1
|
Ems M, Brichkina A, Lauth M. A safe haven for cancer cells: tumor plus stroma control by DYRK1B. Oncogene 2025; 44:341-347. [PMID: 39863750 PMCID: PMC11790486 DOI: 10.1038/s41388-025-03275-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
The development of resistance remains one of the biggest challenges in clinical cancer patient care and it comprises all treatment modalities from chemotherapy to targeted or immune therapy. In solid malignancies, drug resistance is the result of adaptive processes occurring in cancer cells or the surrounding tumor microenvironment (TME). Future therapy attempts will therefore benefit from targeting both, tumor and stroma compartments and drug targets which affect both sides will be highly appreciated. In this review, we describe a seemingly paradoxical oncogenic mediator with this potential: The dual-specificity tyrosine-phosphorylation regulated kinase 1B (DYRK1B). DYRK1B promotes proliferative quiescence and yet is overexpressed or amplified in many hyperproliferative malignancies including ovarian cancer and pancreatic cancer. In particular the latter disease is a paradigmatic example for a therapy-recalcitrant and highly stroma-rich cancer entity. Here, recent evidence suggests that DYRK1B exerts its oncogenic features by installing a protective niche for cancer cells by directly affecting cancer cells but also the TME. Specifically, DYRK1B not only fosters cell-intrinsic processes like cell survival, chemoresistance, and disease recurrence, but it also upregulates TME and cancer cell-protective innate immune checkpoints and down-modulates anti-tumoral macrophage functionality. In this article, we outline the well-established cell-autonomous roles of DYRK1B and extend its importance to the TME and the control of the tumor immune stroma. In summary, DYRK1B appears as a single novel key player creating a safe haven for cancer cells by acting cell-intrinsically and-extrinsically, leading to the promotion of cancer cell survival, chemoresistance, and relapse. Thus, DYRK1B appears as an attractive drug target for future therapeutic approaches.
Collapse
Affiliation(s)
- Miriam Ems
- Department of Gastroenterology, Endocrinology and Metabolism, Center for Tumor and Immune Biology, Philipps University Marburg, Marburg, Germany
| | - Anna Brichkina
- Institute of Systems Immunology, Philipps University Marburg, Marburg, Germany
| | - Matthias Lauth
- Department of Gastroenterology, Endocrinology and Metabolism, Center for Tumor and Immune Biology, Philipps University Marburg, Marburg, Germany.
| |
Collapse
|
2
|
Tolu-Akinnawo O, Ogunniyi KE. Accelerated Coronary Artery Disease in a Patient With Advanced Lung Cancer: Interplay of Cancer, Inflammation, and Treatment Effects. Cureus 2025; 17:e77769. [PMID: 39981449 PMCID: PMC11841481 DOI: 10.7759/cureus.77769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
Cardiovascular disease (CVD) and lung cancer are among the most prevalent causes of death worldwide, representing substantial public health challenges. The relationship between coronary artery disease (CAD) and lung cancer is potentially multifaceted, influenced by common risk factors and the adverse cardiac effects of cancer treatments. However, cases of accelerated CAD occurring within a year of cancer treatment initiation are rarely reported. We present a case of a Black American male undergoing combination therapy (chemotherapy/radiotherapy/immunotherapy) for small-cell lung cancer, who developed ST-elevation myocardial infarction (STEMI) seven months after beginning treatment. Although we do not have the official records, the patient reported that he had previously undergone a cardiac workup (including an echo/stress test) at an outside facility a year prior, due to persistent dyspnea, which was unremarkable. The patient underwent successful percutaneous coronary intervention and was discharged on apixaban and prasugrel. This study underscores the importance of maintaining a high index of suspicion for acute coronary syndromes (ACS) in patients receiving lung cancer treatments, emphasizes the need for early recognition of warning signs, and highlights the critical role of risk factor management and enhanced surveillance in this vulnerable population.
Collapse
|
3
|
Sester S, Wilms G, Ahlburg J, Babendreyer A, Becker W. Elevated expression levels of the protein kinase DYRK1B induce mesenchymal features in A549 lung cancer cells. BMC Cancer 2024; 24:1341. [PMID: 39482615 PMCID: PMC11529244 DOI: 10.1186/s12885-024-13057-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/11/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND The protein kinase DYRK1B is a negative regulator of cell proliferation but has been found to be overexpressed in diverse human solid cancers. While DYRK1B is recognized to promote cell survival and adaption to stressful conditions, the consequences of elevated DYRK1B levels in cancer cells are largely uncharted. METHODS To elucidate the role of DYRK1B in cancer cells, we established a A549 lung adenocarcinoma cell model featuring conditional overexpression of DYRK1B. This system was used to characterize the impact of heightened DYRK1B levels on gene expression and to monitor phenotypic and functional changes. RESULTS A549 cells with induced overexpression of wild type DYRK1B acquired a mesenchymal cell morphology with diminished cell-cell contacts and a reorganization of the pericellular actin cytoskeleton into stress fibers. This transition was not observed in cells overexpressing a catalytically impaired DYRK1B variant. The phenotypic changes were associated with increased expression of the transcription factors SNAIL and SLUG, which are core regulators of epithelial mesenchymal transition (EMT). Further profiling of DYRK1B-overexpressing cells revealed transcriptional changes that are characteristic for the mesenchymal conversion of epithelial cells, including the upregulation of genes that are related to cancer cell invasion and metastasis. Functionally, DYRK1B overexpression enhanced the migratory capacity of A549 cells in a wound healing assay. CONCLUSIONS The present data identify DYRK1B as a regulator of phenotypic plasticity in A549 cells. Increased expression of DYRK1B induces mesenchymal traits in A549 lung adenocarcinoma cells.
Collapse
Affiliation(s)
- Soraya Sester
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Gerrit Wilms
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Joana Ahlburg
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Aachen, Germany
| | - Aaron Babendreyer
- Institute of Molecular Pharmacology, RWTH Aachen University, Aachen, Germany
| | - Walter Becker
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
4
|
Vorwerk VA, Wilms G, Babendreyer A, Becker W. Differential regulation of expression of the protein kinases DYRK1A and DYRK1B in cancer cells. Sci Rep 2024; 14:23926. [PMID: 39397076 PMCID: PMC11471791 DOI: 10.1038/s41598-024-74190-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
The protein kinases DYRK1A and DYRK1B are pivotal regulators of cell cycle progression by promoting cell cycle exit into quiescence. DYRK1B appears to play a more important role in cancer cell quiescence than DYRK1A, as evidenced by its overexpression or copy number variations in human tumour samples. Nonetheless, the stimuli driving DYRK1B upregulation and the potential divergence in expression patterns between DYRK1A and DYRK1B remain largely elusive. In the present study, we scrutinized the regulatory pathways modulating DYRK1B expression relative to DYRK1A in PANC-1 and A549 cancer cell lines across varying conditions. Serum deprivation, pharmacological mTOR inhibition and increased cell density resulted in the differential upregulation of DYRK1B compared to DYRK1A. We then aimed to assess the role of protein kinases MST1 and MST2, which are key transmitters of cell density dependent effects. Unexpectedly, exposure to the MST1/2 inhibitor XMU-MP-1 resulted in increased DYRK1B levels in A549 cells. Further investigation into the off-target effects of XMU-MP-1 unveiled the inhibition of Aurora kinases (AURKA and AURKB) as a potential causative factor. Consistently, AURK inhibitors VX-680 (tozasertib), MLN8237 (alisertib), AZD1152-HQPA (barasertib) resulted in the upregulation of DYRK1B expression in A549 cells. In summary, our findings indicate that the expression of DYRK1A and DYRK1B is differentially regulated in cancer cells and reveal that the kinase inhibitor XMU-MP-1 increases DYRK1B expression likely through off target inhibition of Aurora kinases.
Collapse
Affiliation(s)
- Vincent Andreas Vorwerk
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Gerrit Wilms
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Aaron Babendreyer
- Institute of Molecular Pharmacology, RWTH Aachen University, 52074, Aachen, Germany
| | - Walter Becker
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
5
|
Li L, Zou Y, Shen C, Chen N, Tong M, Liu R, Wang J, Ning G. Hepatic Dyrk1b impairs systemic glucose homeostasis by modulating Wbp2 expression in a kinase activity-dependent manner. Heliyon 2024; 10:e36726. [PMID: 39296215 PMCID: PMC11407929 DOI: 10.1016/j.heliyon.2024.e36726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/05/2024] [Accepted: 08/21/2024] [Indexed: 09/21/2024] Open
Abstract
Patients with gain-of-function mutations of Dyrk1b have higher fasting blood glucose (FBG) levels. However, the role of Dyrk1b in glucose metabolism is not fully elucidated. Herein, we found that hepatic Dyrk1b overexpression in mice impaired systemic glucose tolerance and hepatic insulin signaling. Dyrk1b overexpression in vitro attenuated insulin signaling in a kinase activity-dependent manner, and its kinase activity was required for its effect on systemic glucose homeostasis and hepatic insulin signaling in vivo. Dyrk1b ablation improved systemic glucose tolerance and hepatic insulin signaling in mice. Quantitative proteomic analyses showed that Dyrk1b downregulated WW domain-binding protein 2 (Wbp2) protein abundance. Mechanistically, Dyrk1b enhanced Wbp2 ubiquitylation and proteasomal degradation. Restoration of hepatic Wbp2 partially rescued the impaired glucose homeostasis in Dyrk1b overexpression mice. In addition, Dyrk1b inhibition with AZ191 moderately improved systemic glucose homeostasis. Our study uncovers that hepatic Dyrk1b impairs systemic glucose homeostasis via its modulation of Wbp2 expression in a kinase activity-dependent manner.
Collapse
Affiliation(s)
- Lianju Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Yaoyu Zou
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200025, China
| | - Chongrong Shen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Na Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Muye Tong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| |
Collapse
|
6
|
Mostafa N, Chen PJ, Darwish SS, Su YC, Shiao MH, Piazza GA, Abadi AH, Engel M, Abdel-Halim M. N-Benzylated 5-Hydroxybenzothiophene-2-carboxamides as Multi-Targeted Clk/Dyrk Inhibitors and Potential Anticancer Agents. Cancers (Basel) 2024; 16:2033. [PMID: 38893153 PMCID: PMC11171218 DOI: 10.3390/cancers16112033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Numerous studies have reported that Dyrk1A, Dyrk1B, and Clk1 are overexpressed in multiple cancers, suggesting a role in malignant disease. Here, we introduce a novel class of group-selective kinase inhibitors targeting Dyrk1A, Dyrk1B, and Clk1. This was achieved by modifying our earlier selective Clk1 inhibitors, which were based on the 5-methoxybenzothiophene-2-carboxamide scaffold. By incorporating a 5-hydroxy group, we increased the potential for additional hydrogen bond interactions that broadened the inhibitory effect to include Dyrk1A and Dyrk1B kinases. Within this series, compounds 12 and 17 emerged as the most potent multi-kinase inhibitors against Dyrk1A, Dyrk1B, and Clk1. Furthermore, when assessed against the most closely related kinases also implicated in cancer, the frontrunner compounds revealed additional inhibitory activity against Haspin and Clk2. Compounds 12 and 17 displayed high potency across various cancer cell lines with minimal effect on non-tumor cells. By examining the effect of these inhibitors on cell cycle distribution, compound 17 retained cells in the G2/M phase and induced apoptosis. Compounds 12 and 17 could also increase levels of cleaved caspase-3 and Bax, while decreasing the expression of the antiapoptotic Bcl-2 protein. These findings support the further study and development of these compounds as novel anticancer therapeutics.
Collapse
Affiliation(s)
- Noha Mostafa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt; (N.M.); (S.S.D.); (A.H.A.)
- Department of Pharmaceutical Chemistry, School of Pharmacy, Newgiza University, Cairo 12256, Egypt
| | - Po-Jen Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan;
- Graduate Institute of Medicine, College of Medicine, I-Shou University, Kaohsiung 824410, Taiwan;
| | - Sarah S. Darwish
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt; (N.M.); (S.S.D.); (A.H.A.)
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, Cairo 11578, Egypt
| | - Yu-Chieh Su
- Graduate Institute of Medicine, College of Medicine, I-Shou University, Kaohsiung 824410, Taiwan;
- Division of Hematology-Oncology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 824410, Taiwan
| | - Ming-Hua Shiao
- Taiwan Instrument Research Institute, National Applied Research Laboratories, Hsinchu 300092, Taiwan;
| | - Gary A. Piazza
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36832, USA;
| | - Ashraf H. Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt; (N.M.); (S.S.D.); (A.H.A.)
