1
|
O'Boyle B, Yeung W, Lu JD, Katiyar S, Yaron-Barir TM, Johnson JL, Cantley LC, Kannan N. An atlas of bacterial serine-threonine kinases reveals functional diversity and key distinctions from eukaryotic kinases. Sci Signal 2025; 18:eadt8686. [PMID: 40327749 DOI: 10.1126/scisignal.adt8686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/11/2025] [Indexed: 05/08/2025]
Abstract
Bacterial serine-threonine kinases (STKs) regulate diverse cellular processes associated with cell growth, virulence, and pathogenicity and are evolutionarily related to the druggable eukaryotic STKs. A deeper understanding of how bacterial STKs differ from their eukaryotic counterparts and how they have evolved to regulate diverse bacterial signaling functions is crucial for advancing the discovery and development of new antibiotic therapies. Here, we classified more than 300,000 bacterial STK sequences from the NCBI RefSeq nonredundant and UniProt protein databases into 35 canonical and seven pseudokinase families on the basis of the patterns of evolutionary constraints in the conserved catalytic domain and flanking regulatory domains. Through statistical comparisons, we identified features distinguishing bacterial STKs from eukaryotic STKs, including an arginine residue in a regulatory helix (C helix) that dynamically couples the ATP- and substrate-binding lobes of the kinase domain. Biochemical and peptide library screens demonstrated that evolutionarily constrained residues contributed to substrate specificity and kinase activation in the Mycobacterium tuberculosis kinase PknB. Together, these findings open previously unidentified avenues for investigating bacterial STK functions in cellular signaling and for developing selective bacterial STK inhibitors.
Collapse
Affiliation(s)
- Brady O'Boyle
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Wayland Yeung
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA
| | - Jason D Lu
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Samiksha Katiyar
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA
| | - Tomer M Yaron-Barir
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jared L Johnson
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Natarajan Kannan
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
2
|
O'Boyle B, Yeung W, Lu JD, Katiyar S, Yaron-Barir TM, Johnson JL, Cantley LC, Kannan N. Atlas of the Bacterial Serine-Threonine Kinases expands the functional diversity of the kinome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.12.632604. [PMID: 39868133 PMCID: PMC11760699 DOI: 10.1101/2025.01.12.632604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Bacterial serine-threonine protein kinases (STKs) regulate diverse cellular processes associated with cell growth, virulence, and pathogenicity. They are evolutionarily related to the druggable eukaryotic STKs. However, an incomplete knowledge of how bacterial STKs differ from their eukaryotic counterparts and how they have diverged to regulate diverse bacterial signaling functions presents a bottleneck in targeting them for drug discovery efforts. Here, we classified over 300,000 bacterial STK sequences from the NCBI RefSeq non-redundant and UniProt protein databases into 35 canonical and seven non-canonical (pseudokinase) families based on the patterns of evolutionary constraints in the conserved catalytic domain and flanking regulatory domains. Through statistical comparisons, we identified distinguishing features of bacterial STKs, including a distinctive arginine residue in a regulatory helix (C-Helix) that dynamically couples ATP and substrate binding lobes of the kinase domain. Biochemical and peptide-library screens demonstrated that constrained residues contribute to substrate specificity and kinase activation in the Mycobacterium tuberculosis kinase PknB. Collectively, these findings open new avenues for investigating bacterial STK functions in cellular signaling and for the development of selective bacterial STK inhibitors.
Collapse
|
3
|
Hu X, Wu Y, Ouyang H, Wu J, Yao M, Chen Z, Li Q. Virtual Screening, Molecular Dynamics, and Mechanism Study of Homeodomain-Interacting Protein Kinase 2 Inhibitor in Renal Fibroblasts. Pharmaceuticals (Basel) 2024; 17:1420. [PMID: 39598332 PMCID: PMC11597050 DOI: 10.3390/ph17111420] [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: 10/04/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Homeodomain-interacting protein kinase 2 (HIPK2) is critically involved in the progression of renal fibrosis. This study aims to identify and characterize a novel HIPK2 inhibitor, CHR-6494, and investigate its therapeutic potential. Methods: Using structure-based virtual screening and molecular dynamics simulations, we identified CHR-6494 as a potent HIPK2 inhibitor with an IC50 of 0.97 μM. The effects of CHR-6494 on the phosphorylation of p53 in Normal Rattus norvegicus kidney cells (NRK-49F) induced by transforming growth factor-β (TGF-β) were assessed, along with its impact on TGF-β signaling and downstream profibrotic markers. Results: CHR-6494 significantly reduces p53 phosphorylation induced by TGF-β and enhances the interaction between HIPK2 and seven in absentia 2 (SIAH2), facilitating HIPK2 degradation via proteasomal pathways. Both CHR-6494 and Abemaciclib inhibit NRK-49F cell proliferation and migration induced by TGF-β, suppressing TGF-β/Smad3 signaling and decreasing profibrotic markers such as Fibronectin I (FN-I) Collagen I and α-smooth muscle actin (α-SMA). Additionally, these compounds inhibit nuclear factor kappa-B (NF-κB) signaling and reduce inflammatory cytokine expression. Conclusions: The study highlights the dual functionality of HIPK2 kinase inhibitors like CHR-6494 and Abemaciclib as promising therapeutic candidates for renal fibrosis and inflammation. The findings provide new insights into HIPK2 inhibition mechanisms and suggest pathways for the design of novel HIPK2 inhibitors in the future.
