliated First Hospital, Ningbo University, Ningbo, 315020, China
| | - Jie Wang
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
- The Affiliated First Hospital, Ningbo University, Ningbo, 315020, China
| | - Xia Luo
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
- The Affiliated First Hospital, Ningbo University, Ningbo, 315020, China.
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
- The Affiliated First Hospital, Ningbo University, Ningbo, 315020, China.
| |
Collapse
|
6
|
Protein Kinase CK2 and Epstein-Barr Virus. Biomedicines 2023; 11:biomedicines11020358. [PMID: 36830895 PMCID: PMC9953236 DOI: 10.3390/biomedicines11020358] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Protein kinase CK2 is a pleiotropic protein kinase, which phosphorylates a number of cellular and viral proteins. Thereby, this kinase is implicated in the regulation of cellular signaling, controlling of cell proliferation, apoptosis, angiogenesis, immune response, migration and invasion. In general, viruses use host signaling mechanisms for the replication of their genome as well as for cell transformation leading to cancer. Therefore, it is not surprising that CK2 also plays a role in controlling viral infection and the generation of cancer cells. Epstein-Barr virus (EBV) lytically infects epithelial cells of the oropharynx and B cells. These latently infected B cells subsequently become resting memory B cells when passing the germinal center. Importantly, EBV is responsible for the generation of tumors such as Burkitt's lymphoma. EBV was one of the first human viruses, which was connected to CK2 in the early nineties of the last century. The present review shows that protein kinase CK2 phosphorylates EBV encoded proteins as well as cellular proteins, which are implicated in the lytic and persistent infection and in EBV-induced neoplastic transformation. EBV-encoded and CK2-phosphorylated proteins together with CK2-phosphorylated cellular signaling proteins have the potential to provide efficient virus replication and cell transformation. Since there are powerful inhibitors known for CK2 kinase activity, CK2 might become an attractive target for the inhibition of EBV replication and cell transformation.
Collapse
|
7
|
Trembley JH, Kren BT, Afzal M, Scaria GA, Klein MA, Ahmed K. Protein kinase CK2 – diverse roles in cancer cell biology and therapeutic promise. Mol Cell Biochem 2022; 478:899-926. [PMID: 36114992 PMCID: PMC9483426 DOI: 10.1007/s11010-022-04558-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022]
Abstract
The association of protein kinase CK2 (formerly casein kinase II or 2) with cell growth and proliferation in cells was apparent at early stages of its investigation. A cancer-specific role for CK2 remained unclear until it was determined that CK2 was also a potent suppressor of cell death (apoptosis); the latter characteristic differentiated its function in normal versus malignant cells because dysregulation of both cell growth and cell death is a universal feature of cancer cells. Over time, it became evident that CK2 exerts its influence on a diverse range of cell functions in normal as well as in transformed cells. As such, CK2 and its substrates are localized in various compartments of the cell. The dysregulation of CK2 is documented in a wide range of malignancies; notably, by increased CK2 protein and activity levels with relatively moderate change in its RNA abundance. High levels of CK2 are associated with poor prognosis in multiple cancer types, and CK2 is a target for active research and testing for cancer therapy. Aspects of CK2 cellular roles and targeting in cancer are discussed in the present review, with focus on nuclear and mitochondrial functions and prostate, breast and head and neck malignancies.
Collapse
Affiliation(s)
- Janeen H Trembley
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA.
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Betsy T Kren
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA
| | - Muhammad Afzal
- Department of Biochemistry, Riphah International University, Islamabad, Pakistan
| | - George A Scaria
- Hematology/Oncology Section, Primary Care Service Line, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA
| | - Mark A Klein
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
- Hematology/Oncology Section, Primary Care Service Line, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Khalil Ahmed
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA.
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
- Department of Urology, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
8
|
CSNK2 in cancer: pathophysiology and translational applications. Br J Cancer 2022; 126:994-1003. [PMID: 34773100 PMCID: PMC8980014 DOI: 10.1038/s41416-021-01616-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/29/2021] [Accepted: 10/22/2021] [Indexed: 12/13/2022] Open
Abstract
Protein kinase CSNK2 (CK2) is a pleiotropic serine/threonine kinase frequently dysregulated in solid and hematologic malignancies. To consolidate a wide range of biological and clinically oriented data from this unique kinase in cancer, this systematic review summarises existing knowledge from in vitro, in vivo and pre-clinical studies on CSNK2 across 24 different human cancer types. CSNK2 mRNA transcripts, protein levels and activity were found to be routinely upregulated in cancer, and commonly identified phosphotargets included AKT, STAT3, RELA, PTEN and TP53. Phenotypically, it frequently influenced evasion of apoptosis, enhancement of proliferation, cell invasion/metastasis and cell cycle control. Clinically, it held prognostic significance across 14 different cancers, and its inhibition in xenograft experiments resulted in a positive treatment response in 12. In conjunction with commentary on preliminary studies of CSNK2 inhibitors in humans, this review harmonises an extensive body of CSNK2 data in cancer and reinforces its emergence as an attractive target for cancer therapy. Continuing to investigate CSNK2 will be crucial to advancing our understanding of CSNK2 biology, and offers the promise of important new discoveries scientifically and clinically.
Collapse
|
9
|
Aichinger G. Natural Dibenzo-α-Pyrones: Friends or Foes? Int J Mol Sci 2021; 22:13063. [PMID: 34884865 PMCID: PMC8657677 DOI: 10.3390/ijms222313063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/17/2022] Open
Abstract
Natural dibenzo-α-pyrones (DAPs) can be viewed from two opposite angles. From one angle, the gastrointestinal metabolites urolithins are regarded as beneficial, while from the other, the emerging mycotoxin alternariol and related fungal metabolites are evaluated critically with regards to potential hazardous effects. Thus, the important question is: can the structural characteristics of DAP subgroups be held responsible for distinct bioactivity patterns? If not, certain toxicological and/or pharmacological aspects of natural DAPs might yet await elucidation. Thus, this review focuses on comparing published data on the two groups of natural DAPs regarding both adverse and beneficial effects on human health. Literature on genotoxic, estrogenic, endocrine-disruptive effects, as well as on the induction of the cellular anti-oxidative defense system, anti-inflammatory properties, the inhibition of kinases, the activation of mitophagy and the induction of autophagy, is gathered and critically reviewed. Indeed, comparing published data suggests similar bioactivity profiles of alternariol and urolithin A. Thus, the current stratification into hazardous Alternaria toxins and healthy urolithins seems debatable. An extrapolation of bioactivities to the other DAP sub-class could serve as a promising base for further research. Conclusively, urolithins should be further evaluated toward high-dose toxicity, while alternariol derivatives could be promising chemicals for the development of therapeutics.
Collapse
Affiliation(s)
- Georg Aichinger
- Laboratory of Toxicology, Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
10
|
Jayaraman PS, Gaston K. Targeting protein kinase CK2 in the treatment of cholangiocarcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:434-447. [PMID: 36045705 PMCID: PMC9400764 DOI: 10.37349/etat.2021.00055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/31/2021] [Indexed: 12/23/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a disease with a very poor prognosis and limited treatment options. Although targeted therapies directed towards specific mutations found in CCA are becoming available and are showing great potential, many tumors do not carry actionable mutations and, in those that do, the emergence of drug resistance is a likely consequence of treatment. Therapeutic targeting of enzymes and other proteins that show elevated activity in CCA cells but which are not altered by mutation is a potential strategy for the treatment of target negative and drug-resistant disease. Protein kinase CK2 (CK2) is a ubiquitously expressed kinase that has increased expression and increased activity in a variety of cancer types including CCA. Several potent CK2 inhibitors are in pre-clinical development or under assessment in a variety of clinical trials often in combination with drugs that induce DNA damage. This review outlines the importance of CK2 in CCA and assesses the progress that has been made in the evaluation of CK2 inhibition as a treatment strategy in this disease. Targeting CK2 based on the expression levels or activity of this protein and/or in combination with drugs that induce DNA damage or inhibit cell cycle progression, could be a viable option for tumors that lack actionable mutations, or for tumors that develop resistance to targeted treatments.
Collapse
Affiliation(s)
- Padma-Sheela Jayaraman
- Biodiscovery Institute, University of Nottingham, NG7 2UH, UK
- Division of Translational Medical Sciences, School of Medicine, University of Nottingham, NG7 2UH, UK
| | - Kevin Gaston
- Biodiscovery Institute, University of Nottingham, NG7 2UH, UK
- Division of Translational Medical Sciences, School of Medicine, University of Nottingham, NG7 2UH, UK
| |
Collapse
|
11
|
Protein kinase CK2: a potential therapeutic target for diverse human diseases. Signal Transduct Target Ther 2021; 6:183. [PMID: 33994545 PMCID: PMC8126563 DOI: 10.1038/s41392-021-00567-7] [Citation(s) in RCA: 200] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/04/2023] Open
Abstract
CK2 is a constitutively active Ser/Thr protein kinase, which phosphorylates hundreds of substrates, controls several signaling pathways, and is implicated in a plethora of human diseases. Its best documented role is in cancer, where it regulates practically all malignant hallmarks. Other well-known functions of CK2 are in human infections; in particular, several viruses exploit host cell CK2 for their life cycle. Very recently, also SARS-CoV-2, the virus responsible for the COVID-19 pandemic, has been found to enhance CK2 activity and to induce the phosphorylation of several CK2 substrates (either viral and host proteins). CK2 is also considered an emerging target for neurological diseases, inflammation and autoimmune disorders, diverse ophthalmic pathologies, diabetes, and obesity. In addition, CK2 activity has been associated with cardiovascular diseases, as cardiac ischemia-reperfusion injury, atherosclerosis, and cardiac hypertrophy. The hypothesis of considering CK2 inhibition for cystic fibrosis therapies has been also entertained for many years. Moreover, psychiatric disorders and syndromes due to CK2 mutations have been recently identified. On these bases, CK2 is emerging as an increasingly attractive target in various fields of human medicine, with the advantage that several very specific and effective inhibitors are already available. Here, we review the literature on CK2 implication in different human pathologies and evaluate its potential as a pharmacological target in the light of the most recent findings.