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, D-66123 Saarbrücken, Germany
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt; (N.M.); (S.S.D.); (A.H.A.)
| |
Collapse
|
7
|
Abd El-Rahman YA, Chen PJ, ElHady AK, Chen SH, Lin HC, El-Gamil DS, Aboushady Y, Abadi AH, Engel M, Abdel-Halim M. Development of 5-hydroxybenzothiophene derivatives as multi-kinase inhibitors with potential anti-cancer activity. Future Med Chem 2024; 16:1239-1254. [PMID: 38989990 PMCID: PMC11249150 DOI: 10.1080/17568919.2024.2342708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/09/2024] [Indexed: 07/12/2024] Open
Abstract
Aim: Chemoresistance in cancer challenges the classical therapeutic strategy of 'one molecule-one target'. To combat this, multi-target therapies that inhibit various cancer-relevant targets simultaneously are proposed. Methods & results: We introduce 5-hydroxybenzothiophene derivatives as effective multi-target kinase inhibitors, showing notable growth inhibitory activity across different cancer cell lines. Specifically, compound 16b, featuring a 5-hydroxybenzothiophene hydrazide scaffold, emerged as a potent inhibitor, displaying low IC50 values against key kinases and demonstrating significant anti-cancer effects, particularly against U87MG glioblastoma cells. It induced G2/M cell cycle arrest, apoptosis and inhibited cell migration by modulating apoptotic markers. Conclusion: 16b represents a promising lead for developing new anti-cancer agents targeting multiple kinases with affinity to the hydroxybenzothiophene core.
Collapse
Affiliation(s)
- Yara A Abd El-Rahman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy & Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| | - Po-Jen Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, 824410, Taiwan
- Graduate Institute of Medicine, I-Shou University, Kaohsiung, 824410, Taiwan
| | - Ahmed K ElHady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy & Biotechnology, German University in Cairo, Cairo, 11835, Egypt
- School of Life & Medical Sciences, University of Hertfordshire hosted by Global Academic Foundation, New Administrative Capital, Cairo, Egypt
| | - Shun-Hua Chen
- School of Nursing, Fooyin University, Kaohsiung, 831301, Taiwan
| | - Hsin-Chieh Lin
- Department of Chinese Medicine, E-Da Cancer Hospital, Kaohsiung, 824410, Taiwan
| | - Dalia S El-Gamil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy & Biotechnology, German University in Cairo, Cairo, 11835, Egypt
- Department of Chemistry, Faculty of Pharmacy, Ahram Canadian University, Cairo, 12451, Egypt
| | - Youssef Aboushady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy & Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy & Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| | - Matthias Engel
- Pharmaceutical & Medicinal Chemistry, Saarland University, Campus C2.3, D-66123, Saarbrücken, Germany
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy & Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| |
Collapse
|
8
|
Kokkorakis N, Zouridakis M, Gaitanou M. Mirk/Dyrk1B Kinase Inhibitors in Targeted Cancer Therapy. Pharmaceutics 2024; 16:528. [PMID: 38675189 PMCID: PMC11053710 DOI: 10.3390/pharmaceutics16040528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
During the last years, there has been an increased effort in the discovery of selective and potent kinase inhibitors for targeted cancer therapy. Kinase inhibitors exhibit less toxicity compared to conventional chemotherapy, and several have entered the market. Mirk/Dyrk1B kinase is a promising pharmacological target in cancer since it is overexpressed in many tumors, and its overexpression is correlated with patients' poor prognosis. Mirk/Dyrk1B acts as a negative cell cycle regulator, maintaining the survival of quiescent cancer cells and conferring their resistance to chemotherapies. Many studies have demonstrated the valuable therapeutic effect of Mirk/Dyrk1B inhibitors in cancer cell lines, mouse xenografts, and patient-derived 3D-organoids, providing a perspective for entering clinical trials. Since the majority of Mirk/Dyrk1B inhibitors target the highly conserved ATP-binding site, they exhibit off-target effects with other kinases, especially with the highly similar Dyrk1A. In this review, apart from summarizing the data establishing Dyrk1B as a therapeutic target in cancer, we highlight the most potent Mirk/Dyrk1B inhibitors recently reported. We also discuss the limitations and perspectives for the structure-based design of Mirk/Dyrk1B potent and highly selective inhibitors based on the accumulated structural data of Dyrk1A and the recent crystal structure of Dyrk1B with AZ191 inhibitor.
Collapse
Affiliation(s)
- Nikolaos Kokkorakis
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, 11521 Athens, Greece;
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Marios Zouridakis
- Structural Neurobiology Research Group, Laboratory of Molecular Neurobiology and Immunology, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Maria Gaitanou
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, 11521 Athens, Greece;
| |
Collapse
|
9
|
Zhang S, Liu L, Shi S, He H, Shen Q, Wang H, Qin S, Chang J, Zhong R. Bidirectional Association Between Cardiovascular Disease and Lung Cancer in a Prospective Cohort Study. J Thorac Oncol 2024; 19:80-93. [PMID: 37703998 DOI: 10.1016/j.jtho.2023.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 09/01/2023] [Accepted: 09/01/2023] [Indexed: 09/15/2023]
Abstract
INTRODUCTION The study aimed to prospectively investigate the bidirectional association between cardiovascular disease (CVD) and lung cancer, and whether this association differs across genetic risk levels. METHODS This study prospectively followed 455,804 participants from the United Kingdom Biobank cohort who were free of lung cancer at baseline. Cox proportional hazard models were used to estimate the hazard ratio (HR) for incident lung cancer according to CVD status. In parallel, similar approaches were used to assess the risk of incident CVD according to lung cancer status among 478,756 participants free of CVD at baseline. The bidirectional causal relations between these conditions were assessed using Mendelian randomization analysis. Besides, polygenic risk scores were estimated by integrating genome-wide association studies identified risk variants. RESULTS During 4,007,477 person-years of follow-up, 2006 incident lung cancer cases were documented. Compared with participants without CVD, those with CVD had HRs (95% confidence interval [CI]) of 1.49 (1.30-1.71) for NSCLC, 1.80 (1.39-2.34) for lung squamous cell carcinoma (LUSC), and 1.25 (1.01-1.56) for lung adenocarcinoma (LUAD). After stratification by smoking status, significant associations of CVD with lung cancer risk were observed in former smokers (HR = 1.44, 95% CI: 1.20-1.74) and current smokers (HR = 1.38, 95% CI: 1.13-1.69), but not in never-smokers (HR = 0.98, 95% CI: 0.60-1.61). In addition, CVD was associated with lung cancer risk across each genetic risk level (pheterogeneity = 0.336). In the second analysis, 32,974 incident CVD cases were recorded. Compared with those without lung cancer, the HRs (95% CI) for CVD were 2.33 (1.29-4.21) in NSCLC, 3.66 (1.65-8.14) in LUAD, and 1.98 (0.64-6.14) in LUSC. In particular, participants with lung cancer had a high risk of incident CVD at a high genetic risk level (HR = 3.79, 95% CI: 1.57-9.13). No causal relations between these conditions were observed in Mendelian randomization analysis. CONCLUSIONS CVD is associated with an increased risk of NSCLC including LUSC and LUAD. NSCLC, particularly LUAD, is associated with a higher CVD risk. Awareness of this bidirectional association may improve prevention and treatment strategies for both diseases. Future clinical demands will require a greater focus on cardiac oncology.
Collapse
Affiliation(s)
- Shanshan Zhang
- Department of Epidemiology and Biostatistics and Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lei Liu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shanshan Shi
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Heng He
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, People's Republic of China
| | - Qian Shen
- Department of Epidemiology and Biostatistics and Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Haoxue Wang
- Department of Epidemiology and Biostatistics and Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shifan Qin
- Department of Epidemiology and Biostatistics and Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jiang Chang
- Department of Epidemiology and Biostatistics and Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Rong Zhong
- Department of Epidemiology and Biostatistics and Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
10
|
Bhat Y, Thrishna MR, Banerjee S. Molecular targets and therapeutic strategies for triple-negative breast cancer. Mol Biol Rep 2023; 50:10535-10577. [PMID: 37924450 DOI: 10.1007/s11033-023-08868-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/29/2023] [Indexed: 11/06/2023]
Abstract
Triple-negative breast cancer (TNBC) is known for its heterogeneous complexity and is often difficult to treat. TNBC lacks the expression of major hormonal receptors like estrogen receptor, progesterone receptor, and human epidermal growth factor receptor-2 and is further subdivided into androgen receptor (AR) positive and AR negative. In contrast, AR negative is also known as quadruple-negative breast cancer (QNBC). Compared to AR-positive TNBC, QNBC has a great scarcity of prognostic biomarkers and therapeutic targets. QNBC shows excessive cellular growth and proliferation of tumor cells due to increased expression of growth factors like EGF and various surface proteins. This study briefly reviews the limited data available as protein biomarkers that can be used as molecular targets in treating TNBC as well as QNBC. Targeted therapy and immune checkpoint inhibitors have recently changed cancer treatment. Many studies in medicinal chemistry continue to focus on the synthesis of novel compounds to discover new antiproliferative medicines capable of treating TNBC despite the abundance of treatments currently on the market. Drug repurposing is one of the therapeutic methods for TNBC that has been examined. Moreover, some additional micronutrients, nutraceuticals, and functional foods may be able to lower cancer risk or slow the spread of malignant diseases that have already been diagnosed with cancer. Finally, nanomedicines, or applications of nanotechnology in medicine, introduce nanoparticles with variable chemistry and architecture for the treatment of cancer. This review emphasizes the most recent research on nutraceuticals, medication repositioning, and novel therapeutic strategies for the treatment of TNBC.
Collapse
Affiliation(s)
- Yashasvi Bhat
- School of Bio Science and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - M R Thrishna
- School of Bio Science and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Satarupa Banerjee
- School of Bio Science and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
11
|
Qiao R, Zhu Q, Di F, Liu C, Song Y, Zhang J, Xu T, Wang Y, Dai L, Gu W, Han B, Yang R. Hypomethylation of DYRK4 in peripheral blood is associated with increased lung cancer risk. Mol Carcinog 2023; 62:1745-1754. [PMID: 37530470 DOI: 10.1002/mc.23612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 06/01/2023] [Accepted: 07/19/2023] [Indexed: 08/03/2023]
Abstract
Lung cancer (LC) is the leading cause of cancer-related deaths worldwide. It is urgent to identify new biomarkers for the early detection of LC. DNA methylation in peripheral blood has been reported to be associated with cancers. We conducted two independent case-control studies and a nested case-control study (168 LC cases and 167 controls in study Ⅰ, 677 LC cases and 833 controls in study Ⅱ, 147 precancers and 21 controls in the nested case-control study). The methylation levels of DYRK4 CpG sites were measured using mass spectrometry and their correlations with LC were analyzed by logistic regression and nonparametric tests. Bonferroni correction was used for the multiple comparisons. LC-related decreased DYRK4 methylation was discovered in Study I and validated in Study II (the odds ratios [ORs] for the lowest vs. highest quartile of all three DYRK4 CpG sites ranged from 1.64 to 2.09, all p < 0.001). Combining the two studies, hypomethylation of DYRK4 was observed in stage I cases (ORs per -10% methylation ranged from 1.16 to 1.38, all p < 5.9E-04), and could be enhanced by male gender (ORs ranged from 1.77 to 4.17 via interquartile analyses, all p < 0.017). Hypomethylation of DYRK4_A_CpG_2 was significantly correlated with tumor size, length, and stage (p = 0.034, 0.002, and 0.002, respectively) in LC cases. Our study disclosed the association between DYRK4 hypomethylation in peripheral blood and LC, suggesting the feasibility of blood-based DNA methylation as new biomarker for LC detection.
Collapse
Affiliation(s)
- Rong Qiao
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Qiang Zhu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Feifei Di
- Nanjing TANTICA Biotechnology Co. Ltd., Nanjing, China
| | - Chunlan Liu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yakang Song
- Nanjing TANTICA Biotechnology Co. Ltd., Nanjing, China
| | - Jin Zhang
- Nanjing TANTICA Biotechnology Co. Ltd., Nanjing, China
| | - Tian Xu
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yue Wang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Wanjian Gu
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Baohui Han
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Rongxi Yang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
- Nanjing TANTICA Biotechnology Co. Ltd., Nanjing, China
| |
Collapse
|
12
|
Wongchang T, Pluangnooch P, Hongeng S, Wongkajornsilp A, Thumkeo D, Soontrapa K. Inhibition of DYRK1B suppresses inflammation in allergic contact dermatitis model and Th1/Th17 immune response. Sci Rep 2023; 13:7058. [PMID: 37120440 PMCID: PMC10148813 DOI: 10.1038/s41598-023-34211-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 04/26/2023] [Indexed: 05/01/2023] Open
Abstract
Allergic contact dermatitis (ACD) is a type IV hypersensitivity mainly mediated by Th1/Th17 immune response. Topical corticosteroid is currently the first-line treatment for allergic contact dermatitis (ACD) and systemic administration of immunosuppressive drugs are used in patients with severe disseminated cases. However, increased risk of adverse effects has limited their use. Thus, the development of a novel immunosuppressant for ACD with low toxicity is a challenging issue. In this study, we began our study by using a murine contact hypersensitivity (CHS) model of ACD to examine the immunosuppressive effects of DYRK1B inhibition. We found that mice treated with a selective DYRK1B inhibitor show reduced ear inflammation. In addition, a significant reduction of Th1 and Th17 cells in the regional lymph node upon DYRK1B inhibition was observed by FACS analysis. Studies in vitro further revealed that DYRK1B inhibitor does not only suppressed Th1 and Th17 differentiation, but also promotes regulatory T cells (Treg) differentiation. Mechanistically, FOXO1 signaling was enhanced due to the suppression of FOXO1Ser329 phosphorylation in the presence of DYRK1B inhibitor. Therefore, these findings suggest that DYRK1B regulates CD4 T cell differentiation through FOXO1 phosphorylation and DYRK1B inhibitor has a potential as a novel agent for treatment of ACD.