Collapse
Affiliation(s)
- Xinlan Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; (X.H.); (Y.W.); (H.O.); (J.W.); (M.Y.); (Z.C.)
- Hunan Key Laboratory of Organ Fibrosis, Changsha 410013, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410013, China
| | - Yan Wu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; (X.H.); (Y.W.); (H.O.); (J.W.); (M.Y.); (Z.C.)
- Hunan Key Laboratory of Organ Fibrosis, Changsha 410013, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410013, China
| | - Hanyi Ouyang
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; (X.H.); (Y.W.); (H.O.); (J.W.); (M.Y.); (Z.C.)
- Hunan Key Laboratory of Organ Fibrosis, Changsha 410013, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410013, China
| | - Jiayan Wu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; (X.H.); (Y.W.); (H.O.); (J.W.); (M.Y.); (Z.C.)
- Hunan Key Laboratory of Organ Fibrosis, Changsha 410013, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410013, China
| | - Mengmeng Yao
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; (X.H.); (Y.W.); (H.O.); (J.W.); (M.Y.); (Z.C.)
- Hunan Key Laboratory of Organ Fibrosis, Changsha 410013, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410013, China
| | - Zhuo Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; (X.H.); (Y.W.); (H.O.); (J.W.); (M.Y.); (Z.C.)
- Hunan Key Laboratory of Organ Fibrosis, Changsha 410013, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410013, China
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; (X.H.); (Y.W.); (H.O.); (J.W.); (M.Y.); (Z.C.)
- Hunan Key Laboratory of Organ Fibrosis, Changsha 410013, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha 410013, China
| |
Collapse
|
4
|
Zhang Q, Liu J, Liu H, Ao L, Xi Y, Chen D. Genome-wide epistasis analysis reveals gene-gene interaction network on an intermediate endophenotype P-tau/Aβ 42 ratio in ADNI cohort. Sci Rep 2024; 14:3984. [PMID: 38368488 PMCID: PMC10874417 DOI: 10.1038/s41598-024-54541-8] [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: 10/22/2023] [Accepted: 02/14/2024] [Indexed: 02/19/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia in the elderly worldwide. The exact etiology of AD, particularly its genetic mechanisms, remains incompletely understood. Traditional genome-wide association studies (GWAS), which primarily focus on single-nucleotide polymorphisms (SNPs) with main effects, provide limited explanations for the "missing heritability" of AD, while there is growing evidence supporting the important role of epistasis. In this study, we performed a genome-wide SNP-SNP interaction detection using a linear regression model and employed multiple GPUs for parallel computing, significantly enhancing the speed of whole-genome analysis. The cerebrospinal fluid (CSF) phosphorylated tau (P-tau)/amyloid-[Formula: see text] (A[Formula: see text]) ratio was used as a quantitative trait (QT) to enhance statistical power. Age, gender, and clinical diagnosis were included as covariates to control for potential non-genetic factors influencing AD. We identified 961 pairs of statistically significant SNP-SNP interactions, explaining a high-level variance of P-tau/A[Formula: see text] level, all of which exhibited marginal main effects. Additionally, we replicated 432 previously reported AD-related genes and found 11 gene-gene interaction pairs overlapping with the protein-protein interaction (PPI) network. Our findings may contribute to partially explain the "missing heritability" of AD. The identified subnetwork may be associated with synaptic dysfunction, Wnt signaling pathway, oligodendrocytes, inflammation, hippocampus, and neuronal cells.