Collapse
|
12
|
Zhou Y, Lian H, Shen N, Korm S, Kwok Ping Lam A, Layton O, Huiting LN, Li D, Miao K, Zeng A, Landesman-Bollag E, Seldin DC, Fu H, Hong L, Feng H. The multifaceted role of protein kinase CK2 in high-risk acute lymphoblastic leukemia. Haematologica 2021; 106:1461-1465. [PMID: 32817283 PMCID: PMC8094085 DOI: 10.3324/haematol.2020.246918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Indexed: 11/16/2022] Open
Affiliation(s)
- Yun Zhou
- Department of Gynecology, Wuhan University Renmin Hospital, Wuhan, Hubei, P. R. China
| | - Haiwei Lian
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Ning Shen
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Sovannarith Korm
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Andrew Kwok Ping Lam
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Olivia Layton
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Leah N Huiting
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Dun Li
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Kelly Miao
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Aozhuo Zeng
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Esther Landesman-Bollag
- Department of Medicine, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA
| | - David C Seldin
- Department of Medicine, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA
| | - Hui Fu
- Department of Anatomy and Embryology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, P. R. China
| | - Li Hong
- Department of Gynecology, Wuhan University Renmin Hospital, Wuhan, Hubei, P. R. China
| | - Hui Feng
- Department of Pharmacology and Experimental Therapeutics, Department of Medicine, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
13
|
Borgo C, D'Amore C, Cesaro L, Sarno S, Pinna LA, Ruzzene M, Salvi M. How can a traffic light properly work if it is always green? The paradox of CK2 signaling. Crit Rev Biochem Mol Biol 2021; 56:321-359. [PMID: 33843388 DOI: 10.1080/10409238.2021.1908951] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CK2 is a constitutively active protein kinase that assuring a constant level of phosphorylation to its numerous substrates supports many of the most important biological functions. Nevertheless, its activity has to be controlled and adjusted in order to cope with the varying needs of a cell, and several examples of a fine-tune regulation of its activity have been described. More importantly, aberrant regulation of this enzyme may have pathological consequences, e.g. in cancer, chronic inflammation, neurodegeneration, and viral infection. Our review aims at summarizing our current knowledge about CK2 regulation. In the first part, we have considered the most important stimuli shown to affect protein kinase CK2 activity/expression. In the second part, we focus on the molecular mechanisms by which CK2 can be regulated, discussing controversial aspects and future perspectives.
Collapse
Affiliation(s)
- Christian Borgo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Claudio D'Amore
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Luca Cesaro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Stefania Sarno
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Lorenzo A Pinna
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,CNR Institute of Neurosciences, Padova, Italy
| | - Maria Ruzzene
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,CNR Institute of Neurosciences, Padova, Italy
| | - Mauro Salvi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
14
|
Homma MK, Kiko Y, Hashimoto Y, Nagatsuka M, Katagata N, Masui S, Homma Y, Nomizu T. Intracellular localization of CK2α as a prognostic factor in invasive breast carcinomas. Cancer Sci 2021; 112:619-628. [PMID: 33164285 PMCID: PMC7894005 DOI: 10.1111/cas.14728] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/04/2020] [Accepted: 11/04/2020] [Indexed: 01/15/2023] Open
Abstract
Overexpression of the ubiquitous protein kinase, CK2α, has been reported in various human cancers. Here, we demonstrate that nuclear and nucleolar CK2α localization in invasive ductal carcinomas of the breast is a reliable predictor of poor prognosis. Cellular localization of CK2α in nuclei and nucleoli was analyzed immunohistochemically using surgical tissue blocks from 112 patients, who had undergone surgery without neoadjuvant chemotherapy. Clinical data collection and median follow-up period were for more than 5 y. In total, 93.8% of patients demonstrated elevated CK2α expression in nuclei and 36.6% of them displayed elevated expression predominantly in nucleoli. Clinicopathological malignancy was strongly correlated with elevated nuclear and nucleolar CK2α expression. Recurrence-free survival was significantly worse (P = .0002) in patients with positive nucleolar CK2α staining. The 5-y survival rate decreased to a roughly 50% in nucleolar CK2α-positive patients of triple-negative (P = .0069) and p Stage 3 (P = .0073) groups. In contrast, no patients relapsed or died in the triple-negative group who exhibited a lack of nucleolar CK2α staining. Evaluation of nucleolar CK2α staining showed a high secondary index with a hazard ratio of 6.629 (P = .001), following lymph node metastasis with a hazard ratio of 14.30 (P = .0008). Multivariate analysis demonstrated that nucleolar CK2α is an independent factor for recurrence-free survival. Therefore, we propose that histochemical evaluation of nucleolar CK2α-positive staining may be a new and robust prognostic indicator for patients who need further treatment. Functional consequences of nucleolar CK2 dysfunction may be a starting point to facilitate development of novel treatments for invasive breast carcinoma.
Collapse
Affiliation(s)
- Miwako Kato Homma
- Department of Biomolecular SciencesFukushima Medical University School of MedicineFukushimaJapan
| | - Yuichiro Kiko
- Department of Diagnostic PathologyFukushima Medical University School of MedicineFukushimaJapan
| | - Yuko Hashimoto
- Department of Diagnostic PathologyFukushima Medical University School of MedicineFukushimaJapan
| | - Miki Nagatsuka
- Department of SurgeryHoshi General HospitalFukushimaJapan
| | - Naoto Katagata
- Department of SurgeryHoshi General HospitalFukushimaJapan
| | - Seiichiro Masui
- Medical Research CenterFukushima Medical University School of MedicineFukushimaJapan
| | - Yoshimi Homma
- Department of Biomolecular SciencesFukushima Medical University School of MedicineFukushimaJapan
| | - Tadashi Nomizu
- Department of SurgeryHoshi General HospitalFukushimaJapan
| |
Collapse
|
15
|
Xia LW, Ba MY, Liu W, Cheng W, Hu CP, Zhao Q, Yao YF, Sun MR, Duan YT. Triazol: a privileged scaffold for proteolysis targeting chimeras. Future Med Chem 2019; 11:2919-2973. [PMID: 31702389 DOI: 10.4155/fmc-2019-0159] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Current traditional drugs such as enzyme inhibitors and receptor agonists/antagonists present inherent limitations due to occupancy-driven pharmacology as the mode of action. Proteolysis targeting chimeras (PROTACs) are composed of an E3 ligand, a connecting linker and a target protein ligand, and are an attractive approach to specifically knockdown-targeted proteins utilizing an event-driven mode of action. The length, hydrophilicity and rigidity of connecting linkers play important role in creating a successful PROTAC. Some PROTACs with a triazole linker have displayed promising anticancer activity. This review provides an overview of PROTACs with a triazole scaffold and discusses its structure-activity relationship. Important milestones in the development of PROTACs are addressed and a critical analysis of this drug discovery strategy is also presented.
Collapse
Affiliation(s)
- Li-Wen Xia
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
- Collaborative Innovation Center of Henan New Drug Research & Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Meng-Yu Ba
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
- Collaborative Innovation Center of Henan New Drug Research & Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Wei Liu
- Henan Provincial Key Laboratory of Children's Genetics & Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou 450018, China
| | - Weyland Cheng
- Henan Provincial Key Laboratory of Children's Genetics & Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou 450018, China
| | - Chao-Ping Hu
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
- Collaborative Innovation Center of Henan New Drug Research & Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Qing Zhao
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
- Collaborative Innovation Center of Henan New Drug Research & Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Yong-Fang Yao
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
- Collaborative Innovation Center of Henan New Drug Research & Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Mo-Ran Sun
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, China
- Collaborative Innovation Center of Henan New Drug Research & Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Yong-Tao Duan
- Henan Provincial Key Laboratory of Children's Genetics & Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou 450018, China
| |
Collapse
|
16
|
Ong BX, Yoo Y, Han MG, Park JB, Choi MK, Choi Y, Shin JS, Bahn YS, Cho HS. Structural analysis of fungal pathogenicity-related casein kinase α subunit, Cka1, in the human fungal pathogen Cryptococcus neoformans. Sci Rep 2019; 9:14398. [PMID: 31591414 PMCID: PMC6779870 DOI: 10.1038/s41598-019-50678-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 09/16/2019] [Indexed: 12/28/2022] Open
Abstract
CK2α is a constitutively active and highly conserved serine/threonine protein kinase that is involved in the regulation of key cellular metabolic pathways and associated with a variety of tumours and cancers. The most well-known CK2α inhibitor is the human clinical trial candidate CX-4945, which has recently shown to exhibit not only anti-cancer, but also anti-fungal properties. This prompted us to work on the CK2α orthologue, Cka1, from the pathogenic fungus Cryptococcus neoformans, which causes life-threatening systemic cryptococcosis and meningoencephalitis mainly in immunocompromised individuals. At present, treatment of cryptococcosis remains a challenge due to limited anti-cryptococcal therapeutic strategies. Hence, expanding therapeutic options for the treatment of the disease is highly clinically relevant. Herein, we report the structures of Cka1-AMPPNP-Mg2+ (2.40 Å) and Cka1-CX-4945 (2.09 Å). Structural comparisons of Cka1-AMPPNP-Mg2+ with other orthologues revealed the dynamic architecture of the N-lobe across species. This may explain for the difference in binding affinities and deviations in protein-inhibitor interactions between Cka1-CX-4945 and human CK2α-CX-4945. Supporting it, in vitro kinase assay demonstrated that CX-4945 inhibited human CK2α much more efficiently than Cka1. Our results provide structural insights into the design of more selective inhibitors against Cka1.
Collapse
Affiliation(s)
- Belinda X Ong
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Youngki Yoo
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Myeong Gil Han
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jun Bae Park
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Myung Kyung Choi
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yeseul Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Severance Biomedical Science Institute and Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yong-Sun Bahn
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyun-Soo Cho
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
17
|
Ao Y, Zhang J, Liu Z, Qian M, Li Y, Wu Z, Sun P, Wu J, Bei W, Wen J, Wu X, Li F, Zhou Z, Zhu WG, Liu B, Wang Z. Lamin A buffers CK2 kinase activity to modulate aging in a progeria mouse model. SCIENCE ADVANCES 2019; 5:eaav5078. [PMID: 30906869 PMCID: PMC6426468 DOI: 10.1126/sciadv.aav5078] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/31/2019] [Indexed: 05/15/2023]
Abstract
Defective nuclear lamina protein lamin A is associated with premature aging. Casein kinase 2 (CK2) binds the nuclear lamina, and inhibiting CK2 activity induces cellular senescence in cancer cells. Thus, it is feasible that lamin A and CK2 may cooperate in the aging process. Nuclear CK2 localization relies on lamin A and the lamin A carboxyl terminus physically interacts with the CK2α catalytic core and inhibits its kinase activity. Loss of lamin A in Lmna-knockout mouse embryonic fibroblasts (MEFs) confers increased CK2 activity. Conversely, prelamin A that accumulates in Zmpste24-deficent MEFs exhibits a high CK2α binding affinity and concomitantly reduces CK2 kinase activity. Permidine treatment activates CK2 by releasing the interaction between lamin A and CK2, promoting DNA damage repair and ameliorating progeroid features. These data reveal a previously unidentified function for nuclear lamin A and highlight an essential role for CK2 in regulating senescence and aging.