Collapse
Affiliation(s)
- Thamrong Wongchang
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand
- Division of Pharmacology, Department of Pharmaceutical Care, School of Pharmaceutical Sciences, University of Phayao, Phayao, Thailand
| | - Panwadee Pluangnooch
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Excellent Center for Drug Discovery, Mahidol University, Bangkok, Thailand
| | - Adisak Wongkajornsilp
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand
| | - Dean Thumkeo
- Department of Drug Discovery Medicine, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kitipong Soontrapa
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand.
| |
Collapse
|
13
|
Armanmehr A, Jafari Khamirani H, Zoghi S, Dianatpour M. Analysis of DYRK1B, PPARG, and CEBPB Expression Patterns in Adipose-Derived Stem Cells from Patients Carrying DYRK1B R102C and Healthy Individuals During Adipogenesis. Metab Syndr Relat Disord 2022; 20:576-583. [PMID: 36318489 DOI: 10.1089/met.2021.0140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background: Metabolic syndrome (MetS) is a group of signs and symptoms that are associated with a higher risk of type 2 diabetes mellitus and cardiovascular diseases. The major risk factor for developing MetS is abdominal obesity, which is caused by an increase in adipocyte size or quantity. Increased adipocyte quantity is a result of differentiation of stem cells into adipose tissue. Numerous studies have investigated the expression of key transcription factors, including PPARG and CEBPB during adipocyte differentiation in murine cells such as 3T3-L1 cell lines. To better understand the expression changes during the process of fat accumulation in adipose-derived stem cells (ASCs), we compared the expression of DYRK1B, PPARG, and ẟB in ASCs between the patient (harboring DYRK1B R102C) and control (healthy individuals) groups. Methods: Gene expression was evaluated on the eighth day before induction and days 1, 5, and 15 postinduction. The pluripotent capacity of ASCs and the potential for differentiation into adipocytes were confirmed by flow cytometry analysis of surface markers (CD34, CD44, CD105, and CD90), and Oil Red O staining, respectively. The Expression of DYRK1B, PPARG, and CEBPB were assessed by real-time-polymerase chain reaction in patients and normal individuals. The effects of AZ191, a potent small molecule inhibitor on DYRK1B and CEBPB expression in patients' samples were studied. Result: The expression of DYRK1B kinase and transcription factors (CEBPB and PPARG) are higher in ASCs harboring DYRK1B R102C compared with noncarriers on days 5 and 15 during adipocyte differentiation. These proteins may be helpful to elucidate the mechanisms underlying obesity and obesity-related disorders like MetS. Furthermore, the new compound AZ191 exhibited inhibitory activity toward DYRK1B and CEBPB. We suggest that AZ191 may be helpful in defining the potential roles of DYRK1B and CEBPB in adipogenesis.
Collapse
Affiliation(s)
- Azam Armanmehr
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Jafari Khamirani
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran.,Comprehensive Medical Genetic Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Zoghi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran.,Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
14
|
Zhao L, Xiong X, Liu L, Liang Q, Tong R, Feng X, Bai L, Shi J. Recent research and development of DYRK1A inhibitors. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
15
|
Zhuang L, Jia K, Chen C, Li Z, Zhao J, Hu J, Zhang H, Fan Q, Huang C, Xie H, Lu L, Shen W, Ning G, Wang J, Zhang R, Chen K, Yan X. DYRK1B-STAT3 Drives Cardiac Hypertrophy and Heart Failure by Impairing Mitochondrial Bioenergetics. Circulation 2022; 145:829-846. [PMID: 35235343 DOI: 10.1161/circulationaha.121.055727] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Heart failure is a global public health issue that is associated with increasing morbidity and mortality. Previous studies have suggested that mitochondrial dysfunction plays critical roles in the progression of heart failure; however, the underlying mechanisms remain unclear. Because kinases have been reported to modulate mitochondrial function, we investigated the effects of DYRK1B (dual-specificity tyrosine-regulated kinase 1B) on mitochondrial bioenergetics, cardiac hypertrophy, and heart failure. METHODS We engineered DYRK1B transgenic and knockout mice and used transverse aortic constriction to produce an in vivo model of cardiac hypertrophy. The effects of DYRK1B and its downstream mediators were subsequently elucidated using RNA-sequencing analysis and mitochondrial functional analysis. RESULTS We found that DYRK1B expression was clearly upregulated in failing human myocardium and in hypertrophic murine hearts, as well. Cardiac-specific DYRK1B overexpression resulted in cardiac dysfunction accompanied by a decline in the left ventricular ejection fraction, fraction shortening, and increased cardiac fibrosis. In striking contrast to DYRK1B overexpression, the deletion of DYRK1B mitigated transverse aortic constriction-induced cardiac hypertrophy and heart failure. Mechanistically, DYRK1B was positively associated with impaired mitochondrial bioenergetics by directly binding with STAT3 to increase its phosphorylation and nuclear accumulation, ultimately contributing toward the downregulation of PGC-1α (peroxisome proliferator-activated receptor gamma coactivator-1α). Furthermore, the inhibition of DYRK1B or STAT3 activity using specific inhibitors was able to restore cardiac performance by rejuvenating mitochondrial bioenergetics. CONCLUSIONS Taken together, the findings of this study provide new insights into the previously unrecognized role of DYRK1B in mitochondrial bioenergetics and the progression of cardiac hypertrophy and heart failure. Consequently, these findings may provide new therapeutic options for patients with heart failure.
Collapse
Affiliation(s)
- Lingfang Zhuang
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Kangni Jia
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Chen Chen
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.C.)
| | - Zhigang Li
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jiaxin Zhao
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases (G.N., J.W.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jian Hu
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases (G.N., J.W.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hang Zhang
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases (G.N., J.W.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Qin Fan
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Chunkai Huang
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases (G.N., J.W.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hongyang Xie
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Lin Lu
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Weifeng Shen
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Guang Ning
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases (G.N., J.W.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jiqiu Wang
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases (G.N., J.W.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases (G.N., J.W.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Kang Chen
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Xiaoxiang Yan
- Department of Cardiovascular Medicine (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., K.C., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Ruijin Hospital, Institute of Cardiovascular Diseases (L.Z., K..J., Z.L., J.Z., J.H., H.Z., Q.F., C.H., H.X., L.L., W.S., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| |
Collapse
|
16
|
Kinome-Wide siRNA Screening Identifies DYRK1B as a Potential Therapeutic Target for Triple-Negative Breast Cancer Cells. Cancers (Basel) 2021; 13:cancers13225779. [PMID: 34830933 PMCID: PMC8616396 DOI: 10.3390/cancers13225779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/03/2021] [Accepted: 11/16/2021] [Indexed: 12/23/2022] Open
Abstract
Simple Summary Therapeutic target is limited for patients with triple-negative breast cancer (TNBC). Through kinome-wide siRNA (709 genes) screening, DYRK1B was identified as a potential gene essential for cell proliferation and mobility of TNBC cells, particularly in DYRK1B highly expressed TNBC cells. TNBC patients with high expression of DYRK1B had poor overall survival and disease-free survival. CCDC97 and ZNF581 were positively correlated with DYRK1B expression and might be involved in DYRK1B-mediated tumor malignancy in TNBC patients, providing DYRK1B as a potential theranostic target for TNBC. Abstract Aims: The selective molecules for targeted therapy of triple-negative breast cancer (TNBC) are limited. Several kinases play pivotal roles in cancer development and malignancy. The study aims to determine if any kinases confer to malignancy of TNBC cells, which could serve as a theranostic target for TNBC. Methods: Kinome siRNA library was used to screen selective genes required for the proliferation of TNBC cells. The involvement of DYRK1B in cancer malignancy was evaluated with migration, invasion assays, and spheroid culture. The expression of DYRK1B was confirmed with quantitative PCR and immunoblotting. The clinical correlation of DYRK1B in TNBC patients was examined with tissue microarray and The Cancer Genome Atlas (TCGA) database. Results: Our results showed that silencing DYRK1B significantly suppressed cell viability in DYRK1B-high expressed TNBC cells, likely by arresting the cell cycle at the G1 phase. Nevertheless, silencing DYRK1B had marginal effects on DYRK1B-low expressed TNBC cells. Similarly, the knockdown of DYRK1B decreased tumorsphere formation and increased cell death of the tumorsphere. Moreover, inactivation of DYRK1B by either specific inhibitor or ectopic expressing catalytic mutant of DYRK1B inhibited cell viability and metastatic characteristics, including migration and invasion. In addition, DYRK1B protein expression was elevated in tumor tissues compared to that in adjacent normal tissues of TNBC patients. Further, DYRK1B gene expression was highly correlated with CCDC97 or ZNF581 genes in TNBC cells and patients. High co-expression of DYRK1B with CCDC97 or ZNF581 was significantly associated with unfavorable overall survival and disease-free survival of TNBC patients. Conclusions: our results suggest DYRK1B might be essential for promoting tumor progression and could be a theranostic target for TNBC. Silencing or inactivation of DYRK1B might be a potential targeted therapy for TNBC.
Collapse
|
17
|
Sato K, Padgaonkar AA, Baker SJ, Cosenza SC, Rechkoblit O, Subbaiah DRCV, Domingo-Domenech J, Bartkowski A, Port ER, Aggarwal AK, Ramana Reddy MV, Irie HY, Reddy EP. Simultaneous CK2/TNIK/DYRK1 inhibition by 108600 suppresses triple negative breast cancer stem cells and chemotherapy-resistant disease. Nat Commun 2021; 12:4671. [PMID: 34344863 PMCID: PMC8333338 DOI: 10.1038/s41467-021-24878-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 07/08/2021] [Indexed: 02/07/2023] Open
Abstract
Triple negative breast cancer (TNBC) remains challenging because of heterogeneous responses to chemotherapy. Incomplete response is associated with a greater risk of metastatic progression. Therefore, treatments that target chemotherapy-resistant TNBC and enhance chemosensitivity would improve outcomes for these high-risk patients. Breast cancer stem cell-like cells (BCSCs) have been proposed to represent a chemotherapy-resistant subpopulation responsible for tumor initiation, progression and metastases. Targeting this population could lead to improved TNBC disease control. Here, we describe a novel multi-kinase inhibitor, 108600, that targets the TNBC BCSC population. 108600 treatment suppresses growth, colony and mammosphere forming capacity of BCSCs and induces G2M arrest and apoptosis of TNBC cells. In vivo, 108600 treatment of mice bearing triple negative tumors results in the induction of apoptosis and overcomes chemotherapy resistance. Finally, treatment with 108600 and chemotherapy suppresses growth of pre-established TNBC metastases, providing additional support for the clinical translation of this agent to clinical trials.
Collapse
Affiliation(s)
- Katsutoshi Sato
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amol A Padgaonkar
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stacey J Baker
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephen C Cosenza
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Olga Rechkoblit
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D R C Venkata Subbaiah
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Alison Bartkowski
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elisa R Port
- Department of Surgery, Mount Sinai Hospital, New York, NY, USA
| | - Aneel K Aggarwal
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M V Ramana Reddy
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hanna Y Irie
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - E Premkumar Reddy
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
18
|
Lee Walmsley D, Murray JB, Dokurno P, Massey AJ, Benwell K, Fiumana A, Foloppe N, Ray S, Smith J, Surgenor AE, Edmonds T, Demarles D, Burbridge M, Cruzalegui F, Kotschy A, Hubbard RE. Fragment-Derived Selective Inhibitors of Dual-Specificity Kinases DYRK1A and DYRK1B. J Med Chem 2021; 64:8971-8991. [PMID: 34143631 DOI: 10.1021/acs.jmedchem.1c00024] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The serine/threonine kinase DYRK1A has been implicated in regulation of a variety of cellular processes associated with cancer progression, including cell cycle control, DNA damage repair, protection from apoptosis, cell differentiation, and metastasis. In addition, elevated-level DYRK1A activity has been associated with increased severity of symptoms in Down's syndrome. A selective inhibitor of DYRK1A could therefore be of therapeutic benefit. We have used fragment and structure-based discovery methods to identify a highly selective, well-tolerated, brain-penetrant DYRK1A inhibitor which showed in vivo activity in a tumor model. The inhibitor provides a useful tool compound for further exploration of the effect of DYRK1A inhibition in models of disease.