Collapse
Affiliation(s)
- Qiushi Zhang
- School of Computer Science, Northeast Electric Power University, 169 Changchun Street, Jilin, 132012, China
| | - Junfeng Liu
- School of Computer Science, Northeast Electric Power University, 169 Changchun Street, Jilin, 132012, China
| | - Hongwei Liu
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, 145 Nantong Street, Harbin, China
| | - Lang Ao
- School of Computer Science, Northeast Electric Power University, 169 Changchun Street, Jilin, 132012, China
| | - Yang Xi
- School of Computer Science, Northeast Electric Power University, 169 Changchun Street, Jilin, 132012, China
| | - Dandan Chen
- School of Automation Engineering, Northeast Electric Power University, 169 Changchun Street, Jilin, 132012, China.
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, 145 Nantong Street, Harbin, China.
| |
Collapse
|
5
|
Pei J, Cong Q. Computational analysis of regulatory regions in human protein kinases. Protein Sci 2023; 32:e4764. [PMID: 37632170 PMCID: PMC10503413 DOI: 10.1002/pro.4764] [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: 05/09/2023] [Revised: 08/08/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
Eukaryotic proteins often feature modular domain structures comprising globular domains that are connected by linker regions and intrinsically disordered regions that may contain important functional motifs. The intramolecular interactions of globular domains and nonglobular regions can play critical roles in different aspects of protein function. However, studying these interactions and their regulatory roles can be challenging due to the flexibility of nonglobular regions, the long insertions separating interacting modules, and the transient nature of some interactions. Obtaining the experimental structures of multiple domains and functional regions is more difficult than determining the structures of individual globular domains. High-quality structural models generated by AlphaFold offer a unique opportunity to study intramolecular interactions in eukaryotic proteins. In this study, we systematically explored intramolecular interactions between human protein kinase domains (KDs) and potential regulatory regions, including globular domains, N- and C-terminal tails, long insertions, and distal nonglobular regions. Our analysis identified intramolecular interactions between human KDs and 35 different types of globular domains, exhibiting a variety of interaction modes that could contribute to orthosteric or allosteric regulation of kinase activity. We also identified prevalent interactions between human KDs and their flanking regions (N- and C-terminal tails). These interactions exhibit group-specific characteristics and can vary within each specific kinase group. Although long-range interactions between KDs and nonglobular regions are relatively rare, structural details of these interactions offer new insights into the regulation mechanisms of several kinases, such as HASPIN, MAPK7, MAPK15, and SIK1B.
Collapse
Affiliation(s)
- Jimin Pei
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Qian Cong
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| |
Collapse
|
6
|
Hogg EKJ, Findlay GM. Functions of SRPK, CLK and DYRK kinases in stem cells, development, and human developmental disorders. FEBS Lett 2023; 597:2375-2415. [PMID: 37607329 PMCID: PMC10952393 DOI: 10.1002/1873-3468.14723] [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: 06/05/2023] [Revised: 07/08/2023] [Accepted: 07/18/2023] [Indexed: 08/24/2023]
Abstract
Human developmental disorders encompass a wide range of debilitating physical conditions and intellectual disabilities. Perturbation of protein kinase signalling underlies the development of some of these disorders. For example, disrupted SRPK signalling is associated with intellectual disabilities, and the gene dosage of DYRKs can dictate the pathology of disorders including Down's syndrome. Here, we review the emerging roles of the CMGC kinase families SRPK, CLK, DYRK, and sub-family HIPK during embryonic development and in developmental disorders. In particular, SRPK, CLK, and DYRK kinase families have key roles in developmental signalling and stem cell regulation, and can co-ordinate neuronal development and function. Genetic studies in model organisms reveal critical phenotypes including embryonic lethality, sterility, musculoskeletal errors, and most notably, altered neurological behaviours arising from defects of the neuroectoderm and altered neuronal signalling. Further unpicking the mechanisms of specific kinases using human stem cell models of neuronal differentiation and function will improve our understanding of human developmental disorders and may provide avenues for therapeutic strategies.