Collapse
Affiliation(s)
- Ying Ao
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen 518060, China
- Department of Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Jie Zhang
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen 518060, China
| | - Zuojun Liu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen 518060, China
| | - Minxian Qian
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen 518060, China
| | - Yao Li
- School of Public Health, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Zhuping Wu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen 518060, China
| | - Pengfei Sun
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen 518060, China
| | - Jie Wu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen 518060, China
| | - Weixin Bei
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen 518060, China
| | - Junqu Wen
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen 518060, China
| | - Xuli Wu
- School of Public Health, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Feng Li
- Department of Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Zhongjun Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong
| | - Wei-Guo Zhu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen 518060, China
| | - Baohua Liu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen 518060, China
- Corresponding author. (Z.W.); (B.L.)
| | - Zimei Wang
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen 518060, China
- Corresponding author. (Z.W.); (B.L.)
| |
Collapse
|
18
|
Lian H, Su M, Zhu Y, Zhou Y, Soomro SH, Fu H. Protein Kinase CK2, a Potential Therapeutic Target in Carcinoma Management. Asian Pac J Cancer Prev 2019; 20:23-32. [PMID: 30677865 PMCID: PMC6485562 DOI: 10.31557/apjcp.2019.20.1.23] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The Protein kinase CK2 (formerly known as casein kinase 2) is a highly conserved serine/ threonine kinase
overexpressed in various human carcinomas and its high expression often correlates with poor prognosis. CK2 protein
is localized in the nucleus of many tumor cells and correlates with clinical features in many cases. Increased expression
of CK2 in mice results in the development of various types of carcinomas (both solids and blood related tumors, such
as (breast carcinoma, lymphoma, etc), which reveals its carcinogenic properties. CK2 plays essential roles in many key
biological processes related to carcinoma, including cell apoptosis, DNA damage responses and cell cycle regulation.
CK2 has become a potential anti-carcinoma target. Various CK2 inhibitors have been developed with anti-neoplastic
properties against a variety of carcinomas. Some CK2 inhibitors have showed good results in in vitro and pre-clinical
models, and have even entered in clinical trials. This article will review effects of CK2 and its inhibitors on common
carcinomas in in vitro and pre-clinical studies.
Collapse
Affiliation(s)
- Haiwei Lian
- Department of Human Anatomy, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, P.R, China.
| | | | | | | | | | | |
Collapse
|
19
|
Shahraki A, Ebrahimi A. Binding of ellagic acid and urolithin metabolites to the CK2 protein, based on the ONIOM method and molecular docking calculations. NEW J CHEM 2019. [DOI: 10.1039/c9nj03508g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Using three-layer ONIOM and molecular docking calculations to investigate the binding of urolithins to the active site of the CK2 protein.
Collapse
Affiliation(s)
- Asiyeh Shahraki
- Department of Chemistry
- Computational Quantum Chemistry Laboratory
- University of Sistan and Baluchestan
- Zahedan
- Iran
| | - Ali Ebrahimi
- Department of Chemistry
- Computational Quantum Chemistry Laboratory
- University of Sistan and Baluchestan
- Zahedan
- Iran
| |
Collapse
|
20
|
Chen H, Chen F, Liu N, Wang X, Gou S. Chemically induced degradation of CK2 by proteolysis targeting chimeras based on a ubiquitin-proteasome pathway. Bioorg Chem 2018; 81:536-544. [PMID: 30245235 DOI: 10.1016/j.bioorg.2018.09.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 02/08/2023]
Abstract
As a ubiquitous, highly pleiotropic and constitutively active serine/threonine protein kinase, casein kinase 2 (CK2) is closely associated with tumorigenesis by its overexpression in cancer cells. Here we report several proteolysis targeting chimeras (PROTACs) via "click reaction" to connect a CK2 inhibitor (CX-4945) and pomalidomide for degradation of CK2 protein. Among them, compound 2 degraded CK2 in a dose and time-dependent manner, and kept CK2 at a low basal level by recruiting ubiquitin-proteasome system. The degradation of CK2 resulted in the reduced phosphorylation of Akt and the up-regulation of p53. As a CK2 protein degrader, 2 showed the analogous cytotoxicity to CX-4945 but with a quite different mechanism of action from the CK2 inhibitor, hinting that degradation of CK2 proteins by PROTACs is a potential way for cancer treatments.
Collapse
Affiliation(s)
- Hong Chen
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China
| | - Feihong Chen
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China
| | - Nannan Liu
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China
| | - Xinyi Wang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China
| | - Shaohua Gou
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China.
| |
Collapse
|
21
|
Schnitzler A, Gratz A, Bollacke A, Weyrich M, Kuckländer U, Wünsch B, Götz C, Niefind K, Jose J. A π-Halogen Bond of Dibenzofuranones with the Gatekeeper Phe113 in Human Protein Kinase CK2 Leads to Potent Tight Binding Inhibitors. Pharmaceuticals (Basel) 2018; 11:ph11010023. [PMID: 29462988 PMCID: PMC5874719 DOI: 10.3390/ph11010023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 01/05/2023] Open
Abstract
Human protein kinase CK2 is an emerging target for neoplastic diseases. Potent lead structures for human CK2 inhibitors are derived from dibenzofuranones. Two new derivatives, 7,9-dichloro-1,2-dihydro-8-hydroxy-4-[(4-methoxyphenylamino)-methylene]dibenzo[b,d]furan-3(2H)-one (4a) and (E)-1,3-dichloro-6-[(4-methoxyphenylimino)-methyl]dibenzo[b,d]furan-2,7-diol (5) were tested for inhibition of CK2 and induction of apoptosis in LNCaP cells. Both turned out to be tight binding inhibitors, with IC50 values of 7 nM (4a) and 5 nM (5) and an apparent Ki value of 0.4 nM for both. Compounds 4a and 5 reduced cellular CK2 activity, indicating cell permeability. Cell viability was substantially impaired in LNCaP cells, as well as apoptosis was induced, which was not appearing in non-neoplastic ARPE-19 cells. Co-crystallization of 4a and 5 revealed an unexpected π-halogen bond of the chloro substituent at C9 with the gatekeeper amino acid Phe113, leading to an inverted binding mode in comparison to parent compound 4b, with the Cl at C6 instead, which was co-crystallized as a control. This indicates that the position of the chloro substituent on ring A of the dibenzofuran scaffold is responsible for an inversion of the binding mode that enhances potency.
Collapse
Affiliation(s)
- Alexander Schnitzler
- Institut für Biochemie, Department für Chemie, Universität zu Köln, Zülpicher Straße 47, D-50674 Köln, Germany.
| | - Andreas Gratz
- Institut für Pharmazeutische und Medizinische Chemie, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany.
| | - Andre Bollacke
- Institut für Pharmazeutische und Medizinische Chemie, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany.
| | - Michael Weyrich
- Medizinische Biochemie und Molekularbiologie, Universität des Saarlandes, Kirrberger Str., Geb. 44, D-66421 Homburg, Germany.
| | - Uwe Kuckländer
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany.
| | - Claudia Götz
- Medizinische Biochemie und Molekularbiologie, Universität des Saarlandes, Kirrberger Str., Geb. 44, D-66421 Homburg, Germany.
| | - Karsten Niefind
- Institut für Biochemie, Department für Chemie, Universität zu Köln, Zülpicher Straße 47, D-50674 Köln, Germany.
| | - Joachim Jose
- Institut für Pharmazeutische und Medizinische Chemie, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany.
| |
Collapse
|
22
|
Chua MMJ, Lee M, Dominguez I. Cancer-type dependent expression of CK2 transcripts. PLoS One 2017; 12:e0188854. [PMID: 29206231 PMCID: PMC5714396 DOI: 10.1371/journal.pone.0188854] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 11/14/2017] [Indexed: 01/31/2023] Open
Abstract
A multitude of proteins are aberrantly expressed in cancer cells, including the oncogenic serine-threonine kinase CK2. In a previous report, we found increases in CK2 transcript expression that could explain the increased CK2 protein levels found in tumors from lung and bronchus, prostate, breast, colon and rectum, ovarian and pancreatic cancers. We also found that, contrary to the current notions about CK2, some CK2 transcripts were downregulated in several cancers. Here, we investigate all other cancers using Oncomine to determine whether they also display significant CK2 transcript dysregulation. As anticipated from our previous analysis, we found cancers with all CK2 transcripts upregulated (e.g. cervical), and cancers where there was a combination of upregulation and/or downregulation of the CK2 transcripts (e.g. sarcoma). Unexpectedly, we found some cancers with significant downregulation of all CK2 transcripts (e.g. testicular cancer). We also found that, in some cases, CK2 transcript levels were already dysregulated in benign lesions (e.g. Barrett’s esophagus). We also found that CK2 transcript upregulation correlated with lower patient survival in most cases where data was significant. However, there were two cancer types, glioblastoma and renal cell carcinoma, where CK2 transcript upregulation correlated with higher survival. Overall, these data show that the expression levels of CK2 genes is highly variable in cancers and can lead to different patient outcomes.
Collapse
Affiliation(s)
- Melissa M. J. Chua
- Department of Medicine, Boston University School of Medicine, Boston MA, United States of America
| | - Migi Lee
- Department of Medicine, Boston University School of Medicine, Boston MA, United States of America
| | - Isabel Dominguez
- Department of Medicine, Boston University School of Medicine, Boston MA, United States of America
- * E-mail:
| |
Collapse
|
23
|
Buontempo F, McCubrey JA, Orsini E, Ruzzene M, Cappellini A, Lonetti A, Evangelisti C, Chiarini F, Evangelisti C, Barata JT, Martelli AM. Therapeutic targeting of CK2 in acute and chronic leukemias. Leukemia 2017; 32:1-10. [PMID: 28951560 PMCID: PMC5770594 DOI: 10.1038/leu.2017.301] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/06/2017] [Accepted: 09/08/2017] [Indexed: 12/22/2022]
Abstract
CK2 is a ubiquitously expressed, constitutively active Ser/Thr protein kinase, which is considered the most pleiotropic protein kinase in the human kinome. Such a pleiotropy explains the involvement of CK2 in many cellular events. However, its predominant roles are stimulation of cell growth and prevention of apoptosis. High levels of CK2 messenger RNA and protein are associated with CK2 pathological functions in human cancers. Over the last decade, basic and translational studies have provided evidence of CK2 as a pivotal molecule driving the growth of different blood malignancies. CK2 overexpression has been demonstrated in nearly all the types of hematological cancers, including acute and chronic leukemias, where CK2 is a key regulator of signaling networks critical for cell proliferation, survival and drug resistance. The findings that emerged from these studies suggest that CK2 could be a valuable therapeutic target in leukemias and supported the initiation of clinical trials using CK2 antagonists. In this review, we summarize the recent advances on the understanding of the signaling pathways involved in CK2 inhibition-mediated effects with a particular emphasis on the combinatorial use of CK2 inhibitors as novel therapeutic strategies for treating both acute and chronic leukemia patients.