Collapse
Affiliation(s)
| | - James B Murray
- Vernalis (R&D) Ltd., Granta Park, Cambridge CB21 6GB, U.K
| | - Pawel Dokurno
- Vernalis (R&D) Ltd., Granta Park, Cambridge CB21 6GB, U.K
| | | | - Karen Benwell
- Vernalis (R&D) Ltd., Granta Park, Cambridge CB21 6GB, U.K
| | - Andrea Fiumana
- Vernalis (R&D) Ltd., Granta Park, Cambridge CB21 6GB, U.K
| | | | - Stuart Ray
- Vernalis (R&D) Ltd., Granta Park, Cambridge CB21 6GB, U.K
| | - Julia Smith
- Vernalis (R&D) Ltd., Granta Park, Cambridge CB21 6GB, U.K
| | | | - Thomas Edmonds
- Institut de Recherches Servier, 125 Chemin de Ronde, Croissy-sur-Seine 78290, France
| | - Didier Demarles
- Technologie Servier, 27 Rue Eugène Vignat, Orleans 45000, France
| | - Mike Burbridge
- Institut de Recherches Servier, 125 Chemin de Ronde, Croissy-sur-Seine 78290, France
| | - Francisco Cruzalegui
- Institut de Recherches Servier, 125 Chemin de Ronde, Croissy-sur-Seine 78290, France
| | - Andras Kotschy
- Servier Research Institute of Medicinal Chemistry, Záhony u. 7., Budapest H-1031, Hungary
| | | |
Collapse
|
19
|
Mohamed YA, Hassaneen HM, El-Dessouky MA, Safwat G, Hassan NAM, Amr K. Study of DYRK1B gene expression and its association with metabolic syndrome in a small cohort of Egyptians. Mol Biol Rep 2021; 48:5497-5502. [PMID: 34291393 DOI: 10.1007/s11033-021-06560-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 07/12/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND A cluster of many risk factors for type 2 diabetes and cardiovascular disease is used to describe the metabolic syndrome (MetS). Moreover, genetic differences associated with metabolic syndrome play a key role in its prevalence and side effects. This study aims to investigate the expression of DYRK1B and its association with metabolic syndrome in a small cohort of Egyptian. MATERIALS AND METHODS A total of 100 adult Egyptians (50 with MetS and 50 healthy control subjects) were included to this study. Clinical, biochemical and anthropometric analysis were assessed. Relative gene expressions of DYRK1B were compared between two groups of subjects using real time PCR. RESULTS We observed marked overexpression in DYRK1B (p < 0.05) in MetS subjects when compared with the healthy control subjects. CONCLUSION This is the first study to provide evidence that DYRK1B is highly expressed among the MetS subjects.
Collapse
Affiliation(s)
- Yara Ahmed Mohamed
- Faculty of Biotechnology, October University for Modern Sciences and Arts University (MSA), No. 12567, 54 Anwar El-Sadat street, Al-Haram, Giza, Egypt.
| | - H M Hassaneen
- Faculty of Science, Chemistry Department, Cairo University, Giza, Egypt
| | | | - Gehan Safwat
- Faculty of Biotechnology, October University for Modern Sciences and Arts University (MSA), No. 12567, 54 Anwar El-Sadat street, Al-Haram, Giza, Egypt
| | - Naglaa Abu-Mandil Hassan
- Medical Research Division, Biological Anthropology Department, National Research Centre, Giza, Egypt
| | - Khalda Amr
- Human Genetics and Genome Research Division, Medical Molecular Genetics Department, National Research Centre, Giza, Egypt
| |
Collapse
|
20
|
Li L, Wei JR, Song Y, Fang S, Du Y, Li Z, Zeng TT, Zhu YH, Li Y, Guan XY. TROAP switches DYRK1 activity to drive hepatocellular carcinoma progression. Cell Death Dis 2021; 12:125. [PMID: 33500384 PMCID: PMC7838256 DOI: 10.1038/s41419-021-03422-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the common malignancy and lacks effective therapeutic targets. Here, we demonstrated that ectopic expression of trophinin-associated protein (TROAP) dramatically drove HCC cell growth assessed by foci formation in monolayer culture, colony formation in soft agar and orthotopic liver transplantation in nude mice. Inversely, silencing TROAP expression with short-hairpin RNA attenuated the malignant proliferation of HCC cells in vitro and in vivo. Next, mechanistic investigation revealed that TROAP directly bound to dual specificity tyrosine phosphorylation regulated kinase 1A/B (DYRK1A/B), resulting in the cytoplasmic retention of proteins DYRK1A/B and promoting cell cycle process via activation of Akt/GSK-3β signaling. Combination of cisplatin with an inhibitor of DYRK1 AZ191 effectively inhibited tumor growth in mouse model for HCC cells with high level of TROAP. Clinically, TROAP was significantly upregulated by miR-142-5p in HCC tissues, which predicted the poor survival of patients with HCC. Therefore, TROAP/DYRK1/Akt axis may be a promising therapeutic target and prognostic indicator for patients with HCC.
Collapse
Affiliation(s)
- Lei Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.
- Department of Clinical Oncology, State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China.
- Department of Clinical Oncology Center, The University of Hongkong-Shenzhen Hospital, 518053, Shenzhen, China.
| | - Jia-Ru Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 510060, Guangzhou, China
| | - Ye Song
- Affiliated Cancer Hospital & Institutes of Guangzhou Medical University, Guangzhou Key Medical Discipline Construction Project, 510095, Guangzhou, China
| | - Shuo Fang
- The Seventh Affiliated Hospital, Sun Yat-sen University, 518100, Shenzhen, China
| | - Yanyu Du
- The Seventh Affiliated Hospital, Sun Yat-sen University, 518100, Shenzhen, China
| | - Zhuo Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China
| | - Ting-Ting Zeng
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China
| | - Ying-Hui Zhu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China
| | - Yan Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China
| | - Xin-Yuan Guan
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.
- Department of Clinical Oncology, State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China.
- Department of Clinical Oncology Center, The University of Hongkong-Shenzhen Hospital, 518053, Shenzhen, China.
| |
Collapse
|
21
|
Kokkorakis N, Gaitanou M. Minibrain-related kinase/dual-specificity tyrosine-regulated kinase 1B implication in stem/cancer stem cells biology. World J Stem Cells 2020; 12:1553-1575. [PMID: 33505600 PMCID: PMC7789127 DOI: 10.4252/wjsc.v12.i12.1553] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/29/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
Dual-specificity tyrosine phosphorylation-regulated kinase 1B (DYRK1B), also known as minibrain-related kinase (MIRK) is one of the best functionally studied members of the DYRK kinase family. DYRKs comprise a family of protein kinases that are emerging modulators of signal transduction pathways, cell proliferation and differentiation, survival, and cell motility. DYRKs were found to participate in several signaling pathways critical for development and cell homeostasis. In this review, we focus on the DYRK1B protein kinase from a functional point of view concerning the signaling pathways through which DYRK1B exerts its cell type-dependent function in a positive or negative manner, in development and human diseases. In particular, we focus on the physiological role of DYRK1B in behavior of stem cells in myogenesis, adipogenesis, spermatogenesis and neurogenesis, as well as in its pathological implication in cancer and metabolic syndrome. Thus, understanding of the molecular mechanisms that regulate signaling pathways is of high importance. Recent studies have identified a close regulatory connection between DYRK1B and the hedgehog (HH) signaling pathway. Here, we aim to bring together what is known about the functional integration and cross-talk between DYRK1B and several signaling pathways, such as HH, RAS and PI3K/mTOR/AKT, as well as how this might affect cellular and molecular processes in development, physiology, and pathology. Thus, this review summarizes the major known functions of DYRK1B kinase, as well as the mechanisms by which DYRK1B exerts its functions in development and human diseases focusing on the homeostasis of stem and cancer stem cells.
Collapse
Affiliation(s)
- Nikolaos Kokkorakis
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens 11521, Greece
| | - Maria Gaitanou
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens 11521, Greece.
| |
Collapse
|
22
|
Boni J, Rubio-Perez C, López-Bigas N, Fillat C, de la Luna S. The DYRK Family of Kinases in Cancer: Molecular Functions and Therapeutic Opportunities. Cancers (Basel) 2020; 12:cancers12082106. [PMID: 32751160 PMCID: PMC7465136 DOI: 10.3390/cancers12082106] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022] Open
Abstract
DYRK (dual-specificity tyrosine-regulated kinases) are an evolutionary conserved family of protein kinases with members from yeast to humans. In humans, DYRKs are pleiotropic factors that phosphorylate a broad set of proteins involved in many different cellular processes. These include factors that have been associated with all the hallmarks of cancer, from genomic instability to increased proliferation and resistance, programmed cell death, or signaling pathways whose dysfunction is relevant to tumor onset and progression. In accordance with an involvement of DYRK kinases in the regulation of tumorigenic processes, an increasing number of research studies have been published in recent years showing either alterations of DYRK gene expression in tumor samples and/or providing evidence of DYRK-dependent mechanisms that contribute to tumor initiation and/or progression. In the present article, we will review the current understanding of the role of DYRK family members in cancer initiation and progression, providing an overview of the small molecules that act as DYRK inhibitors and discussing the clinical implications and therapeutic opportunities currently available.
Collapse
Affiliation(s)
- Jacopo Boni
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Dr Aiguader 88, 08003 Barcelona, Spain;
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Carlota Rubio-Perez
- Cancer Science Programme, Institute for Research in Biomedicine (IRB), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain; (C.R.-P.); (N.L.-B.)
| | - Nuria López-Bigas
- Cancer Science Programme, Institute for Research in Biomedicine (IRB), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain; (C.R.-P.); (N.L.-B.)
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig Lluís Companys 23, 08010 Barcelona, Spain
| | - Cristina Fillat
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), 28029 Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain;
| | - Susana de la Luna
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Dr Aiguader 88, 08003 Barcelona, Spain;
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), 28029 Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig Lluís Companys 23, 08010 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Dr Aiguader 88, 08003 Barcelona, Spain
- Correspondence: ; Tel.: +34-933-160-144
| |
Collapse
|
23
|
Szamborska-Gbur A, Rutkowska E, Dreas A, Frid M, Vilenchik M, Milik M, Brzózka K, Król M. How to design potent and selective DYRK1B inhibitors? Molecular modeling study. J Mol Model 2019; 25:41. [PMID: 30673861 DOI: 10.1007/s00894-018-3921-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 12/26/2018] [Indexed: 12/19/2022]
Abstract
DYRK1B protein kinase is an emerging anticancer target due to its overexpression in a variety of cancers and its role in cancer chemoresistance through maintaining cancer cells in the G0 (quiescent) state. Consequently, there is a growing interest in the development of potent and selective DYRK1B inhibitors for anticancer therapy. One of the major off-targets is another protein kinase, GSK3β, which phosphorylates an important regulator of cell cycle progression on the same residue as DYRK1B and is involved in multiple signaling pathways. In the current work, we performed a detailed comparative structural analysis of DYRK1B and GSK3β ATP-binding sites and identified key regions responsible for selectivity. As the crystal structure of DYRK1B has never been reported, we built and optimized a homology model by comparative modeling and metadynamics simulations. Calculation of interaction energies between docked ligands in the ATP-binding sites of both kinases allowed us to pinpoint key residues responsible for potency and selectivity. Specifically, the role of the gatekeeper residues in DYRK1B and GSK3β is discussed in detail, and two other residues are identified as key to selectivity of DYRK1B inhibition versus GSK3β. The analysis presented in this work was used to support the design of potent and selective azaindole-quinoline-based DYRK1B inhibitors and can facilitate development of more selective inhibitors for DYRK kinases.
Collapse
Affiliation(s)
| | | | | | - Michael Frid
- Felicitex Therapeutics, Inc., 27 Strathmore Road, Natick, MA, 01760, USA
| | - Maria Vilenchik
- Felicitex Therapeutics, Inc., 27 Strathmore Road, Natick, MA, 01760, USA
| | - Mariusz Milik
- Selvita S.A., Bobrzyńskiego 14, 30-348, Kraków, Poland
| | | | - Marcin Król
- Selvita S.A., Bobrzyńskiego 14, 30-348, Kraków, Poland.
| |
Collapse
|
24
|
He M, Gu J, Zhu J, Wang X, Wang C, Duan C, Ni Y, Lu X, Li J. Up-regulation of Dyrk1b promote astrocyte activation following lipopolysaccharide-induced neuroinflammation. Neuropeptides 2018; 69:76-83. [PMID: 29751999 DOI: 10.1016/j.npep.2018.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 02/11/2018] [Accepted: 04/15/2018] [Indexed: 02/07/2023]
Abstract
Astrocytes become activated in response to different stimulation. Dyrk1b is an arginine-directed serine/threonineprotein kinase that is expressed at elevated levels in many cancers but remains unknown in the pathologies of neuroinflammation. In this study, in vivo, we demonstrated that Dyrk1b expression was significantly increased and reached a peak at 12 h after LPS injection via Western blot. Double immunofluorescence staining showed that Dyrk1b co-located with GFAP and Ki67. In vitro, the expression of Dyrk1b, Ki67 and cyclinD1 was gradually increased and reached a peak at 12 h in a time-dependent manner after 1 μg/mL LPS stimulation. Knockdown of Dyrk1b significantly reduced the expression of Ki67 and cyclinD1. In addition, the data exhibited that silenced Dyrk1b decreased the expression of p-STAT3 in primary astrocyte cells, and Dyrk1b interacted with STAT3 in LPS-induced neuroinflammation. In conclusion, these results suggested that Dyrk1b is increased and may play a crucial role in regulating astrocyte cell activation via interact with STAT3 in LPS-induced neuroinflammation.