Collapse
Affiliation(s)
- Elizabeth K. J. Hogg
- The MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life SciencesUniversity of DundeeUK
| | - Greg M. Findlay
- The MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life SciencesUniversity of DundeeUK
| |
Collapse
|
7
|
Araki S, Ohori M, Yugami M. Targeting pre-mRNA splicing in cancers: roles, inhibitors, and therapeutic opportunities. Front Oncol 2023; 13:1152087. [PMID: 37342192 PMCID: PMC10277747 DOI: 10.3389/fonc.2023.1152087] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/09/2023] [Indexed: 06/22/2023] Open
Abstract
Accumulating evidence has indicated that pre-mRNA splicing plays critical roles in a variety of physiological processes, including development of multiple diseases. In particular, alternative splicing is profoundly involved in cancer progression through abnormal expression or mutation of splicing factors. Small-molecule splicing modulators have recently attracted considerable attention as a novel class of cancer therapeutics, and several splicing modulators are currently being developed for the treatment of patients with various cancers and are in the clinical trial stage. Novel molecular mechanisms modulating alternative splicing have proven to be effective for treating cancer cells resistant to conventional anticancer drugs. Furthermore, molecular mechanism-based combination strategies and patient stratification strategies for cancer treatment targeting pre-mRNA splicing must be considered for cancer therapy in the future. This review summarizes recent progress in the relationship between druggable splicing-related molecules and cancer, highlights small-molecule splicing modulators, and discusses future perspectives of splicing modulation for personalized and combination therapies in cancer treatment.
Collapse
|
8
|
Grygier P, Pustelny K, Nowak J, Golik P, Popowicz GM, Plettenburg O, Dubin G, Menezes F, Czarna A. Silmitasertib (CX-4945), a Clinically Used CK2-Kinase Inhibitor with Additional Effects on GSK3β and DYRK1A Kinases: A Structural Perspective. J Med Chem 2023; 66:4009-4024. [PMID: 36883902 PMCID: PMC10041529 DOI: 10.1021/acs.jmedchem.2c01887] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
A clinical casein kinase 2 inhibitor, CX-4945 (silmitasertib), shows significant affinity toward the DYRK1A and GSK3β kinases, involved in down syndrome phenotypes, Alzheimer's disease, circadian clock regulation, and diabetes. This off-target activity offers an opportunity for studying the effect of the DYRK1A/GSK3β kinase system in disease biology and possible line extension. Motivated by the dual inhibition of these kinases, we solved and analyzed the crystal structures of DYRK1A and GSK3β with CX-4945. We built a quantum-chemistry-based model to rationalize the compound affinity for CK2α, DYRK1A, and GSK3β kinases. Our calculations identified a key element for CK2α's subnanomolar affinity to CX-4945. The methodology is expandable to other kinase selectivity modeling. We show that the inhibitor limits DYRK1A- and GSK3β-mediated cyclin D1 phosphorylation and reduces kinase-mediated NFAT signaling in the cell. Given the CX-4945's clinical and pharmacological profile, this inhibitory activity makes it an interesting candidate with potential for application in additional disease areas.
Collapse
Affiliation(s)
- Przemyslaw Grygier
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Krakow, Poland
| | - Katarzyna Pustelny
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Krakow, Poland
| | - Jakub Nowak
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Krakow, Poland
| | | | - Grzegorz M Popowicz
- Institute of Structural Biology, Helmholtz Zentrum Muenchen, Ingolstaedter Landstrasse 1, Neuherberg 85764, Germany
- Biomolecular NMR and Center for Integrated Protein Science Munich at Department Chemie, Technical University of Munich, Lichtenbergstrasse 4, Garching 85747, Germany
| | - Oliver Plettenburg
- Institute of Medicinal Chemistry, Helmholtz Munich, Ingolstaedter Landstrasse 1, Neuherberg 85764, Germany
- Institute of Organic Chemistry, Centre of Biomolecular Drug Research (BMWZ) and Laboratory of Nano and Quantum Engineering (LNQE), Leibniz University Hannover, Schneiderberg 1b, Hannover 30167, Germany
- German Center for Diabetes Research (DZD), Ingolstaedter Landstrasse 1, Neuherberg 85764, Germany
- Institute of Lung Health (ILH), Aulweg 130, Giessen 35392, Germany
| | - Grzegorz Dubin
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Krakow, Poland
| | - Filipe Menezes
- Institute of Structural Biology, Helmholtz Zentrum Muenchen, Ingolstaedter Landstrasse 1, Neuherberg 85764, Germany
- Biomolecular NMR and Center for Integrated Protein Science Munich at Department Chemie, Technical University of Munich, Lichtenbergstrasse 4, Garching 85747, Germany
| | - Anna Czarna
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Krakow, Poland
| |
Collapse
|
9
|
Lindberg MF, Deau E, Arfwedson J, George N, George P, Alfonso P, Corrionero A, Meijer L. Comparative Efficacy and Selectivity of Pharmacological Inhibitors of DYRK and CLK Protein Kinases. J Med Chem 2023; 66:4106-4130. [PMID: 36876904 DOI: 10.1021/acs.jmedchem.2c02068] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Dual-specificity, tyrosine phosphorylation-regulated kinases (DYRKs) and cdc2-like kinases (CLKs) play a large variety of cellular functions and are involved in several diseases (cognitive disorders, diabetes, cancers, etc.). There is, thus, growing interest in pharmacological inhibitors as chemical probes and potential drug candidates. This study presents an unbiased evaluation of the kinase inhibitory activity of a library of 56 reported DYRK/CLK inhibitors on the basis of comparative, side-by-side, catalytic activity assays on a panel of 12 recombinant human kinases, enzyme kinetics (residence time and Kd), in-cell inhibition of Thr-212-Tau phosphorylation, and cytotoxicity. The 26 most active inhibitors were modeled in the crystal structure of DYRK1A. The results show a rather large diversity of potencies and selectivities among the reported inhibitors and emphasize the difficulties to avoid "off-targets" in this area of the kinome. The use of a panel of DYRKs/CLKs inhibitors is suggested to analyze the functions of these kinases in cellular processes.