Collapse
Affiliation(s)
- F Buontempo
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - J A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - E Orsini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - M Ruzzene
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - A Cappellini
- Department of Human, Social and Health Sciences, University of Cassino, Cassino, Italy
| | - A Lonetti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - C Evangelisti
- Institute of Molecular Genetics, National Research Council, Bologna, Italy.,Cell and Molecular Biology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - F Chiarini
- Institute of Molecular Genetics, National Research Council, Bologna, Italy.,Cell and Molecular Biology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - C Evangelisti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - J T Barata
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - A M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
24
|
Wadey KS, Brown BA, Sala-Newby GB, Jayaraman PS, Gaston K, George SJ. Protein kinase CK2 inhibition suppresses neointima formation via a proline-rich homeodomain-dependent mechanism. Vascul Pharmacol 2017; 99:34-44. [PMID: 28927755 PMCID: PMC5718878 DOI: 10.1016/j.vph.2017.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 09/05/2017] [Accepted: 09/14/2017] [Indexed: 11/19/2022]
Abstract
Neointimal hyperplasia is a product of VSMC replication and consequent accumulation within the blood vessel wall. In this study, we determined whether inhibition of protein kinase CK2 and the resultant stabilisation of proline-rich homeodomain (PRH) could suppress VSMC proliferation. Both silencing and pharmacological inhibition of CK2 with K66 antagonised replication of isolated VSMCs. SiRNA-induced knockdown as well as ectopic overexpression of proline-rich homeodomain indicated that PRH disrupts cell cycle progression. Mutation of CK2 phosphorylation sites Ser163 and Ser177 within the PRH homeodomain enabled prolonged cell cycle arrest by PRH. Concomitant knockdown of PRH and inhibition of CK2 with K66 indicated that the anti-proliferative action of K66 required the presence of PRH. Both K66 and adenovirus-mediated gene transfer of S163C:S177C PRH impaired neointima formation in human saphenous vein organ cultures. Importantly, neither intervention had notable effects on cell cycle progression, cell survival or migration in cultured endothelial cells.
Collapse
MESH Headings
- Animals
- Casein Kinase II/antagonists & inhibitors
- Casein Kinase II/genetics
- Casein Kinase II/metabolism
- Cell Cycle Checkpoints/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/enzymology
- Humans
- Hyperplasia
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Mutation
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Neointima
- Phosphorylation
- Proline-Rich Protein Domains
- Protein Kinase Inhibitors/pharmacology
- RNA Interference
- Rats
- Saphenous Vein/drug effects
- Saphenous Vein/enzymology
- Saphenous Vein/pathology
- Signal Transduction/drug effects
- Tissue Culture Techniques
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transfection
Collapse
Affiliation(s)
- K S Wadey
- School of Clinical Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK; Department of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - B A Brown
- School of Clinical Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - G B Sala-Newby
- School of Clinical Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - P-S Jayaraman
- Division of Immunity and Infection, College of Medicine, University Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - K Gaston
- Department of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - S J George
- School of Clinical Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK.
| |
Collapse
|
25
|
Chua MMJ, Ortega CE, Sheikh A, Lee M, Abdul-Rassoul H, Hartshorn KL, Dominguez I. CK2 in Cancer: Cellular and Biochemical Mechanisms and Potential Therapeutic Target. Pharmaceuticals (Basel) 2017; 10:E18. [PMID: 28134850 PMCID: PMC5374422 DOI: 10.3390/ph10010018] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/23/2017] [Accepted: 01/23/2017] [Indexed: 01/09/2023] Open
Abstract
CK2 genes are overexpressed in many human cancers, and most often overexpression is associated with worse prognosis. Site-specific expression in mice leads to cancer development (e.g., breast, lymphoma) indicating the oncogenic nature of CK2. CK2 is involved in many key aspects of cancer including inhibition of apoptosis, modulation of signaling pathways, DNA damage response, and cell cycle regulation. A number of CK2 inhibitors are now available and have been shown to have activity against various cancers in vitro and in pre-clinical models. Some of these inhibitors are now undergoing exploration in clinical trials as well. In this review, we will examine some of the major cancers in which CK2 inhibition has promise based on in vitro and pre-clinical studies, the proposed cellular and signaling mechanisms of anti-cancer activity by CK2 inhibitors, and the current or recent clinical trials using CK2 inhibitors.
Collapse
Affiliation(s)
- Melissa M J Chua
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| | - Charina E Ortega
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| | - Ayesha Sheikh
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| | - Migi Lee
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| | - Hussein Abdul-Rassoul
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| | - Kevan L Hartshorn
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| | - Isabel Dominguez
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| |
Collapse
|
26
|
Franchin C, Borgo C, Zaramella S, Cesaro L, Arrigoni G, Salvi M, Pinna LA. Exploring the CK2 Paradox: Restless, Dangerous, Dispensable. Pharmaceuticals (Basel) 2017; 10:ph10010011. [PMID: 28117670 PMCID: PMC5374415 DOI: 10.3390/ph10010011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/12/2017] [Accepted: 01/16/2017] [Indexed: 12/28/2022] Open
Abstract
The history of protein kinase CK2 is crowded with paradoxes and unanticipated findings. Named after a protein (casein) that is not among its physiological substrates, CK2 remained in search of its targets for more than two decades after its discovery in 1954, but it later came to be one of the most pleiotropic protein kinases. Being active in the absence of phosphorylation and/or specific stimuli, it looks unsuitable to participate in signaling cascades, but its “lateral” implication in a variety of signaling pathways is now soundly documented. At variance with many “onco-kinases”, CK2 is constitutively active, and no oncogenic CK2 mutant is known; still high CK2 activity correlates to neoplasia. Its pleiotropy and essential role may cast doubts on the actual “druggability” of CK2; however, a CK2 inhibitor is now in Phase II clinical trials for the treatment of cancer, and cell clones viable in the absence of CK2 are providing information about the mechanism by which cancer becomes addicted to high CK2 levels. A phosphoproteomics analysis of these CK2 null cells suggests that CK2 pleiotropy may be less pronounced than expected and supports the idea that the phosphoproteome generated by this kinase is flexible and not rigidly pre-determined.
Collapse
Affiliation(s)
- Cinzia Franchin
- Department of Biomedical Sciences, University of Padova, via U. Bassi, 58/B, 35131 Padova, Italy.
- Proteomics Center, University of Padova and Azienda Ospedaliera di Padova, via G. Orus, 2/B, 35129 Padova, Italy.
| | - Christian Borgo
- Department of Biomedical Sciences, University of Padova, via U. Bassi, 58/B, 35131 Padova, Italy.
| | - Silvia Zaramella
- Proteomics Center, University of Padova and Azienda Ospedaliera di Padova, via G. Orus, 2/B, 35129 Padova, Italy.
| | - Luca Cesaro
- Department of Biomedical Sciences, University of Padova, via U. Bassi, 58/B, 35131 Padova, Italy.
| | - Giorgio Arrigoni
- Department of Biomedical Sciences, University of Padova, via U. Bassi, 58/B, 35131 Padova, Italy.
- Proteomics Center, University of Padova and Azienda Ospedaliera di Padova, via G. Orus, 2/B, 35129 Padova, Italy.
| | - Mauro Salvi
- Department of Biomedical Sciences, University of Padova, via U. Bassi, 58/B, 35131 Padova, Italy.
| | - Lorenzo A Pinna
- Department of Biomedical Sciences, University of Padova, via U. Bassi, 58/B, 35131 Padova, Italy.
- CNR Neurosciences Institute, via U. Bassi, 58/B, 35131 Padova, Italy.
| |
Collapse
|
27
|
Bandyopadhyay M, Arbet S, Bishop CP, Bidwai AP. Drosophila Protein Kinase CK2: Genetics, Regulatory Complexity and Emerging Roles during Development. Pharmaceuticals (Basel) 2016; 10:E4. [PMID: 28036067 PMCID: PMC5374408 DOI: 10.3390/ph10010004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 12/12/2016] [Accepted: 12/19/2016] [Indexed: 01/06/2023] Open
Abstract
CK2 is a Ser/Thr protein kinase that is highly conserved amongst all eukaryotes. It is a well-known oncogenic kinase that regulates vital cell autonomous functions and animal development. Genetic studies in the fruit fly Drosophila are providing unique insights into the roles of CK2 in cell signaling, embryogenesis, organogenesis, neurogenesis, and the circadian clock, and are revealing hitherto unknown complexities in CK2 functions and regulation. Here, we review Drosophila CK2 with respect to its structure, subunit diversity, potential mechanisms of regulation, developmental abnormalities linked to mutations in the gene encoding CK2 subunits, and emerging roles in multiple aspects of eye development. We examine the Drosophila CK2 "interaction map" and the eye-specific "transcriptome" databases, which raise the prospect that this protein kinase has many additional targets in the developing eye. We discuss the possibility that CK2 functions during early retinal neurogenesis in Drosophila and mammals bear greater similarity than has been recognized, and that this conservation may extend to other developmental programs. Together, these studies underscore the immense power of the Drosophila model organism to provide new insights and avenues to further investigate developmentally relevant targets of this protein kinase.
Collapse
Affiliation(s)
| | - Scott Arbet
- Department of Biology, West Virginia University, Morgantown, WV 26506, USA.
| | - Clifton P Bishop
- Department of Biology, West Virginia University, Morgantown, WV 26506, USA.
| | - Ashok P Bidwai
- Department of Biology, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
28
|
Götz C, Montenarh M. Protein kinase CK2 in development and differentiation. Biomed Rep 2016; 6:127-133. [PMID: 28357063 DOI: 10.3892/br.2016.829] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/06/2016] [Indexed: 12/15/2022] Open
Abstract
Among the human kinomes, protein kinase CK2 (formerly termed casein kinase II) is considered to be essential, as it is implicated in the regulation of various cellular processes. Experiments with pharmacological inhibitors of the kinase activity of CK2 provide evidence that CK2 is essential for development and differentiation. Therefore, the present review addresses the role of CK2 during embryogenesis, neuronal, adipogenic, osteogenic and myogenic differentiation in established model cell lines, and in embryonic, neural and mesenchymal stem cells. CK2 kinase activity appears to be essential in the early stages of differentiation, as CK2 inhibition at early time points generally prevents differentiation. In addition, the present review reports on target proteins of CK2 in embryogenesis and differentiation.