Collapse
Affiliation(s)
- Mingqing He
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing 210011, Jiangsu, China
| | - Jun Gu
- Department of Orthopaedics, XiShan People's Hospital, Wuxi 214011, Jiangsu, China
| | - Jinzhou Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Xiaoyan Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Chengniu Wang
- Basic Medical Research Centre, Medical College, Nantong University, Nantong 226001,Jiangsu, China
| | - Chengwei Duan
- The Second People's Hospital of Nantong, Nantong 226002, Jiangsu, China
| | - Yingjie Ni
- Department of Orthopaedics, XiShan People's Hospital, Wuxi 214011, Jiangsu, China
| | - Xiang Lu
- Department of Geriatrics, The Affiliated Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing 210011, Jiangsu, China.
| | - Jianzhong Li
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China.
| |
Collapse
|
25
|
Pérez-Sánchez G, Jiménez A, Quezada-Ramírez MA, Estudillo E, Ayala-Sarmiento AE, Mendoza-Hernández G, Hernández-Soto J, Hernández-Hernández FC, Cázares-Raga FE, Segovia J. Annexin A1, Annexin A2, and Dyrk 1B are upregulated during GAS1-induced cell cycle arrest. J Cell Physiol 2018; 233:4166-4182. [PMID: 29030970 DOI: 10.1002/jcp.26226] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 10/03/2017] [Indexed: 12/18/2022]
Abstract
GAS1 is a pleiotropic protein that has been investigated because of its ability to induce cell proliferation, cell arrest, and apoptosis, depending on the cellular or the physiological context in which it is expressed. At this point, we have information about the molecular mechanisms by which GAS1 induces proliferation and apoptosis; but very few studies have been focused on elucidating the mechanisms by which GAS1 induces cell arrest. With the aim of expanding our knowledge on this subject, we first focused our research on finding proteins that were preferentially expressed in cells arrested by serum deprivation. By using a proteomics approach and mass spectrometry analysis, we identified 17 proteins in the 2-DE protein profile of serum deprived NIH3T3 cells. Among them, Annexin A1 (Anxa1), Annexin A2 (Anxa2), dual specificity tyrosine-phosphorylation-regulated kinase 1B (Dyrk1B), and Eukaryotic translation initiation factor 3, F (eIf3f) were upregulated at transcriptional the level in proliferative NIH3T3 cells. Moreover, we demonstrated that Anxa1, Anxa2, and Dyrk1b are upregulated at both the transcriptional and translational levels by the overexpression of GAS1. Thus, our results suggest that the upregulation of Anxa1, Anxa2, and Dyrk1b could be related to the ability of GAS1 to induce cell arrest and maintain cell viability. Finally, we provided further evidence showing that GAS1 through Dyrk 1B leads not only to the arrest of NIH3T3 cells but also maintains cell viability.
Collapse
Affiliation(s)
- Gilberto Pérez-Sánchez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Adriana Jiménez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Marco A Quezada-Ramírez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Enrique Estudillo
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Alberto E Ayala-Sarmiento
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | | | - Justino Hernández-Soto
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Fidel C Hernández-Hernández
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Febe E Cázares-Raga
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Jose Segovia
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| |
Collapse
|
26
|
Becker W. A wake-up call to quiescent cancer cells - potential use of DYRK1B inhibitors in cancer therapy. FEBS J 2018; 285:1203-1211. [PMID: 29193696 DOI: 10.1111/febs.14347] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/25/2017] [Accepted: 11/24/2017] [Indexed: 12/27/2022]
Abstract
Nondividing cancer cells are relatively resistant to chemotherapeutic drugs and environmental stress factors. Promoting cell cycle re-entry of quiescent cancer cells is a potential strategy to enhance the cytotoxicity of agents that target cycling cells. It is therefore important to elucidate the mechanisms by which these cells are maintained in the quiescent state. The protein kinase dual specificity tyrosine phosphorylation-regulated kinase 1B (DYRK1B) is overexpressed in a subset of cancers and maintains cellular quiescence by counteracting G0 /G1 -S phase transition. Specifically, DYRK1B controls the S phase checkpoint by stabilizing the cyclin-dependent kinase (CDK) inhibitor p27Kip1 and inducing the degradation of cyclin D. DYRK1B also stabilizes the DREAM complex that represses cell cycle gene expression in G0 arrested cells. In addition, DYRK1B enhances cell survival by upregulating antioxidant gene expression and reducing intracellular levels of reactive oxygen species (ROS). Substantial evidence indicates that depletion or inhibition of DYRK1B drives cell cycle re-entry and enhances apoptosis of those quiescent cancer cells with high expression of DYRK1B. Furthermore, small molecule DYRK1B inhibitors sensitize cells to the cytotoxic effects of anticancer drugs that target proliferating cells. These encouraging findings justify continued efforts to investigate the use of DYRK1B inhibitors to disrupt the quiescent state and overturn chemoresistance of noncycling cancer cells.
Collapse
Affiliation(s)
- Walter Becker
- Institute of Pharmacology and Toxicology, Medical Faculty of the RWTH Aachen University, Germany
| |
Collapse
|
27
|
Chen H, Shen J, Choy E, Hornicek FJ, Shan A, Duan Z. Targeting DYRK1B suppresses the proliferation and migration of liposarcoma cells. Oncotarget 2017; 9:13154-13166. [PMID: 29568347 PMCID: PMC5862568 DOI: 10.18632/oncotarget.22743] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/30/2017] [Indexed: 12/24/2022] Open
Abstract
Liposarcoma is a common subtype of soft tissue sarcoma and accounts for 20% of all sarcomas. Conventional chemotherapeutic agents have limited efficacy in liposarcoma patients. Expression and activation of serine/threonine-protein kinase dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1B (DYRK1B) is associated with growth and survival of many types of cancer cells. However, the role of DYRK1B in liposarcoma remains unknown. In this study, we investigated the functional and therapeutic relevance of DYRK1B in liposarcoma. Tissue microarray and immunohistochemistry analysis showed that higher expression levels of DYRK1B correlated with a worse prognosis. RNA interference-mediated knockdown of DYRK1B or targeting DYRK1B with the kinase inhibitor AZ191 inhibited liposarcoma cell growth, decreased cell motility, and induced apoptosis. Moreover, combined AZ191 with doxorubicin demonstrated an increased anti-cancer effect on liposarcoma cells. These findings suggest that DYRK1B is critical for the growth of liposarcoma cells. Targeting DYRK1B provides a new rationale for treatment of liposarcoma.
Collapse
Affiliation(s)
- Hua Chen
- Department of Emergency Surgery, ShenZhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, Guangdong Province, China, 518020.,Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Jacson Shen
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Edwin Choy
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Francis J Hornicek
- Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6902, USA
| | - Aijun Shan
- Department of Emergency Surgery, ShenZhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, Guangdong Province, China, 518020
| | - Zhenfeng Duan
- Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6902, USA
| |
Collapse
|
28
|
Singh R, Lauth M. Emerging Roles of DYRK Kinases in Embryogenesis and Hedgehog Pathway Control. J Dev Biol 2017; 5:E13. [PMID: 29615569 PMCID: PMC5831797 DOI: 10.3390/jdb5040013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 11/17/2017] [Accepted: 11/18/2017] [Indexed: 12/19/2022] Open
Abstract
Hedgehog (Hh)/GLI signaling is an important instructive cue in various processes during embryonic development, such as tissue patterning, stem cell maintenance, and cell differentiation. It also plays crucial roles in the development of many pediatric and adult malignancies. Understanding the molecular mechanisms of pathway regulation is therefore of high interest. Dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs) comprise a group of protein kinases which are emerging modulators of signal transduction, cell proliferation, survival, and cell differentiation. Work from the last years has identified a close regulatory connection between DYRKs and the Hh signaling system. In this manuscript, we outline the mechanistic influence of DYRK kinases on Hh signaling with a focus on the mammalian situation. We furthermore aim to bring together what is known about the functional consequences of a DYRK-Hh cross-talk and how this might affect cellular processes in development, physiology, and pathology.
Collapse
Affiliation(s)
- Rajeev Singh
- Philipps University Marburg, Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor and Immune Biology (ZTI), Hans-Meerwein-Str. 3, 35043 Marburg, Germany.
| | - Matthias Lauth
- Philipps University Marburg, Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor and Immune Biology (ZTI), Hans-Meerwein-Str. 3, 35043 Marburg, Germany.
| |
Collapse
|
29
|
Masoudkabir F, Sarrafzadegan N, Gotay C, Ignaszewski A, Krahn AD, Davis MK, Franco C, Mani A. Cardiovascular disease and cancer: Evidence for shared disease pathways and pharmacologic prevention. Atherosclerosis 2017; 263:343-351. [PMID: 28624099 PMCID: PMC6207942 DOI: 10.1016/j.atherosclerosis.2017.06.001] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 05/08/2017] [Accepted: 06/01/2017] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease (CVD) and cancer are leading causes of mortality and morbidity worldwide. Strategies to improve their treatment and prevention are global priorities and major focus of World Health Organization's joint prevention programs. Emerging evidence suggests that modifiable risk factors including diet, sedentary lifestyle, obesity and tobacco use are central to the pathogenesis of both diseases and are reflected in common genetic, cellular, and signaling mechanisms. Understanding this important biological overlap is critical and may help identify novel therapeutic and preventative strategies for both disorders. In this review, we will discuss the shared genetic and molecular factors central to CVD and cancer and how the strategies commonly used for the prevention of atherosclerotic vascular disease can be applied to cancer prevention.
Collapse
Affiliation(s)
- Farzad Masoudkabir
- Cardiac Primary Prevention Research Center, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nizal Sarrafzadegan
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran; School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Carolyn Gotay
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada; Cancer Control Research Program, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Andrew Ignaszewski
- Division of Cardiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew D Krahn
- Division of Cardiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Margot K Davis
- Division of Cardiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher Franco
- Division of Cardiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Arya Mani
- Yale Cardiovascular Genetics Program, Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
30
|
Chen Y, Wang S, He Z, Sun F, Huang Y, Ni Q, Wang H, Wang Y, Cheng C. Dyrk1B overexpression is associated with breast cancer growth and a poor prognosis. Hum Pathol 2017; 66:48-58. [PMID: 28554575 DOI: 10.1016/j.humpath.2017.02.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 01/29/2017] [Accepted: 02/23/2017] [Indexed: 01/12/2023]
Abstract
Dyrk1B, also called minibrain-related kinase (Mirk), is a member of the dual-specificity tyrosine phosphorylation-regulated kinase (Dyrk)/minibrain family of dual-specificity protein kinases. It is a serine/threonine kinase involved in the regulation of tumor progression and cell proliferation. In this study, the role of Dyrk1B in breast cancer development was investigated. The expression of Dyrk1B was detected by Western blot and immunohistochemistry staining, both of which demonstrated that Dyrk1B was overexpressed in breast cancer tissues and cells. Statistical analysis showed that the extent of Dyrk1B expression was associated with multiple clinicopathologic factors, including tumor size, grade, estrogen receptor status, and Ki-67 expression, and that high expression predicted a poor prognosis. The growth of breast cancer cells was inhibited significantly after knockout of DYRK1B by small interfering RNA (siRNA). Moreover, FoxO1 could be phosphorylated by Dyrk1B, and then FoxO1 was shuttled from the cell nucleus into the cytoplasm, which might be the mechanism of Dyrk1B-mediated survival in breast cancer cells. The results suggest that Dyrk1B plays a key role in the progression of breast cancer and provides a new target for breast cancer therapy.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Immunology, Medical College, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China; Department of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Shuo Wang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Zhixian He
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Fulan Sun
- Department of General Surgery, The Second People's Hospital of Nantong, Nantong 226001, China
| | - Yeqing Huang
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Qichao Ni
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Hua Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Yingying Wang
- Department of Immunology, Medical College, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China.
| | - Chun Cheng
- Department of Immunology, Medical College, Nantong University, Nantong 226001, Jiangsu Province, People's Republic of China.
| |
Collapse
|
31
|
Hutterer C, Milbradt J, Hamilton S, Zaja M, Leban J, Henry C, Vitt D, Steingruber M, Sonntag E, Zeitträger I, Bahsi H, Stamminger T, Rawlinson W, Strobl S, Marschall M. Inhibitors of dual-specificity tyrosine phosphorylation-regulated kinases (DYRK) exert a strong anti-herpesviral activity. Antiviral Res 2017; 143:113-121. [PMID: 28400201 DOI: 10.1016/j.antiviral.2017.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/26/2017] [Accepted: 04/07/2017] [Indexed: 11/26/2022]
Abstract
Infection with human cytomegalovirus (HCMV) is a serious medical problem, particularly in immunocompromised individuals and neonates. The success of (val)ganciclovir therapy is hampered by low drug compatibility and induction of viral resistance. A novel strategy of antiviral treatment is based on the exploitation of cell-directed signaling, e. g. pathways with a known relevance for carcinogenesis and tumor drug development. Here we describe a principle for putative antiviral drugs based on targeting dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs). DYRKs constitute an evolutionarily conserved family of protein kinases with key roles in the control of cell proliferation and differentiation. Members of the DYRK family are capable of phosphorylating a number of substrate proteins, including regulators of the cell cycle, e.g. DYRK1B can induce cell cycle arrest, a critical step for the regulation of HCMV replication. Here we provide first evidence for a critical role of DYRKs during viral replication and the high antiviral potential of DYRK inhibitors (SC84227, SC97202 and SC97208, Harmine and AZ-191). Using established replication assays for laboratory and clinically relevant strains of HCMV, concentration-dependent profiles of inhibition were obtained. Mean inhibitory concentrations (EC50) of 0.98 ± 0.08 μM/SC84227, 0.60 ± 0.02 μM/SC97202, 6.26 ± 1.64 μM/SC97208, 0.71 ± 0.019 μM/Harmine and 0.63 ± 0.23 μM/AZ-191 were determined with HCMV strain AD169-GFP for the infection of primary human fibroblasts. A first analysis of the mode of antiviral action suggested a block of viral replication at the early-late stage of HCMV gene expression. Moreover, rhesus macaque cytomegalovirus (RhCMV), varicella-zoster virus (VZV) and herpes simplex virus (HSV-1) showed a similarly high sensitivity to these compounds. Thus, we conclude that DYRK signaling represents a promising target pathway for the development of novel anti-herpesviral strategies.