Collapse
Affiliation(s)
| | - Emmanuel Deau
- Perha Pharmaceuticals, Perharidy Peninsula, 29680 Roscoff, France
| | - Jonas Arfwedson
- Perha Pharmaceuticals, Perharidy Peninsula, 29680 Roscoff, France
| | - Nicolas George
- Oncodesign, 25-27 avenue du Québec, 91140 Villebon-sur-Yvette, France
| | - Pascal George
- Perha Pharmaceuticals, Perharidy Peninsula, 29680 Roscoff, France
| | - Patricia Alfonso
- Enzymlogic, Qube Technology Park, C/Santiago Grisolía, 2, 28760 Madrid, Spain
| | - Ana Corrionero
- Enzymlogic, Qube Technology Park, C/Santiago Grisolía, 2, 28760 Madrid, Spain
| | - Laurent Meijer
- Perha Pharmaceuticals, Perharidy Peninsula, 29680 Roscoff, France
| |
Collapse
|
10
|
Sardina F, Conte A, Paladino S, Pierantoni GM, Rinaldo C. HIPK2 in the physiology of nervous system and its implications in neurological disorders. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119465. [PMID: 36935052 DOI: 10.1016/j.bbamcr.2023.119465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/21/2023]
Abstract
HIPK2 is an evolutionary conserved serine/threonine kinase with multifunctional roles in stress response, embryonic development and pathological conditions, such as cancer and fibrosis. The heterogeneity of its interactors and targets makes HIPK2 activity strongly dependent on the cellular context, and allows it to modulate multiple signaling pathways, ultimately regulating cell fate and proliferation. HIPK2 is highly expressed in the central and peripheral nervous systems, and its genetic ablation causes neurological defects in mice. Moreover, HIPK2 is involved in processes, such as endoplasmic reticulum stress response and protein aggregate accumulation, and pathways, including TGF-β and BMP signaling, that are crucial in the pathogenesis of neurological disorders. Here, we review the data about the role of HIPK2 in neuronal development, survival, and homeostasis, highlighting the implications in the pathogenesis of neurological disorders, and pointing out HIPK2 potentiality as therapeutic target and diagnostic or prognostic marker.
Collapse
Affiliation(s)
- F Sardina
- Institute of Molecular Biology and Pathology (IBPM), Consiglio Nazionale delle Ricerche (CNR), c/o Sapienza University, Rome, Italy
| | - A Conte
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - S Paladino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - G M Pierantoni
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy.
| | - C Rinaldo
- Institute of Molecular Biology and Pathology (IBPM), Consiglio Nazionale delle Ricerche (CNR), c/o Sapienza University, Rome, Italy.
| |
Collapse
|
11
|
Yang Y, Fan X, Liu Y, Ye D, Liu C, Yang H, Su Z, Zhang Y, Liu Y. Function and Inhibition of DYRK1A: emerging roles of treating multiple human diseases. Biochem Pharmacol 2023; 212:115521. [PMID: 36990324 DOI: 10.1016/j.bcp.2023.115521] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) is an evolutionarily conserved protein kinase and the most studied member of the Dual-specificity tyrosine-regulated kinase (DYRK) family. It has been shown that it participates in the development of plenty of diseases, and both the low or high expression of DYRK1A protein could lead to disorder. Thus, DYRK1A is recognized as a key target for the therapy for these diseases, and the studies on natural or synthetic DYRK1A inhibitors have become more and more popular. Here, we provide a comprehensive review for DYRK1A from the structure and function of DYRK1A, the roles of DYRK1A in various types of diseases, including diabetes mellitus, neurodegenerative diseases, and kinds of cancers, and the studies of its natural and synthetic inhibitors.