Collapse
Affiliation(s)
- Claudia Götz
- Department of Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Germany
| | - Mathias Montenarh
- Department of Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Germany
| |
Collapse
|
29
|
Gowda C, Song C, Kapadia M, Payne JL, Hu T, Ding Y, Dovat S. Regulation of cellular proliferation in acute lymphoblastic leukemia by Casein Kinase II (CK2) and Ikaros. Adv Biol Regul 2016; 63:71-80. [PMID: 27666503 DOI: 10.1016/j.jbior.2016.09.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 09/09/2016] [Indexed: 12/23/2022]
Abstract
The IKZF1 gene encodes the Ikaros protein, a zinc finger transcriptional factor that acts as a master regulator of hematopoiesis and a tumor suppressor in leukemia. Impaired activity of Ikaros is associated with the development of high-risk acute lymphoblastic leukemia (ALL) with a poor prognosis. The molecular mechanisms that regulate Ikaros' function as a tumor suppressor and regulator of cellular proliferation are not well understood. We demonstrated that Ikaros is a substrate for Casein Kinase II (CK2), an oncogenic kinase that is overexpressed in ALL. Phosphorylation of Ikaros by CK2 impairs Ikaros' DNA-binding ability, as well as Ikaros' ability to regulate gene expression and function as a tumor suppressor in leukemia. Targeting CK2 with specific inhibitors restores Ikaros' function as a transcriptional regulator and tumor suppressor resulting in a therapeutic, anti-leukemia effect in a preclinical model of ALL. Here, we review the genes and pathways that are regulated by Ikaros and the molecular mechanisms through which Ikaros and CK2 regulate cellular proliferation in leukemia.
Collapse
Affiliation(s)
- Chandrika Gowda
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Chunhua Song
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Malika Kapadia
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Jonathon L Payne
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; Loma Linda University, Loma Linda, CA, USA
| | - Tommy Hu
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Yali Ding
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Sinisa Dovat
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
30
|
Verma S, Goyal S, Jamal S, Singh A, Grover A. Hsp90: Friends, clients and natural foes. Biochimie 2016; 127:227-240. [PMID: 27295069 DOI: 10.1016/j.biochi.2016.05.018] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/29/2016] [Indexed: 12/13/2022]
Abstract
Hsp90, a homodimeric ATPase, is responsible for the correct folding of a number of newly synthesized polypeptides in addition to the correct folding of denatured/misfolded client proteins. It requires several co-chaperones and other partner proteins for chaperone activity. Due to the involvement of Hsp90-dependent client proteins in a variety of oncogenic signaling pathways, Hsp90 inhibition has emerged as one of the leading strategies for anticancer chemotherapeutics. Most of Hsp90 inhibitors blocks the N terminal ATP binding pocket and prevents the conformational changes which are essential for the loading of co-chaperones and client proteins. Several other inhibitors have also been reported which disrupt chaperone cycle in ways other than binding to N terminal ATP binding pocket. The Hsp90 inhibition is associated with heat shock response, mediated by HSF-1, to overcome the loss of Hsp90 and sustain cell survival. This review is an attempt to give an over view of all the important players of chaperone cycle.
Collapse
Affiliation(s)
- Sharad Verma
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Sukriti Goyal
- Department of Bioscience and Biotechnology, Banasthali University, Tonk, Rajasthan 304022, India.
| | - Salma Jamal
- Department of Bioscience and Biotechnology, Banasthali University, Tonk, Rajasthan 304022, India.
| | - Aditi Singh
- Department of Biotechnology, TERI University, VasantKunj, New Delhi 110 070, India.
| | - Abhinav Grover
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
31
|
Rosenberger AFN, Morrema THJ, Gerritsen WH, van Haastert ES, Snkhchyan H, Hilhorst R, Rozemuller AJM, Scheltens P, van der Vies SM, Hoozemans JJM. Increased occurrence of protein kinase CK2 in astrocytes in Alzheimer's disease pathology. J Neuroinflammation 2016; 13:4. [PMID: 26732432 PMCID: PMC4702323 DOI: 10.1186/s12974-015-0470-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 12/24/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disease. In addition to the occurrence of amyloid deposits and widespread tau pathology, AD is associated with a neuroinflammatory response characterized by the activation of microglia and astrocytes. Protein kinase 2 (CK2, former casein kinase II) is involved in a wide variety of cellular processes. Previous studies on CK2 in AD showed controversial results, and the involvement of CK2 in neuroinflammation in AD remains elusive. METHODS In this study, we used immunohistochemical and immunofluorescent staining methods to investigate the localization of CK2 in the hippocampus and temporal cortex of patients with AD and non-demented controls. We compared protein levels with Western blotting analysis, and we investigated CK2 activity in human U373 astrocytoma cells and human primary adult astrocytes stimulated with IL-1β or TNF-α. RESULTS We report increased levels of CK2 in the hippocampus and temporal cortex of AD patients compared to non-demented controls. Immunohistochemical analysis shows CK2 immunoreactivity in astrocytes in AD and control cases. In AD, the presence of CK2 immunoreactive astrocytes is increased. CK2 immunopositive astrocytes are associated with amyloid deposits, suggesting an involvement of CK2 in the neuroinflammatory response. In U373 cells and human primary astrocytes, the selective CK2 inhibitor CX-4945 shows a dose-dependent reduction of the IL-1β or TNF-α induced MCP-1 and IL-6 secretion. CONCLUSIONS This data suggests that CK2 in astrocytes is involved in the neuroinflammatory response in AD. The reduction in pro-inflammatory cytokine secretion by human astrocytes using the selective CK2 inhibitor CX-4945 indicates that CK2 could be a potential target to modulate neuroinflammation in AD.
Collapse
Affiliation(s)
- Andrea F N Rosenberger
- Alzheimer center & Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands.
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Tjado H J Morrema
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Wouter H Gerritsen
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Elise S van Haastert
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Hripsime Snkhchyan
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Riet Hilhorst
- PamGene International BV, Wolvenhoek 10, 5211 HH, 's-Hertogenbosch, The Netherlands.
| | - Annemieke J M Rozemuller
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Philip Scheltens
- Alzheimer center & Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands.
| | - Saskia M van der Vies
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Jeroen J M Hoozemans
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
32
|
Abstract
INTRODUCTION The conventional term 'casein kinase' (CK) denotes three classes of kinases - CK1, CK2 and Golgi-CK (G-CK)/Fam20C (family with sequence similarity 20, member C) - sharing the ability to phoshorylate casein in vitro, but otherwise unrelated to each other. All CKs have been reported to be implicated in human diseases, and reviews individually dealing with the druggability of CK1 and CK2 are available. Our aim is to provide a comparative analysis of the three classes of CKs as therapeutic targets. AREAS COVERED CK2 is the CK for which implication in neoplasia is best documented, with the survival of cancer cells often relying on its overexpression. An ample variety of cell-permeable CK2 inhibitors have been developed, with a couple of these now in clinical trials. Isoform-specific CK1 inhibitors that are expected to play a beneficial role in oncology and neurodegeneration have been also developed. In contrast, the pathogenic potential of G-CK/Fam20C is caused by its loss of function. Activators of Fam20C, notably sphingolipids and their analogs, may prove beneficial in this respect. EXPERT OPINION Optimization of CK2 and CK1 inhibitors will prove useful to develop new therapeutic strategies for treating cancer and neurodegenerative disorders, while the design of potent activators of G-CK/Fam20C will provide a new tool in the fields of bio-mineralization and hypophosphatemic diseases.
Collapse
Affiliation(s)
- Giorgio Cozza
- a 1 University of Padova, Department of Biomedical Sciences , Via Ugo Bassi 58B, 35131 Padova, Italy
| | - Lorenzo A Pinna
- a 1 University of Padova, Department of Biomedical Sciences , Via Ugo Bassi 58B, 35131 Padova, Italy .,b 2 University of Padova, Department of Biomedical Sciences and CNR Institute of Neurosciences , Padova, Italy ;
| |
Collapse
|
33
|
Song C, Gowda C, Pan X, Ding Y, Tong Y, Tan BH, Wang H, Muthusami S, Ge Z, Sachdev M, Amin SG, Desai D, Gowda K, Gowda R, Robertson GP, Schjerven H, Muschen M, Payne KJ, Dovat S. Targeting casein kinase II restores Ikaros tumor suppressor activity and demonstrates therapeutic efficacy in high-risk leukemia. Blood 2015; 126:1813-22. [PMID: 26219304 PMCID: PMC4600018 DOI: 10.1182/blood-2015-06-651505] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 07/16/2015] [Indexed: 12/13/2022] Open
Abstract
Ikaros (IKZF1) is a tumor suppressor that binds DNA and regulates expression of its target genes. The mechanism of Ikaros activity as a tumor suppressor and the regulation of Ikaros function in leukemia are unknown. Here, we demonstrate that Ikaros controls cellular proliferation by repressing expression of genes that promote cell cycle progression and the phosphatidylinositol-3 kinase (PI3K) pathway. We show that Ikaros function is impaired by the pro-oncogenic casein kinase II (CK2), and that CK2 is overexpressed in leukemia. CK2 inhibition restores Ikaros function as transcriptional repressor of cell cycle and PI3K pathway genes, resulting in an antileukemia effect. In high-risk leukemia where one IKZF1 allele has been deleted, CK2 inhibition restores the transcriptional repressor function of the remaining wild-type IKZF1 allele. CK2 inhibition demonstrated a potent therapeutic effect in a panel of patient-derived primary high-risk B-cell acute lymphoblastic leukemia xenografts as indicated by prolonged survival and a reduction of leukemia burden. We demonstrate the efficacy of a novel therapeutic approach for high-risk leukemia: restoration of Ikaros tumor suppressor activity via inhibition of CK2. These results provide a rationale for the use of CK2 inhibitors in clinical trials for high-risk leukemia, including cases with deletion of one IKZF1 allele.