Collapse
Affiliation(s)
- Corina Hutterer
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany.
| | - Jens Milbradt
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - Stuart Hamilton
- Serology and Virology Division, SEALS Microbiology Prince of Wales Hospital Randwick NSW 2013 and SOMS and BABS, University of NSW, Sydney, Australia
| | - Mirko Zaja
- 4SC Discovery GmbH, Martinsried, Germany
| | | | | | | | - Mirjam Steingruber
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - Eric Sonntag
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - Isabel Zeitträger
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - Hanife Bahsi
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - Thomas Stamminger
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - William Rawlinson
- Serology and Virology Division, SEALS Microbiology Prince of Wales Hospital Randwick NSW 2013 and SOMS and BABS, University of NSW, Sydney, Australia
| | | | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany.
| |
Collapse
|
32
|
Wang X, Zhang Y, Nilsson CL, Berven FS, Andrén PE, Carlsohn E, Horvatovich P, Malm J, Fuentes M, Végvári Á, Welinder C, Fehniger TE, Rezeli M, Edula G, Hober S, Nishimura T, Marko-Varga G. Association of chromosome 19 to lung cancer genotypes and phenotypes. Cancer Metastasis Rev 2016; 34:217-26. [PMID: 25982285 DOI: 10.1007/s10555-015-9556-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The Chromosome 19 Consortium, a part of the Chromosome-Centric Human Proteome Project (C-HPP, http://www.C-HPP.org ), is tasked with the understanding chromosome 19 functions at the gene and protein levels, as well as their roles in lung oncogenesis. Comparative genomic hybridization (CGH) studies revealed chromosome aberration in lung cancer subtypes, including ADC, SCC, LCC, and SCLC. The most common abnormality is 19p loss and 19q gain. Sixty-four aberrant genes identified in previous genomic studies and their encoded protein functions were further validated in the neXtProt database ( http://www.nextprot.org/ ). Among those, the loss of tumor suppressor genes STK11, MUM1, KISS1R (19p13.3), and BRG1 (19p13.13) is associated with lung oncogenesis or remote metastasis. Gene aberrations include translocation t(15, 19) (q13, p13.1) fusion oncogene BRD4-NUT, DNA repair genes (ERCC1, ERCC2, XRCC1), TGFβ1 pathway activation genes (TGFB1, LTBP4), Dyrk1B, and potential oncogenesis protector genes such as NFkB pathway inhibition genes (NFKBIB, PPP1R13L) and EGLN2. In conclusion, neXtProt is an effective resource for the validation of gene aberrations identified in genomic studies. It promises to enhance our understanding of lung cancer oncogenesis.
Collapse
Affiliation(s)
- Xiangdong Wang
- Zhongshan Hospital, Shanghai Institute of Clinical Bioinformatics, Fudan University, Shanghai, China,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Radhakrishnan A, Nanjappa V, Raja R, Sathe G, Puttamallesh VN, Jain AP, Pinto SM, Balaji SA, Chavan S, Sahasrabuddhe NA, Mathur PP, Kumar MM, Prasad TSK, Santosh V, Sukumar G, Califano JA, Rangarajan A, Sidransky D, Pandey A, Gowda H, Chatterjee A. A dual specificity kinase, DYRK1A, as a potential therapeutic target for head and neck squamous cell carcinoma. Sci Rep 2016; 6:36132. [PMID: 27796319 PMCID: PMC5086852 DOI: 10.1038/srep36132] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 10/10/2016] [Indexed: 12/18/2022] Open
Abstract
Despite advances in clinical management, 5-year survival rate in patients with late-stage head and neck squamous cell carcinoma (HNSCC) has not improved significantly over the past decade. Targeted therapies have emerged as one of the most promising approaches to treat several malignancies. Though tyrosine phosphorylation accounts for a minority of total phosphorylation, it is critical for activation of signaling pathways and plays a significant role in driving cancers. To identify activated tyrosine kinase signaling pathways in HNSCC, we compared the phosphotyrosine profiles of a panel of HNSCC cell lines to a normal oral keratinocyte cell line. Dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A (DYRK1A) was one of the kinases hyperphosphorylated at Tyr-321 in all HNSCC cell lines. Inhibition of DYRK1A resulted in an increased apoptosis and decrease in invasion and colony formation ability of HNSCC cell lines. Further, administration of the small molecular inhibitor against DYRK1A in mice bearing HNSCC xenograft tumors induced regression of tumor growth. Immunohistochemical labeling of DYRK1A in primary tumor tissues using tissue microarrays revealed strong to moderate staining of DYRK1A in 97.5% (39/40) of HNSCC tissues analyzed. Taken together our results suggest that DYRK1A could be a novel therapeutic target in HNSCC.
Collapse
Affiliation(s)
- Aneesha Radhakrishnan
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry 605014, India
| | - Vishalakshi Nanjappa
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
- Amrita School of Biotechnology, Amrita University, Kollam 690 525, India
| | - Remya Raja
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
| | - Gajanan Sathe
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
- Manipal University, Madhav Nagar, Manipal 576104, India
| | - Vinuth N. Puttamallesh
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
- Amrita School of Biotechnology, Amrita University, Kollam 690 525, India
| | - Ankit P. Jain
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
- School of Biotechnology, KIIT University, Bhubaneswar 751024, India
| | - Sneha M. Pinto
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
| | - Sai A. Balaji
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Sandip Chavan
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
- Manipal University, Madhav Nagar, Manipal 576104, India
| | | | - Premendu P. Mathur
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry 605014, India
- School of Biotechnology, KIIT University, Bhubaneswar 751024, India
| | - Mahesh M. Kumar
- Department of Neuro-Virology, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - T. S. Keshava Prasad
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
- Amrita School of Biotechnology, Amrita University, Kollam 690 525, India
- YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University, Mangalore 575018, India
| | - Vani Santosh
- Department of Pathology, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Geethanjali Sukumar
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
| | - Joseph A. Califano
- Milton J. Dance Head and Neck Center, Greater Baltimore Medical Center, Baltimore, MD 21204, USA
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Annapoorni Rangarajan
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine,Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Harsha Gowda
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
- YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University, Mangalore 575018, India
| | - Aditi Chatterjee
- Institute of Bioinformatics, International Technology Park, Bangalore, 560 066, India
- YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University, Mangalore 575018, India
| |
Collapse
|
34
|
|
35
|
Ashford AL, Dunkley TPJ, Cockerill M, Rowlinson RA, Baak LM, Gallo R, Balmanno K, Goodwin LM, Ward RA, Lochhead PA, Guichard S, Hudson K, Cook SJ. Identification of DYRK1B as a substrate of ERK1/2 and characterisation of the kinase activity of DYRK1B mutants from cancer and metabolic syndrome. Cell Mol Life Sci 2016; 73:883-900. [PMID: 26346493 PMCID: PMC4735261 DOI: 10.1007/s00018-015-2032-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 08/25/2015] [Accepted: 08/27/2015] [Indexed: 12/18/2022]
Abstract
The dual-specificity tyrosine-phosphorylation-regulated kinase, DYRK1B, is expressed de novo during myogenesis, amplified or mutated in certain cancers and mutated in familial cases of metabolic syndrome. DYRK1B is activated by cis auto-phosphorylation on tyrosine-273 (Y273) within the activation loop during translation but few other DYRK1B phosphorylation sites have been characterised to date. Here, we demonstrate that DYRK1B also undergoes trans-autophosphorylation on serine-421 (S421) in vitro and in cells and that this site contributes to DYRK1B kinase activity. Whilst a DYRK1B(S421A) mutant was completely defective for p-S421 in cells, DYRK1B inhibitors caused only a partial loss of p-S421 suggesting the existence of an additional kinase that could also phosphorylate DYRK1B S421. Indeed, a catalytically inactive DYRK1B(D239A) mutant exhibited very low levels of p-S421 in cells but this was increased by KRAS(G12V). In addition, selective activation of the RAF-MEK1/2-ERK1/2 signalling pathway rapidly increased p-S421 in cells whereas activation of the stress kinases JNK or p38 could not. S421 resides within a Ser-Pro phosphoacceptor motif that is typical for ERK1/2 and recombinant ERK2 phosphorylated DYRK1B at S421 in vitro. Our results show that DYRK1B is a novel ERK2 substrate, uncovering new links between two kinases involved in cell fate decisions. Finally, we show that DYRK1B mutants that have recently been described in cancer and metabolic syndrome exhibit normal or reduced intrinsic kinase activity.
Collapse
Affiliation(s)
- Anne L Ashford
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| | - Tom P J Dunkley
- AstraZeneca, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
- Roche Innovation Center Basel, Basel, Switzerland
| | - Mark Cockerill
- AstraZeneca, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
- Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK
| | | | - Lisa M Baak
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Raffaella Gallo
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Kathryn Balmanno
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Louise M Goodwin
- AstraZeneca, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
| | - Richard A Ward
- AstraZeneca, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
| | - Pamela A Lochhead
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Sylvie Guichard
- AstraZeneca, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
- AstraZeneca, Waltham, MA, 02451, USA
| | - Kevin Hudson
- AstraZeneca, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
| | - Simon J Cook
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| |
Collapse
|
36
|
Zhou N, Yuan S, Wang R, Zhang W, Chen JJ. Role of dual specificity tyrosine-phosphorylation-regulated kinase 1B (Dyrk1B) in S-phase entry of HPV E7 expressing cells from quiescence. Oncotarget 2015; 6:30745-61. [PMID: 26307683 PMCID: PMC4741565 DOI: 10.18632/oncotarget.5222] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 08/08/2015] [Indexed: 12/18/2022] Open
Abstract
The high-risk human papillomavirus (HPV) is the causative agent for cervical cancer. The HPV E7 oncogene promotes S-phase entry from quiescent state in the presence of elevated cell cycle inhibitor p27Kip1, a function that may contribute to carcinogenesis. However, the mechanism by which HPV E7 induces quiescent cells to entry into S-phase is not fully understood. Interestingly, we found that Dyrk1B, a dual-specificity kinase and negative regulator of cell proliferation in quiescent cells, was upregulated in E7 expressing cells. Surprisingly and in contrast to what was previously reported, Dyrk1B played a positive role in S-phase entry of quiescent HPV E7 expressing cells. Mechanistically, Dyrk1B contributed to p27 phosphorylation (at serine 10 and threonine 198), which was important for the proliferation of HPV E7 expressing cells. Moreover, Dyrk1B up-regulated HPV E7. Taken together, our studies uncovered a novel function of Dyrk1B in high-risk HPV E7-mediated cell proliferation. Dyrk1B may serve as a target for therapy in HPV-associated cancers.
Collapse
Affiliation(s)
- Na Zhou
- Cancer Research Center, Shandong University School of Medicine, Jinan, Shandong, China
| | - Shoudao Yuan
- Cancer Research Center, Shandong University School of Medicine, Jinan, Shandong, China
| | - Rongchun Wang
- Biology Institute of Shandong Academy of Sciences, Jinan, Shandong, China
| | - Weifang Zhang
- Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Jason J. Chen
- Cancer Research Center, Shandong University School of Medicine, Jinan, Shandong, China
| |
Collapse
|
37
|
Masaki S, Kii I, Sumida Y, Kato-Sumida T, Ogawa Y, Ito N, Nakamura M, Sonamoto R, Kataoka N, Hosoya T, Hagiwara M. Design and synthesis of a potent inhibitor of class 1 DYRK kinases as a suppressor of adipogenesis. Bioorg Med Chem 2015; 23:4434-4441. [PMID: 26145823 DOI: 10.1016/j.bmc.2015.06.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 06/05/2015] [Accepted: 06/06/2015] [Indexed: 12/28/2022]
Abstract
Dysregulation of dual-specificity tyrosine-phosphorylation-regulated kinase 1A (DYRK1A) has been demonstrated in several pathological conditions, including Alzheimer's disease and cancer progression. It has been recently reported that a gain of function-mutation in the human DYRK1B gene exacerbates metabolic syndrome by enhancing obesity. In the previous study, we developed an inhibitor of DYRK family kinases (INDY) and demonstrated that INDY suppresses the pathological phenotypes induced by overexpression of DYRK1A or DYRK1B in cellular and animal models. In this study, we designed and synthesized a novel inhibitor of DYRK family kinases based on the crystal structure of the DYRK1A/INDY complex by replacing the phenol group of INDY with dibenzofuran to produce a derivative, named BINDY. This compound exhibited potent and selective inhibitory activity toward DYRK family kinases in an in vitro assay. Furthermore, treatment of 3T3-L1 pre-adipocytes with BINDY hampered adipogenesis by suppressing gene expression of the critical transcription factors PPARγ and C/EBPα. This study indicates the possibility of BINDY as a potential drug for metabolic syndrome.