Collapse
Affiliation(s)
- Chunhua Song
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA
| | - Chandrika Gowda
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA
| | - Xiaokang Pan
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA
| | - Yali Ding
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA
| | - Yongqing Tong
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA
| | - Bi-Hua Tan
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA
| | - Haijun Wang
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA
| | - Sunil Muthusami
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA
| | - Zheng Ge
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA; Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Mansi Sachdev
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA
| | - Shantu G Amin
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Dhimant Desai
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Krishne Gowda
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Raghavendra Gowda
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Gavin P Robertson
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Hilde Schjerven
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA; and
| | - Markus Muschen
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA; and
| | - Kimberly J Payne
- Department of Pathology and Human Anatomy, Loma Linda University, Loma Linda, CA
| | - Sinisa Dovat
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA
| |
Collapse
|
34
|
Protein kinase CK2 is necessary for the adipogenic differentiation of human mesenchymal stem cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2207-16. [DOI: 10.1016/j.bbamcr.2015.05.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 05/21/2015] [Accepted: 05/23/2015] [Indexed: 11/17/2022]
|
35
|
Filhol O, Giacosa S, Wallez Y, Cochet C. Protein kinase CK2 in breast cancer: the CK2β regulatory subunit takes center stage in epithelial plasticity. Cell Mol Life Sci 2015; 72:3305-22. [PMID: 25990538 PMCID: PMC11113558 DOI: 10.1007/s00018-015-1929-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 05/06/2015] [Accepted: 05/11/2015] [Indexed: 12/11/2022]
Abstract
Structurally, protein kinase CK2 consists of two catalytic subunits (α and α') and two regulatory subunits (β), which play a critical role in targeting specific CK2 substrates. Compelling evidence shows the complexity of the CK2 cellular signaling network and supports the view that this enzyme is a key component of regulatory protein kinase networks that are involved in several aspects of cancer. CK2 both activates and suppresses the expression of a number of essential oncogenes and tumor suppressors, and its expression and activity are upregulated in blood tumors and virtually all solid tumors. The prognostic significance of CK2α expression in association with various clinicopathological parameters highlighted this kinase as an adverse prognostic marker in breast cancer. In addition, several recent studies reported its implication in the regulation of the epithelial-to-mesenchymal transition (EMT), an early step in cancer invasion and metastasis. In this review, we briefly overview the contribution of CK2 to several aspects of cancer and discuss how in mammary epithelial cells, the expression of its CK2β regulatory subunit plays a critical role in maintaining an epithelial phenotype through CK2-mediated control of key EMT-related transcription factors. Importantly, decreased CK2β expression in breast tumors is correlated with inefficient phosphorylation and nuclear translocation of Snail1 and Foxc2, ultimately leading to EMT induction. This review highlights the pivotal role played by CK2β in the mammary epithelial phenotype and discusses how a modest alteration in its expression may be sufficient to induce dramatic effects facilitating the early steps in tumor cell dissemination through the coordinated regulation of two key transcription factors.
Collapse
Affiliation(s)
- Odile Filhol
- Institut National de la Santé et de la Recherche Médicale, U1036, Grenoble, France
- Institute of Life Sciences Research and Technologies, Biology of Cancer and Infection, Commissariat à l’Energie Atomique, Grenoble, France
- Unité Mixte de Recherche-S1036, University of Grenoble Alpes, Grenoble, France
| | - Sofia Giacosa
- Institut National de la Santé et de la Recherche Médicale, U1036, Grenoble, France
- Institute of Life Sciences Research and Technologies, Biology of Cancer and Infection, Commissariat à l’Energie Atomique, Grenoble, France
- Unité Mixte de Recherche-S1036, University of Grenoble Alpes, Grenoble, France
| | - Yann Wallez
- Institut National de la Santé et de la Recherche Médicale, U1036, Grenoble, France
- Institute of Life Sciences Research and Technologies, Biology of Cancer and Infection, Commissariat à l’Energie Atomique, Grenoble, France
- Unité Mixte de Recherche-S1036, University of Grenoble Alpes, Grenoble, France
| | - Claude Cochet
- Institut National de la Santé et de la Recherche Médicale, U1036, Grenoble, France
- Institute of Life Sciences Research and Technologies, Biology of Cancer and Infection, Commissariat à l’Energie Atomique, Grenoble, France
- Unité Mixte de Recherche-S1036, University of Grenoble Alpes, Grenoble, France
| |
Collapse
|
36
|
Cheeseman K, Weitzman JB. Host–parasite interactions: an intimate epigenetic relationship. Cell Microbiol 2015; 17:1121-32. [DOI: 10.1111/cmi.12471] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 05/28/2015] [Accepted: 06/01/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Kevin Cheeseman
- Sorbonne Paris Cité Epigenetics and Cell Fate UMR 7216 CNRS Université Paris Diderot Paris France
| | - Jonathan B. Weitzman
- Sorbonne Paris Cité Epigenetics and Cell Fate UMR 7216 CNRS Université Paris Diderot Paris France
| |
Collapse
|
37
|
Briguglio I, Piras S, Corona P, Gavini E, Nieddu M, Boatto G, Carta A. Benzotriazole: An overview on its versatile biological behavior. Eur J Med Chem 2015; 97:612-48. [PMID: 25293580 PMCID: PMC7115563 DOI: 10.1016/j.ejmech.2014.09.089] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 09/25/2014] [Accepted: 09/28/2014] [Indexed: 12/13/2022]
Abstract
Discovered in late 1960, azoles are heterocyclic compounds class which constitute the largest group of available antifungal drugs. Particularly, the imidazole ring is the chemical component that confers activity to azoles. Triazoles are obtained by a slight modification of this ring and similar or improved activities as well as less adverse effects are reported for triazole derivatives. Consequently, it is not surprising that benzimidazole/benzotriazole derivatives have been found to be biologically active. Since benzimidazole has been widely investigated, this review is focused on defining the place of benzotriazole derivatives in biomedical research, highlighting their versatile biological properties, the mode of action and Structure Activity Relationship (SAR) studies for a variety of antimicrobial, antiparasitic, and even antitumor, choleretic, cholesterol-lowering agents.
Collapse
Affiliation(s)
- I Briguglio
- Department of Chemistry and Pharmacy, University of Sassari, Via Muroni 23/A, 07100 Sassari, Italy
| | - S Piras
- Department of Chemistry and Pharmacy, University of Sassari, Via Muroni 23/A, 07100 Sassari, Italy
| | - P Corona
- Department of Chemistry and Pharmacy, University of Sassari, Via Muroni 23/A, 07100 Sassari, Italy
| | - E Gavini
- Department of Chemistry and Pharmacy, University of Sassari, Via Muroni 23/A, 07100 Sassari, Italy
| | - M Nieddu
- Department of Chemistry and Pharmacy, University of Sassari, Via Muroni 23/A, 07100 Sassari, Italy
| | - G Boatto
- Department of Chemistry and Pharmacy, University of Sassari, Via Muroni 23/A, 07100 Sassari, Italy
| | - A Carta
- Department of Chemistry and Pharmacy, University of Sassari, Via Muroni 23/A, 07100 Sassari, Italy.
| |
Collapse
|
38
|
Kim SW, Hasanuzzaman M, Cho M, Heo YR, Ryu MJ, Ha NY, Park HJ, Park HY, Shin JG. Casein Kinase 2 (CK2)-mediated Phosphorylation of Hsp90β as a Novel Mechanism of Rifampin-induced MDR1 Expression. J Biol Chem 2015; 290:17029-40. [PMID: 25995454 DOI: 10.1074/jbc.m114.624106] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Indexed: 12/15/2022] Open
Abstract
The P-glycoprotein (P-gp) encoded by the MDR1 gene is a drug-exporting transporter located in the cellular membrane. P-gp induction is regarded as one of the main mechanisms underlying drug-induced resistance. Although there is great interest in the regulation of P-gp expression, little is known about its underlying regulatory mechanisms. In this study, we demonstrate that casein kinase 2 (CK2)-mediated phosphorylation of heat shock protein 90β (Hsp90β) and subsequent stabilization of PXR is a key mechanism in the regulation of MDR1 expression. Furthermore, we show that CK2 is directly activated by rifampin. Upon exposure to rifampin, CK2 catalyzes the phosphorylation of Hsp90β at the Ser-225/254 residues. Phosphorylated Hsp90β then interacts with PXR, causing a subsequent increase in its stability, leading to the induction of P-gp expression. In addition, inhibition of CK2 and Hsp90β enhances the down-regulation of PXR and P-gp expression. The results of this study may facilitate the development of new strategies to prevent multidrug resistance and provide a plausible mechanism for acquired drug resistance by CK2-mediated regulation of P-gp expression.
Collapse
Affiliation(s)
- So Won Kim
- From the Department of Pharmacology and the Institute for Clinical and Translational Research, Catholic Kwandong University College of Medicine, Gangneung 210-701, Korea, the Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 614-735, Korea, the Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan 614-735, Korea
| | - Md Hasanuzzaman
- the Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 614-735, Korea
| | - Munju Cho
- the Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 614-735, Korea
| | - Ye Rang Heo
- the Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 614-735, Korea
| | - Min-Jung Ryu
- the Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 614-735, Korea
| | - Na-Young Ha
- the Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 614-735, Korea
| | - Hyun June Park
- the Bio-MAX Institute, Seoul National University, Seoul 151-742, Korea, and
| | - Hyung-Yeon Park
- the Agriculture and Biotechnology Department, Noroo Holdings Co. Ltd., Seoul 135-983, Korea
| | - Jae-Gook Shin
- the Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 614-735, Korea, the Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan 614-735, Korea,
| |
Collapse
|
39
|
Abstract
The IKZF1 gene at 7p12.2 codes for IKAROS (also termed IKZF1), an essential transcription factor in haematopoiesis involved primarily in lymphoid differentiation. Its importance is underlined by the fact that deregulation of IKAROS results in leukaemia in both mice and men. During recent years, constitutional as well as acquired genetic changes of IKZF1 have been associated with human disease. For example, certain germline single nucleotide polymorphisms in IKZF1 have been shown to increase the risk of some disorders and abnormal expression and somatic rearrangements, mutations and deletions of IKZF1 (ΔIKZF1) have been detected in a wide variety of human malignancies. Of immediate clinical importance is the fact that ΔIKZF1 occurs in 15% of paediatric B-cell precursor acute lymphoblastic leukaemia (BCP ALL) and that the presence of ΔIKZF1 is associated with an increased risk of relapse and a poor outcome; in some studies such deletions have been shown to be an independent risk factor also when minimal residual disease data are taken into account. However, cooperative genetic changes, such as ERG deletions and CRLF2 rearrangements, may modify the prognostic impact of ΔIKZF1, for better or worse. This review summarizes our current knowledge of IKZF1 abnormalities in human disease, with an emphasis on BCP ALL.
Collapse
Affiliation(s)
- Linda Olsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | | |
Collapse
|
40
|
Srivastava R, Akthar S, Sharma R, Mishra S. Identification of Ellagic acid analogues as potent inhibitor of protein Kinase CK2:A chemopreventive role in oral Cancer. Bioinformation 2015; 11:21-6. [PMID: 25780276 PMCID: PMC4349935 DOI: 10.6026/97320630011021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 03/18/2014] [Indexed: 11/23/2022] Open
Abstract
Over expression of Protein kinase (CK2) suppresses apoptosis induced by a variety of agents, whereas down-regulation of CK2 sensitizes cells to induction of apoptosis. In this study, we have built quantitative structure activity relationship (QSAR) models, which were trained and tested on experimentally verified 38 enzyme׳s inhibitors having inhibitory value IC50 in µM. These inhibitors were docked at the active site of CK2 (PDB id: 2ZJW) using AutoDock software, which resulted in energy-based descriptors such as binding energy, intermol energy, torsional energy, internal energy and docking energy. For QSAR modeling, Multiple Linear Regression (MLR) model was engendered using energy-based descriptors yielding correlation coefficient r(2) of 0.4645. To assess the predictive performance of QSAR models, different cross-validation procedures were adopted. Our results suggests that ligand-receptor binding interactions for CK2 employing QSAR modeling seems to be a promising approach for prediction of IC50 value of a new ligand molecule against CK2.Further, twenty analogues of ellagic acid were docked with CK2 structure. After docking, two compounds CID 46229200 and CID 10003463 had lower docking energy even lower than standard control Ellagic acid with CK2 was selected as potent candidate drugs for Oral cancer. The biological activity of two compounds in terms of IC50 was predicted based on QSAR model, which could be used as a guideline for anticancerous activity of compounds before their synthesis.