Collapse
Affiliation(s)
- So Masaki
- Laboratory for Malignancy Control Research, Medical Innovation Center, Graduate School of Medicine, Kyoto University, 53, Shigoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Isao Kii
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuto Sumida
- Laboratory of Chemical Bioscience, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Tomoe Kato-Sumida
- Laboratory of Chemical Bioscience, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Yasushi Ogawa
- Department of Dermatology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Nobutoshi Ito
- Department of Structural Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Mitsuhiro Nakamura
- Division of Natural Sciences, Graduate School of Integrated Arts and Sciences, University of Tokushima, Tokushima 770-8502, Japan
| | - Rie Sonamoto
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Naoyuki Kataoka
- Laboratory for Malignancy Control Research, Medical Innovation Center, Graduate School of Medicine, Kyoto University, 53, Shigoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takamitsu Hosoya
- Laboratory of Chemical Bioscience, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Masatoshi Hagiwara
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
38
|
Martin KA, Mani MV, Mani A. New targets to treat obesity and the metabolic syndrome. Eur J Pharmacol 2015; 763:64-74. [PMID: 26001373 DOI: 10.1016/j.ejphar.2015.03.093] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 03/19/2015] [Accepted: 03/30/2015] [Indexed: 01/12/2023]
Abstract
Metabolic syndrome (MetS) is a cluster ofassociated metabolic traits that collectively confer unsurpassed risk for development of cardiovascular disease (CVD) and type 2 diabetes compared to any single CVD risk factor. Truncal obesity plays an exceptionally critical role among all metabolic traits of the MetS. Consequently, the prevalence of the MetS has steadily increased with the growing epidemic of obesity. Pharmacotherapy has been available for obesity for more than one decade, but with little success in improving the metabolic profiles. The serotonergic drugs and inhibitors of pancreatic lipases were among the few drugs that were initially approved to treat obesity. At the present time, only the pancreatic lipase inhibitor orlistat is approved for long-term treatment of obesity. New classes of anti-diabetic drugs, including glucagon-like peptide 1 receptor (GLP-1R) agonists and Dipeptidyl-peptidase IV (DPP-IV) inhibitors, are currently being evaluated for their effects on obesity and metabolic traits. The genetic studies of obesity and metabolic syndrome have identified novel molecules acting on the hunger and satiety peptidergic signaling of the gut-hypothalamus axis or the melanocortin system of the brain and are promising targets for future drug development. The goal is to develop drugs that not only treat obesity, but also favorably impact its associated traits.
Collapse
Affiliation(s)
- Kathleen A Martin
- Department of Internal Medicine, Yale University School of Medicine, USA
| | | | - Arya Mani
- Department of Internal Medicine, Yale University School of Medicine, USA; Department of Genetics, Yale University School of Medicine, USA.
| |
Collapse
|
39
|
Fernández-Martínez P, Zahonero C, Sánchez-Gómez P. DYRK1A: the double-edged kinase as a protagonist in cell growth and tumorigenesis. Mol Cell Oncol 2015; 2:e970048. [PMID: 27308401 PMCID: PMC4905233 DOI: 10.4161/23723548.2014.970048] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 09/03/2014] [Accepted: 09/03/2014] [Indexed: 01/12/2023]
Abstract
DYRK1A (dual-specificity tyrosine-regulated kinase 1A) is a kinase with multiple implications for embryonic development, especially in the nervous system where it regulates the balance between proliferation and differentiation of neural progenitors. The DYRK1A gene is located in the Down syndrome critical region and may play a significant role in the developmental brain defects, early neurodegeneration, and cancer susceptibility of individuals with this syndrome. DYRK1A is also expressed in adults, where it might participate in the regulation of cell cycle, survival, and tumorigenesis, thus representing a potential therapeutic target for certain types of cancer. However, the final readout of DYRK1A overexpression or inhibition depends strongly on the cellular context, as it has both tumor suppressor and oncogenic activities. Here, we will discuss the functions and substrates of DYRK1A associated with the control of cell growth and tumorigenesis with a focus on the potential use of DYRK1A inhibitors in cancer therapy.
Collapse
Affiliation(s)
- P Fernández-Martínez
- Instituto de Medicina Molecular Aplicada; Universidad CEU-San Pablo ; Madrid, Spain
| | - C Zahonero
- Neuro-oncology Unit; Instituto de Salud Carlos III-UFIEC ; Madrid, Spain
| | - P Sánchez-Gómez
- Neuro-oncology Unit; Instituto de Salud Carlos III-UFIEC ; Madrid, Spain
| |
Collapse
|
40
|
Li L, Liu Y, Zhang Q, Zhou H, Zhang Y, Yan B. Comparison of cancer cell survival triggered by microtubule damage after turning Dyrk1B kinase on and off. ACS Chem Biol 2014; 9:731-42. [PMID: 24377315 DOI: 10.1021/cb4005589] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Using a tubulin polymerization inhibitor and a tubulin polymerization/Dyrk1B dual inhibitor, we intentionally allowed or blocked the Dyrk1B-coordinated cell survival process in response to microtubule damage. By examining the resulting differential effects on cell function and phenotype, we have elucidated key molecular interactions involved in the Dyrk1B-coordinated cell survival process as well as the associated overall cellular impact. Dyrk1B activation that is induced by microtubule damage triggers microtubule stabilization and promotes the mitochondrial translocation of p21(Cip1/waf1) (referred to as p21 hereafter) to suppress apoptosis. These coordinated survival events rapidly repair microtubules, relieve cell G2/M arrest for 42% of the cells, suppress apoptosis for 27% of the cells, and increase cell viability by 10-fold. That is, the dual inhibitor is 10 times more potent in the inhibition of cancer cell viability. This approach affords a novel drug discovery strategy by targeting both the therapeutic targets and the associated cell survival pathway using a single therapeutic agent.
Collapse
Affiliation(s)
- Liwen Li
- School of Chemistry and Chemical Engineering, Shandong University , Jinan, China 250100
| | | | | | | | | | | |
Collapse
|
41
|
Hartono SB, Yu M, Gu W, Yang J, Strounina E, Wang X, Qiao S, Yu C. Synthesis of multi-functional large pore mesoporous silica nanoparticles as gene carriers. NANOTECHNOLOGY 2014; 25:055701. [PMID: 24406311 DOI: 10.1088/0957-4484/25/5/055701] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The development of functional nanocarriers that can enhance the cellular delivery of a variety of nucleic acid agents is important in many biomedical applications such as siRNA therapy. We report the synthesis of large pore mesoporous silica nanoparticles (LPMSN) loaded with iron oxide and covalently modified by polyethyleneimine (denoted PEI-Fe-LPMSN) as carriers for gene delivery. The LPMSN have a particle size of ∼200 nm and a large pore size of 11 nm. The large pore size is essential for the formation of large iron oxide nanoparticles to increase the magnetic properties and the adsorption capacity of siRNA molecules. The magnetic property facilitates the cellular uptake of nanocarriers under an external magnetic field. PEI is covalently grafted on the silica surface to enhance the nanocarriers' affinity against siRNA molecules and to improve gene silencing performance. The PEI-Fe-LPMSN delivered siRNA-PLK1 effectively into osteosarcoma cancer cells, leading to cell viability inhibition of 80%, higher compared to the 50% reduction when the same dose of siRNA was delivered by a commercial product, oligofectamine.
Collapse
Affiliation(s)
- Sandy B Hartono
- ARC Centre of Excellence for Functional Nanomaterials, Australian Institute for Bioengineering and Nanotechnology, University of Queensland, QLD 4072, Australia
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Ashford AL, Oxley D, Kettle J, Hudson K, Guichard S, Cook SJ, Lochhead PA. A novel DYRK1B inhibitor AZ191 demonstrates that DYRK1B acts independently of GSK3β to phosphorylate cyclin D1 at Thr(286), not Thr(288). Biochem J 2014; 457:43-56. [PMID: 24134204 DOI: 10.1042/bj20130461] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
DYRK1B (dual-specificity tyrosine phosphorylation-regulated kinase 1B) is amplified in certain cancers and may be an oncogene; however, our knowledge of DYRK1B has been limited by the lack of selective inhibitors. In the present study we describe AZ191, a potent small molecule inhibitor that selectively inhibits DYRK1B in vitro and in cells. CCND1 (cyclin D1), a key regulator of the mammalian G1-S-phase transition, is phosphorylated on Thr(286) by GSK3β (glycogen synthase kinase 3β) to promote its degradation. DYRK1B has also been proposed to promote CCND1 turnover, but was reported to phosphorylate Thr(288) rather than Thr(286). Using in vitro kinase assays, phospho-specific immunoblot analysis and MS in conjunction with AZ191 we now show that DYRK1B phosphorylates CCND1 at Thr(286), not Thr(288), in vitro and in cells. In HEK (human embryonic kidney)-293 and PANC-1 cells (which exhibit DYRK1B amplification) DYRK1B drives Thr(286) phosphorylation and proteasome-dependent turnover of CCND1 and this is abolished by AZ191 or DYRK1B RNAi, but not by GSK3β inhibitors or GSK3β RNAi. DYRK1B expression causes a G1-phase cell-cycle arrest, but overexpression of CCND1 (wild-type or T286A) fails to overcome this; indeed, DYRK1B also promotes the expression of p21CIP1 (21 kDa CDK-interacting protein 1) and p27KIP1 (CDK-inhibitory protein 1). The results of the present study demonstrate for the first time that DYRK1B is a novel Thr(286)-CCND1 kinase that acts independently of GSK3β to promote CCND1 degradation. Furthermore, we anticipate that AZ191 may prove useful in defining further substrates and biological functions of DYRK1B.
Collapse
Affiliation(s)
- Anne L Ashford
- *Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, U.K
| | | | | | | | | | | | | |
Collapse
|
43
|
Gao J, Zhao Y, Lv Y, Chen Y, Wei B, Tian J, Yang Z, Kong F, Pang J, Liu J, Shi H. Mirk/Dyrk1B mediates G0/G1 to S phase cell cycle progression and cell survival involving MAPK/ERK signaling in human cancer cells. Cancer Cell Int 2013; 13:2. [PMID: 23311607 PMCID: PMC3575355 DOI: 10.1186/1475-2867-13-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 01/08/2013] [Indexed: 01/09/2023] Open
Abstract
Background Mirk/Dyrk1B contributes to G0 arrest by destabilization of cyclin D1 and stabilization of p27kip1 to maintain the viability of quiescent human cancer cells, and it could be negatively regulated by mitogenic-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling. This study was performed to investigate the effect of Mirk/Dyrk1B on cell cycle and survival of human cancer cells involving MAPK/ERK signaling. Methods The correlations between Mirk/Dyrk1B expression and active ERK1/2 detected by western blot in both ovarian cancer and non-small cell lung cancer (NSCLC) cells were analyzed by simple regression. Mirk/Dyrk1B unique phosphopeptides with sites associated with Mirk/Dyrk1B protein were isolated and quantitated by liquid chromatography coupled to tandem mass/mass spectrometry (LC-MS/MS) proteomics analysis. The human cancer cells were treated with small interfering RNAs (siRNAs) and/or U0126, an inhibitor of MEK for indicated duration, followed by investigating the alterations of cell cycle and apoptosis as well as related proteins examined by flow cytometry and Western blot, respectively. Results Our study demonstrated the widely expressed Mirk/Dyrk1B proteins in the human cancer cells were positively correlated with the levels of activated ERK1/2. Moreover, Mirk/Dyrk1B protein expressions consistent with the tyrosine autophosphorylated levels in the human cancer cells were increased by U0126 or growth factor-depleted culture. Conversely, knockdown of Mirk/Dyrk1B by siRNA led to up-regulated activation of c-Raf-MEK-ERK1/2 pathway and subsequent changes in cell cycle proteins (cyclin D1, p27kip1), accompanied by increased growth rate and cells from G0/G1 into S of cell cycle which could be blocked by U0126 in a dose-dependent manner, indicating Mirk/Dyrk1B may sequester MAPK/ERK pathway, and vice versa. Whereas, combined Mirk siRNA and U0126 induced cell apoptosis in the human cancer cells. Conclusions These data together show that Mirk/Dyrk1B mediates cell cycle and survival via interacting with MAPK/ERK signals and simultaneous inhibition of both pathways may be a novel therapeutic target for human cancer.
Collapse
Affiliation(s)
- Jingchun Gao
- Department of Obstetrics & Gynecology, First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, Liaoning 116011, China
| | - Yi Zhao
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, China
| | - Yunyi Lv
- Department of Obstetrics & Gynecology, First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, Liaoning 116011, China
| | - Yamin Chen
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, China
| | - Bing Wei
- Department of Obstetrics & Gynecology, First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, Liaoning 116011, China
| | - Jianxin Tian
- Department of Obstetrics & Gynecology, First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, Liaoning 116011, China
| | - Zhihai Yang
- Department of Obstetrics & Gynecology, First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, Liaoning 116011, China
| | - Fandou Kong
- Department of Obstetrics & Gynecology, First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, Liaoning 116011, China
| | - Jian Pang
- Department of Obstetrics & Gynecology, First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, Liaoning 116011, China
| | - Jiwei Liu
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, China
| | - Hong Shi
- Department of Obstetrics & Gynecology, First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, Liaoning 116011, China
| |
Collapse
|
44
|
Yousef AF, Fonseca GJ, Cohen MJ, Mymryk JS. The C-terminal region of E1A: a molecular tool for cellular cartography. Biochem Cell Biol 2012; 90:153-63. [DOI: 10.1139/o11-080] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The adenovirus E1A proteins function via protein–protein interactions. By making many connections with the cellular protein network, individual modules of this virally encoded hub reprogram numerous aspects of cell function and behavior. Although many of these interactions have been thoroughly studied, those mediated by the C-terminal region of E1A are less well understood. This review focuses on how this region of E1A affects cell cycle progression, apoptosis, senescence, transformation, and conversion of cells to an epithelial state through interactions with CTBP1/2, DYRK1A/B, FOXK1/2, and importin-α. Furthermore, novel potential pathways that the C-terminus of E1A influences through these connections with the cellular interaction network are discussed.
Collapse
Affiliation(s)
- Ahmed F. Yousef
- Departments of Microbiology and Immunology and Oncology, The University of Western Ontario, London Regional Cancer Program, London, ON N6A 4L6, Canada
| | - Gregory J. Fonseca
- Departments of Microbiology and Immunology and Oncology, The University of Western Ontario, London Regional Cancer Program, London, ON N6A 4L6, Canada
| | - Michael J. Cohen
- Departments of Microbiology and Immunology and Oncology, The University of Western Ontario, London Regional Cancer Program, London, ON N6A 4L6, Canada
| | - Joe S. Mymryk
- Departments of Microbiology and Immunology and Oncology, The University of Western Ontario, London Regional Cancer Program, London, ON N6A 4L6, Canada
| |
Collapse
|
45
|
GAO JINGCHUN, YANG XIANGJUN, YIN PING, HU WENFENG, LIAO HONGFENG, MIAO ZHIHUI, PAN CHAO, LI NA. The involvement of FoxO in cell survival and chemosensitivity mediated by Mirk/Dyrk1B in ovarian cancer. Int J Oncol 2012; 40:1203-9. [PMID: 22159921 PMCID: PMC3584584 DOI: 10.3892/ijo.2011.1293] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 11/18/2011] [Indexed: 12/13/2022] Open
Abstract
Minibrain-related kinase (Mirk) is a serine/threonine kinase which is also known as the dual specificity tyrosine-phosphorylation-regulated kinase 1B (Dyrk1B). It is known that Dyrk1A, the closest family member to Mirk/Dyrk1B can mediate cellular localization of mammalian forkhead subclass O (FoxO1), a transcription factor, although the effect of Mirk/Dyrk1B on FoxO factors remains to be defined. In this study, we showed that Mirk/Dyrk1B protein was overexpressed in 5 of 8 ovarian cancer cell lines and negatively correlated with the protein level of FoxO factors (FoxO1+FoxO3A). Knockdown of Mirk by small interfering RNA (siRNA) resulted in cell apoptosis and sensitized cells to cisplatin accompanied by nuclear translocation of FoxO1 and/or FoxO3A as well as increased Bim, TRADD, cleaved caspase-3 and PARP. Furthermore, combined siRNAs of Mirk with FoxO1 and/or FoxO3A led to fewer apoptotic cells and cisplatin sensitivity compared to Mirk siRNA alone, suggesting that FoxO is involved in Mirk-mediated cell survival and chemosensitivity of ovarian cancer. Taken together, Mirk/Dyrk1B plays an important role in ovarian cancer cell survival through modulating FoxO translocation and may be a novel therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- JINGCHUN GAO
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning 116011
| | - XIANGJUN YANG
- Department of Obstetrics and Gynecology, Zhongshan Hospital Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - PING YIN
- Department of Pathology, Zhongshan Hospital Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - WENFENG HU
- Department of Obstetrics and Gynecology, Zhongshan Hospital Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - HONGFENG LIAO
- Department of Pathology, Zhongshan Hospital Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - ZHIHUI MIAO
- Department of Obstetrics and Gynecology, Zhongshan Hospital Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - CHAO PAN
- Department of Pathology, Zhongshan Hospital Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - NA LI
- Department of Obstetrics and Gynecology, Zhongshan Hospital Xiamen University, Xiamen, Fujian 361004, P.R. China
| |
Collapse
|
46
|
Hartono SB, Gu W, Kleitz F, Liu J, He L, Middelberg APJ, Yu C, Lu GQM, Qiao SZ. Poly-L-lysine functionalized large pore cubic mesostructured silica nanoparticles as biocompatible carriers for gene delivery. ACS NANO 2012; 6:2104-2117. [PMID: 22385282 DOI: 10.1021/nn2039643] [Citation(s) in RCA: 201] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Large pore mesoporous silica nanoparticles (LP-MSNs) functionalized with poly-L-lysine (PLL) were designed as a new carrier material for gene delivery applications. The synthesized LP-MSNs are 100-200 nm in diameter and are composed of cage-like pores organized in a cubic mesostructure. The size of the cavities is about 28 nm with an entrance size of 13.4 nm. Successful grafting of PLL onto the silica surface through covalent immobilization was confirmed by X-ray photoelectron spectroscopy, solid-state (13)C magic-angle spinning nuclear magnetic resonance, Fourier transformed infrared, and thermogravimetric analysis. As a result of the particle modification with PLL, a significant increase of the nanoparticle binding capacity for oligo-DNAs was observed compared to the native unmodified silica particles. Consequently, PLL-functionalized nanoparticles exhibited a strong ability to deliver oligo DNA-Cy3 (a model for siRNA) to Hela cells. Furthermore, PLL-functionalized nanoparticles were proven to be superior as gene carriers compared to amino-functionalized nanoparticles and the native nanoparticles. The system was tested to deliver functional siRNA against minibrain-related kinase and polo-like kinase 1 in osteosarcoma cancer cells. Here, the functionalized particles demonstrated great potential for efficient gene transfer into cancer cells as a decrease of the cellular viability of the osteosarcoma cancer cells was induced. Moreover, the PLL-modified silica nanoparticles also exhibit a high biocompatibility, with low cytotoxicity observed up to 100 μg/mL.
Collapse
Affiliation(s)
- Sandy B Hartono
- ARC Centre of Excellence for Functional Nanomaterials, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Miyata Y, Nishida E. DYRK1A binds to an evolutionarily conserved WD40-repeat protein WDR68 and induces its nuclear translocation. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1813:1728-39. [PMID: 21777625 DOI: 10.1016/j.bbamcr.2011.06.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 06/10/2011] [Accepted: 06/30/2011] [Indexed: 12/25/2022]
Abstract
DYRK1A is encoded in the Down's syndrome critical region on human chromosome 21, and plays an important role in the functional and developmental regulation of many types of cells, including neuronal cells. Here we have identified WDR68, an evolutionarily conserved protein with WD40-repeat domains, as a cellular binding partner of DYRK1A. WDR68 was originally identified in petunia as AN11 that controls the pigmentation of flowers by stimulating the transcription of anthocyanin biosynthetic genes. Experiments with RNA interference showed that WDR68 was indispensable for the optimal proliferation and survival of mammalian cultured cell, and WDR68 depletion induced cell apoptosis. DYRK1A and DYRK1B, but not DYRK2, DYRK3, or DYRK4, bound to endogenous and expressed WDR68. The N-terminal domain, but not the catalytic kinase domain or the C-terminal domain of DYRK1A, was responsible for the WDR68 binding. Deletions in the N-terminal or C-terminal region outside of the central WD40-repeats of WDR68 abolished its binding to DYRK1A, suggesting that WD40 repeats are not sufficient for the association with DYRK1A. Immunofluorescent staining revealed that WDR68 was distributed throughout the cell. Importantly, nuclear accumulation of WDR68 was observed upon co-expression of the wild type and a kinase-dead mutant of DYRK1A. Taken together, these results suggest that DYRK1A binds specifically to WDR68 in cells, and that the binding, but not the phosphorylation event, induces the nuclear translocation of WDR68.
Collapse
Affiliation(s)
- Yoshihiko Miyata
- Department of Cell and Developmental Biology, Kyoto University, Kyoto, Japan.
| | | |
Collapse
|
48
|
Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, Castedo M, Kroemer G. Molecular mechanisms of cisplatin resistance. Oncogene 2011; 31:1869-83. [PMID: 21892204 DOI: 10.1038/onc.2011.384] [Citation(s) in RCA: 1989] [Impact Index Per Article: 142.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Platinum-based drugs, and in particular cis-diamminedichloroplatinum(II) (best known as cisplatin), are employed for the treatment of a wide array of solid malignancies, including testicular, ovarian, head and neck, colorectal, bladder and lung cancers. Cisplatin exerts anticancer effects via multiple mechanisms, yet its most prominent (and best understood) mode of action involves the generation of DNA lesions followed by the activation of the DNA damage response and the induction of mitochondrial apoptosis. Despite a consistent rate of initial responses, cisplatin treatment often results in the development of chemoresistance, leading to therapeutic failure. An intense research has been conducted during the past 30 years and several mechanisms that account for the cisplatin-resistant phenotype of tumor cells have been described. Here, we provide a systematic discussion of these mechanism by classifying them in alterations (1) that involve steps preceding the binding of cisplatin to DNA (pre-target resistance), (2) that directly relate to DNA-cisplatin adducts (on-target resistance), (3) concerning the lethal signaling pathway(s) elicited by cisplatin-mediated DNA damage (post-target resistance) and (4) affecting molecular circuitries that do not present obvious links with cisplatin-elicited signals (off-target resistance). As in some clinical settings cisplatin constitutes the major therapeutic option, the development of chemosensitization strategies constitute a goal with important clinical implications.
Collapse
Affiliation(s)
- L Galluzzi
- INSERM, U848 Apoptosis, Cancer and Immunity, Villejuif, France
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Rosenthal AS, Tanega C, Shen M, Mott BT, Bougie JM, Nguyen DT, Misteli T, Auld DS, Maloney DJ, Thomas CJ. Potent and selective small molecule inhibitors of specific isoforms of Cdc2-like kinases (Clk) and dual specificity tyrosine-phosphorylation-regulated kinases (Dyrk). Bioorg Med Chem Lett 2011; 21:3152-8. [PMID: 21450467 PMCID: PMC3085634 DOI: 10.1016/j.bmcl.2011.02.114] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 02/24/2011] [Accepted: 02/28/2011] [Indexed: 11/22/2022]
Abstract
Continued examination of substituted 6-arylquinazolin-4-amines as Clk4 inhibitors resulted in selective inhibitors of Clk1, Clk4, Dyrk1A and Dyrk1B. Several of the most potent inhibitors were validated as being highly selective within a comprehensive kinome scan.
Collapse
Affiliation(s)
- Andrew S. Rosenthal
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - Cordelle Tanega
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - Min Shen
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - Bryan T. Mott
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - James M. Bougie
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - Dac-Trung Nguyen
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - Tom Misteli
- Cell Biology of Genomes, National Cancer Institute, NIH, 41 Library Drive, Bethesda, MD 20892 USA
| | - Douglas S. Auld
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - David J. Maloney
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| | - Craig J. Thomas
- NIH Chemical Genomics Center, National Human Genome Research Institute, NIH 9800 Medical Center Drive, MSC 3370 Bethesda, MD 20892-3370 USA
| |
Collapse
|
50
|
Aranda S, Laguna A, de la Luna S. DYRK family of protein kinases: evolutionary relationships, biochemical properties, and functional roles. FASEB J 2011; 25:449-62. [PMID: 21048044 DOI: 10.1096/fj.10-165837] [Citation(s) in RCA: 242] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dual-specificity tyrosine-regulated kinases (DYRKs) comprise a family of protein kinases within the CMGC group of the eukaryotic kinome. Members of the DYRK family are found in 4 (animalia, plantae, fungi, and protista) of the 5 main taxa or kingdoms, and all DYRK proteins studied to date share common structural, biochemical, and functional properties with their ancestors in yeast. Recent work on DYRK proteins indicates that they participate in several signaling pathways critical for developmental processes and cell homeostasis. In this review, we focus on the DYRK family of proteins from an evolutionary, biochemical, and functional point of view and discuss the most recent, relevant, and controversial contributions to the study of these kinases.
Collapse
Affiliation(s)
- Sergi Aranda
- Center for Genomic Regulation, University Pompeu Fabra, Barcelona, Spain
| | | | | |
Collapse
|