Collapse
Affiliation(s)
- Rashi Srivastava
- Department of Biotechnology, IFTM University, Moradabad-244 001, U.P., India
| | - Salman Akthar
- Department of Biotechnology, Integral University, Lucknow 226 015, U.P. India
| | - Rolee Sharma
- Department of Biotechnology, Integral University, Lucknow 226 015, U.P. India
| | - Sanjay Mishra
- Department of Biotechnology, IFTM University, Moradabad-244 001, U.P., India
| |
Collapse
|
41
|
Wang H, Song C, Gurel Z, Song N, Ma J, Ouyang H, Lai L, Payne KJ, Dovat S. Protein phosphatase 1 (PP1) and Casein Kinase II (CK2) regulate Ikaros-mediated repression of TdT in thymocytes and T-cell leukemia. Pediatr Blood Cancer 2014; 61:2230-5. [PMID: 25214003 PMCID: PMC4205270 DOI: 10.1002/pbc.25221] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 07/20/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND Ikaros is a DNA-binding protein that acts as master-regulator of hematopoiesis and a tumor suppressor. In thymocytes and T-cell leukemia, Ikaros negatively regulates transcription of terminal deoxynucleotide transferase (TdT), a key protein in lymphocyte differentiation. The signaling pathways that regulate Ikaros-mediated repression of TdT are unknown. Our previous work identified Casein Kinase II (CK2) and Protein Phosphatase 1 (PP1) as regulators of Ikaros DNA binding activity. Here, we investigated the role of PP1 and CK2 in regulating Ikaros-mediated control of TdT expression. PROCEDURES Ikaros phosphomimetic and phosphoresistant mutants and specific CK2 and PP1 inhibitors were used in combination with quantitative chromatin immunoprecipitation (qChIP) and quantitative reverse transcriptase-PCR (q RT-PCR) assays to evaluate the role of CK2 and PP1 in regulating the ability of Ikaros to bind the TdT promoter and to regulate TdT expression. RESULTS We demonstrate that phosphorylation of Ikaros by pro-oncogenic CK2 decreases Ikaros binding to the promoter of the TdT gene and reduces the ability of Ikaros to repress TdT expression during thymocyte differentiation. CK2 inhibition and PP1 activity restore Ikaros DNA-binding affinity toward the TdT promoter, as well as Ikaros-mediated transcriptional repression of TdT in primary thymocytes and in leukemia. CONCLUSION These data establish that PP1 and CK2 signal transduction pathways regulate Ikaros-mediated repression of TdT in thymocytes and leukemia. These findings reveal that PP1 and CK2 have opposing effects on Ikaros-mediated repression of TdT and establish novel roles for PP1 and CK2 signaling in thymocyte differentiation and leukemia.
Collapse
Affiliation(s)
- Haijun Wang
- School of Pharmacology, Wenzhou Medical University, Wenzhou 325035, China,Pennsylvania State University College of Medicine, Hershey, PA 17033,College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Chunhua Song
- Pennsylvania State University College of Medicine, Hershey, PA 17033
| | | | - Na Song
- School of Pharmacology, Wenzhou Medical University, Wenzhou 325035, China
| | - Jisheng Ma
- School of Pharmacology, Wenzhou Medical University, Wenzhou 325035, China
| | - Hongsheng Ouyang
- College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Liangxue Lai
- College of Animal Sciences, Jilin University, Changchun 130062, China
| | | | - Sinisa Dovat
- Pennsylvania State University College of Medicine, Hershey, PA 17033,Corresponding author: Sinisa Dovat, MD PhD, Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, Phone: 717-531-6012, FAX: 717-531-4789,
| |
Collapse
|
42
|
Wang X, Gupta P, Fairbanks J, Hansen D. Protein kinase CK2 both promotes robust proliferation and inhibits the proliferative fate in the C. elegans germ line. Dev Biol 2014; 392:26-41. [PMID: 24824786 DOI: 10.1016/j.ydbio.2014.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 04/02/2014] [Accepted: 05/02/2014] [Indexed: 11/18/2022]
Abstract
Stem cells are capable of both self-renewal (proliferation) and differentiation. Determining the regulatory mechanisms controlling the balance between stem cell proliferation and differentiation is not only an important biological question, but also holds the key for using stem cells as therapeutic agents. The Caenorhabditis elegans germ line has emerged as a valuable model to study the molecular mechanisms controlling stem cell behavior. In this study, we describe a large-scale RNAi screen that identified kin-10, which encodes the β subunit of protein kinase CK2, as a novel factor regulating stem cell proliferation in the C. elegans germ line. While a loss of kin-10 in an otherwise wild-type background results in a decrease in the number of proliferative cells, loss of kin-10 in sensitized genetic backgrounds results in a germline tumor. Therefore, kin-10 is not only necessary for robust proliferation, it also inhibits the proliferative fate. We found that kin-10's regulatory role in inhibiting the proliferative fate is carried out through the CK2 holoenzyme, rather than through a holoenzyme-independent function, and that it functions downstream of GLP-1/Notch signaling. We propose that a loss of kin-10 leads to a defect in CK2 phosphorylation of its downstream targets, resulting in abnormal activity of target protein(s) that are involved in the proliferative fate vs. differentiation decision. This eventually causes a shift towards the proliferative fate in the stem cell fate decision.
Collapse
Affiliation(s)
- Xin Wang
- Department of Biological Sciences, University of Calgary, 2500 University Drive, Calgary, Alberta, Canada T2N 1N4
| | - Pratyush Gupta
- Department of Biological Sciences, University of Calgary, 2500 University Drive, Calgary, Alberta, Canada T2N 1N4
| | - Jared Fairbanks
- Department of Biological Sciences, University of Calgary, 2500 University Drive, Calgary, Alberta, Canada T2N 1N4
| | - Dave Hansen
- Department of Biological Sciences, University of Calgary, 2500 University Drive, Calgary, Alberta, Canada T2N 1N4.
| |
Collapse
|
43
|
Lee YH, Kang BS, Bae YS. Premature senescence in human breast cancer and colon cancer cells by tamoxifen-mediated reactive oxygen species generation. Life Sci 2013; 97:116-22. [PMID: 24361399 DOI: 10.1016/j.lfs.2013.12.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 11/26/2013] [Accepted: 12/07/2013] [Indexed: 02/07/2023]
Abstract
AIMS Cellular senescence is an important tumor suppression process in vivo. Tamoxifen is a well-known anti-breast cancer drug; however, its molecular function is poorly understood. Here, we examined whether tamoxifen promotes senescence in breast cancer and colon cancer cells for the first time. MAIN METHODS Human breast cancer MCF-7, T47D, and MDA-MB-435 and colorectal cancer HCT116 cells were treated with tamoxifen. Cellular senescence was measured by SA-β-gal staining and based on the protein expression of p53 and p21(Cip1/WAF1). The production of reactive oxygen species (ROS) was determined by staining with CM-H2DCFDA and dihydroethidium (DHE). CK2 activity was assessed with a specific peptide substrate. KEY FINDINGS Tamoxifen promoted senescence phenotype and ROS generation in MCF-7 and HCT116 cells. The ROS scavenger, N-acetyl-l-cysteine (NAC), and the NADPH oxidase inhibitor, apocynin, almost completely abolished this event. Tamoxifen inhibited the catalytic activity of CK2. Overexpression of CK2α antagonized senescence mediated by tamoxifen, indicating that tamoxifen induced senescence via a CK2-dependent pathway. A well-known CK2 inhibitor, 5,6-dichloro-1-β-d-ribofuranosylbenzimidazole (DRB), also stimulated ROS production and senescence in MCF-7 cells. Finally, experiments using T47D (wild-type p53) and MDA-MB-435 (mutant p53) cell lines suggested that tamoxifen induces p53-independent ROS production as well as p53-dependent senescence in breast cancer cells. SIGNIFICANCE These results demonstrate that tamoxifen promotes senescence through a ROS-p53-p21(Cip1/WAF1) dependent pathway by inhibiting CK2 activity in breast cancer and colon cancer cells.
Collapse
Affiliation(s)
- Young-Hoon Lee
- School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Beom Sik Kang
- School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Young-Seuk Bae
- School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea.
| |
Collapse
|
44
|
Identification of a novel protein interaction motif in the regulatory subunit of casein kinase 2. Mol Cell Biol 2013; 34:246-58. [PMID: 24216761 DOI: 10.1128/mcb.00968-13] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Casein kinase 2 (CK2) regulates multiple cellular processes and can promote oncogenesis. Interactions with the CK2β regulatory subunit of the enzyme target its catalytic subunit (CK2α or CK2α') to specific substrates; however, little is known about the mechanisms by which these interactions occur. We previously showed that by binding CK2β, the Epstein-Barr virus (EBV) EBNA1 protein recruits CK2 to promyelocytic leukemia (PML) nuclear bodies, where increased CK2-mediated phosphorylation of PML proteins triggers their degradation. Here we have identified a KSSR motif near the dimerization interface of CK2β as forming part of a protein interaction pocket that mediates interaction with EBNA1. We show that the EBNA1-CK2β interaction is primed by phosphorylation of EBNA1 on S393 (within a polyserine region). This phosphoserine is critical for EBNA1-induced PML degradation but does not affect EBNA1 functions in EBV replication or segregation. Using comparative proteomics of wild-type (WT) and KSSR mutant CK2β, we identified an uncharacterized cellular protein, C18orf25/ARKL1, that also binds CK2β through the KSSR motif and show that this involves a polyserine sequence resembling the CK2β binding sequence in EBNA1. Therefore, we have identified a new mechanism of CK2 interaction used by viral and cellular proteins.
Collapse
|
45
|
Structural and functional insights into the regulation mechanism of CK2 by IP6 and the intrinsically disordered protein Nopp140. Proc Natl Acad Sci U S A 2013; 110:19360-5. [PMID: 24218616 DOI: 10.1073/pnas.1304670110] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Protein kinase CK2 is a ubiquitous kinase that can phosphorylate hundreds of cellular proteins and plays important roles in cell growth and development. Deregulation of CK2 is related to a variety of human cancers, and CK2 is regarded as a suppressor of apoptosis; therefore, it is a target of anticancer therapy. Nucleolar phosphoprotein 140 (Nopp140), which is an intrinsically disordered protein, interacts with CK2 and inhibits the latter's catalytic activity in vitro. Interestingly, the catalytic activity of CK2 is recovered in the presence of d-myo-inositol 1,2,3,4,5,6-hexakisphosphate (IP6). IP6 is widely distributed in animal cells, but the molecular mechanisms that govern its cellular functions in animal cells have not been completely elucidated. In this study, the crystal structure of CK2 in complex with IP6 showed that the lysine-rich cluster of CK2 plays an important role in binding to IP6. The biochemical experiments revealed that a Nopp140 fragment (residues 568-596) and IP6 competitively bind to the catalytic subunit of CK2 (CK2α), and phospho-Ser574 of Nopp140 significantly enhances its interaction with CK2α. Substitutions of K74E, K76E, and K77E in CK2α significantly reduced the interactions of CK2α with both IP6 and the Nopp140-derived peptide. Our study gives an insight into the regulation of CK2. In particular, our work suggests that CK2 activity is inhibited by Nopp140 and reactivated by IP6 by competitive binding at the substrate recognition site of CK2.
Collapse
|
46
|
Turowec JP, Vilk G, Gabriel M, Litchfield DW. Characterizing the convergence of protein kinase CK2 and caspase-3 reveals isoform-specific phosphorylation of caspase-3 by CK2α': implications for pathological roles of CK2 in promoting cancer cell survival. Oncotarget 2013; 4:560-71. [PMID: 23599180 PMCID: PMC3720604 DOI: 10.18632/oncotarget.948] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Protein kinase CK2 has emerged as a promising candidate for the treatment of a number of cancers. This enzyme is comprised of two catalytic subunits (CK2 and/or CK2α′) that form complexes with homodimers of regulatory CK2β subunits. While catalytic and regulatory CK2 subunits are generally expressed at similar levels to form tetrameric complexes, asymmetric expression of CK2 subunits has been associated with various forms of cancer and the enhanced survival of cancer cells. To elucidate mechanisms responsible for regulation of cancer cell survival by CK2, we recently employed computational and experimental strategies that revealed widespread overlap between sites for CK2 phosphorylation and caspase cleavage. Among candidates with overlapping CK2 and caspase cleavage sites was caspase-3 that is phosphorylated by CK2 to prevent its activation by upstream caspases. To elucidate the precise relationship between CK2 and caspase-3, we modulated expression of individual CK2 subunits and demonstrated that CK2α′ exhibits a striking preference for caspase-3 phosphorylation in cells as compared to CK2α and that CK2β exhibits the capacity to abolish caspase-3 phosphorylation. Since caspase-3 represents the first CK2 substrate selectively phosphorylated by CK2α′ in cells, our work highlights divergent functions of the different forms of CK2. Given the involvement of CK2 in a diverse series of biological events and its association with various cancers, this work has important implications for identifying pathological roles of distinct forms of CK2 that could instruct efforts to selectively target individual CK2 subunits for therapy.
Collapse
Affiliation(s)
- Jacob P Turowec
- Department of Biochemistry, Schulich School of Medicine, Western University, London, ON, Canada
| | | | | | | |
Collapse
|
47
|
Lee YH, Yuk HJ, Park KH, Bae YS. Coumestrol induces senescence through protein kinase CKII inhibition-mediated reactive oxygen species production in human breast cancer and colon cancer cells. Food Chem 2013; 141:381-8. [PMID: 23768371 DOI: 10.1016/j.foodchem.2013.03.053] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 03/10/2013] [Accepted: 03/15/2013] [Indexed: 12/31/2022]
Abstract
An inhibitor of the protein kinase CKII (CKII) was purified from leaves of Glycine max (L.) Merrill and was identified as coumestrol by structural analysis. Coumestrol inhibited the phosphotransferase activity of CKII toward β-casein, with an IC50 of about 5 μM. It acted as a competitive inhibitor with respect to ATP as a substrate, with an apparent Ki value of 7.67 μM. Coumestrol at 50μM resulted in 50% and 30% growth inhibition of human breast cancer MCF-7 and colorectal cancer HCT116 cells, respectively. Coumestrol promoted senescence through the p53-p21(Cip1/WAF1) pathway by inducing reactive oxygen species (ROS) production in MCF-7 and HCT116 cells. The ROS scavenger N-acetyl-l-cysteine (NAC), NADPH oxidase inhibitor apocynin and p22(phox) siRNA almost completely abolished this event. Overexpression of CKIIα antagonised cellular senescence mediated by coumestrol, indicating that this compound induced senescence via a CKII-dependent pathway. Since senescence is an important tumour suppression process in vivo, these results suggest that coumestrol can function by inhibiting oncogenic disease, at least in part, through CKII inhibition-mediated cellular senescence.
Collapse
Affiliation(s)
- Young-Hoon Lee
- School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | | | | | | |
Collapse
|
48
|
HUNG MINGSZU, XU ZHIDONG, CHEN YU, SMITH EMMANUEL, MAO JIANHUA, HSIEH DAVID, LIN YUCHING, YANG CHENGTA, JABLONS DAVIDM, YOU LIANG. Hematein, a casein kinase II inhibitor, inhibits lung cancer tumor growth in a murine xenograft model. Int J Oncol 2013; 43:1517-1522. [PMID: 24008396 PMCID: PMC3823374 DOI: 10.3892/ijo.2013.2087] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 08/09/2013] [Indexed: 12/27/2022] Open
Abstract
Casein kinase II (CK2) inhibitors suppress cancer cell growth. In this study, we examined the inhibitory effects of a novel CK2 inhibitor, hematein, on tumor growth in a murine xenograft model. We found that in lung cancer cells, hematein inhibited cancer cell growth, Akt/PKB Ser129 phosphorylation, the Wnt/TCF pathway and increased apoptosis. In a murine xenograft model of lung cancer, hematein inhibited tumor growth without significant toxicity to the mice tested. Molecular docking showed that hematein binds to CK2α in durable binding sites. Collectively, our results suggest that hematein is an allosteric inhibitor of protein kinase CK2 and has antitumor activity to lung cancer.
Collapse
Affiliation(s)
- MING-SZU HUNG
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94115,
USA
| | - ZHIDONG XU
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94115,
USA
| | - YU CHEN
- Department of Molecular Medicine, College of Medicine, University of South Florida, Tampa, FL
| | - EMMANUEL SMITH
- Department of Molecular Medicine, College of Medicine, University of South Florida, Tampa, FL
| | - JIAN-HUA MAO
- Life Sciences Division, Lawrence Berkeley National Laboratory, University of California, Berkeley, CA,
USA
| | - DAVID HSIEH
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94115,
USA
| | - YU-CHING LIN
- Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi branch
| | - CHENG-TA YANG
- Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan branch
- Department of Respiratory Care, College of Medicine, Chang Gung University, Taoyuan, Taiwan,
R.O.C
| | - DAVID M. JABLONS
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94115,
USA
| | - LIANG YOU
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94115,
USA
| |
Collapse
|
49
|
Abstract
Hsp90 is a major molecular chaperone that is expressed abundantly and plays a pivotal role in assisting correct folding and functionality of its client proteins in cells. The Hsp90 client proteins include a wide variety of signal transducing molecules such as protein kinases and steroid hormone receptors. Cancer is a complex disease, but most types of human cancer share common hallmarks, including self-sufficiency in growth signals, insensitivity to growth-inhibitory mechanism, evasion of programmed cell death, limitless replicative potential, sustained angiogenesis, and tissue invasion and metastasis. A surprisingly large number of Hsp90-client proteins play crucial roles in establishing cancer cell hallmarks. We start the review by describing the structure and function of Hsp90 since conformational changes during the ATPase cycle of Hsp90 are closely related to its function. Many co-chaperones, including Hop, p23, Cdc37, Aha1, and PP5, work together with Hsp90 by modulating the chaperone machinery. Post-translational modifications of Hsp90 and its cochaperones are vital for their function. Many tumor-related Hsp90-client proteins, including signaling kinases, steroid hormone receptors, p53, and telomerase, are described. Hsp90 and its co-chaperones are required for the function of these tumor-promoting client proteins; therefore, inhibition of Hsp90 by specific inhibitors such as geldanamycin and its derivatives attenuates the tumor progression. Hsp90 inhibitors can be potential and effective cancer chemotherapeutic drugs with a unique profile and have been examined in clinical trials. We describe possible mechanisms why Hsp90 inhibitors show selectivity to cancer cells even though Hsp90 is essential also for normal cells. Finally, we discuss the "Hsp90-addiction" of cancer cells, and suggest a role for Hsp90 in tumor evolution.
Collapse
Affiliation(s)
- Yoshihiko Miyata
- Department of Cell & Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan.
| | | | | |
Collapse
|
50
|
Miyata Y, Nakamoto H, Neckers L. The therapeutic target Hsp90 and cancer hallmarks. Curr Pharm Des 2013; 19:347-65. [PMID: 22920906 DOI: 10.2174/138161213804143725] [Citation(s) in RCA: 258] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 08/15/2012] [Indexed: 01/22/2023]
Abstract
Hsp90 is a major molecular chaperone that is expressed abundantly and plays a pivotal role in assisting correct folding and functionality of its client proteins in cells. The Hsp90 client proteins include a wide variety of signal transducing molecules such as protein kinases and steroid hormone receptors. Cancer is a complex disease, but most types of human cancer share common hallmarks, including self-sufficiency in growth signals, insensitivity to growth-inhibitory mechanism, evasion of programmed cell death, limitless replicative potential, sustained angiogenesis, and tissue invasion and metastasis. A surprisingly large number of Hsp90-client proteins play crucial roles in establishing cancer cell hallmarks. We start the review by describing the structure and function of Hsp90 since conformational changes during the ATPase cycle of Hsp90 are closely related to its function. Many co-chaperones, including Hop, p23, Cdc37, Aha1, and PP5, work together with Hsp90 by modulating the chaperone machinery. Post-translational modifications of Hsp90 and its cochaperones are vital for their function. Many tumor-related Hsp90-client proteins, including signaling kinases, steroid hormone receptors, p53, and telomerase, are described. Hsp90 and its co-chaperones are required for the function of these tumor-promoting client proteins; therefore, inhibition of Hsp90 by specific inhibitors such as geldanamycin and its derivatives attenuates the tumor progression. Hsp90 inhibitors can be potential and effective cancer chemotherapeutic drugs with a unique profile and have been examined in clinical trials. We describe possible mechanisms why Hsp90 inhibitors show selectivity to cancer cells even though Hsp90 is essential also for normal cells. Finally, we discuss the "Hsp90-addiction" of cancer cells, and suggest a role for Hsp90 in tumor evolution.
Collapse
Affiliation(s)
- Yoshihiko Miyata
- Department of Cell & Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan.
| | | | | |
Collapse
|