Collapse
|
12
|
Di Segni M, Virdia I, Verdina A, Amoreo CA, Baldari S, Toietta G, Diodoro MG, Mottolese M, Sperduti I, Moretti F, Buglioni S, Soddu S, Di Rocco G. HIPK2 Cooperates with KRAS Signaling and Associates with Colorectal Cancer Progression. Mol Cancer Res 2022; 20:686-698. [PMID: 35082165 DOI: 10.1158/1541-7786.mcr-21-0628] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/25/2021] [Accepted: 01/21/2022] [Indexed: 11/16/2022]
Abstract
UNLABELLED Homeodomain-interacting protein kinase 2 (HIPK2) is an evolutionary conserved kinase that has gained attention as a fine tuner of multiple signaling pathways, among which those commonly altered in colorectal cancer. The aim of this study was to evaluate the relationship of HIPK2 expression with progression markers and mutational pattern and gain insights into the contribution of HIPK2 activity in colorectal cancer. We evaluated a retrospective cohort of colorectal cancer samples by IHC for HIPK2 expression and by next-generation sequencing (NGS) for the detection of mutations of cancer associated genes. We show that the percentage of HIPK2-positive cells increases with tumor progression, significantly correlates with tumor-node-metastasis (TNM) staging and associates with a worse outcome. In addition, we observed that high HIPK2 expression significantly associates with KRAS mutations but not with other cancer-related genes. Functional characterization of the link between HIPK2 and KRAS show that activation of the RAS pathway either due to KRAS mutation or via upstream receptor stimulation, increases HIPK2 expression at the protein level. Of note, HIPK2 physically participates in the active RAS complex while HIPK2 depletion impairs ERK phosphorylation and the growth of tumors derived from KRAS mutated colorectal cancer cells. Overall, this study identifies HIPK2 as a prognostic biomarker candidate in patients with colorectal cancer and underscores a previously unknown functional link between HIPK2 and the KRAS signaling pathway. IMPLICATIONS Our data indicate HIPK2 as a new player in the complex picture of the KRAS signaling network, providing rationales for future clinical studies and new treatment strategies for KRAS mutated colorectal cancer.
Collapse
Affiliation(s)
- Micol Di Segni
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Department of Science, Roma Tre University, Rome, Italy
| | - Ilaria Virdia
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alessandra Verdina
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Carla Azzurra Amoreo
- Pathology Division, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Baldari
- Unit of Tumor Immunology and Immunotherapy, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Gabriele Toietta
- Unit of Tumor Immunology and Immunotherapy, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Grazia Diodoro
- Pathology Division, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marcella Mottolese
- Pathology Division, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Isabella Sperduti
- Clinical Trial Center, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Fabiola Moretti
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Monterotondo, Italy
| | - Simonetta Buglioni
- Pathology Division, Biostatistics and Bioinformatic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Soddu
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giuliana Di Rocco
- Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
13
|
Hu L, Wang G, Zhao C, Peng Z, Tao L, Chen Z, Hu G, Li Q. Identification of selective homeodomain interacting protein kinase 2 inhibitors, a potential treatment for renal fibrosis. Bioorg Chem 2022; 126:105866. [DOI: 10.1016/j.bioorg.2022.105866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/25/2022] [Accepted: 05/06/2022] [Indexed: 11/16/2022]
|
14
|
Kaltheuner IH, Anand K, Moecking J, Düster R, Wang J, Gray NS, Geyer M. Abemaciclib is a potent inhibitor of DYRK1A and HIP kinases involved in transcriptional regulation. Nat Commun 2021; 12:6607. [PMID: 34785661 PMCID: PMC8595372 DOI: 10.1038/s41467-021-26935-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 10/28/2021] [Indexed: 11/09/2022] Open
Abstract
Homeodomain-interacting protein kinases (HIPKs) belong to the CMGC kinase family and are closely related to dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs). HIPKs are regulators of various signaling pathways and involved in the pathology of cancer, chronic fibrosis, diabetes, and multiple neurodegenerative diseases. Here, we report the crystal structure of HIPK3 in its apo form at 2.5 Å resolution. Recombinant HIPKs and DYRK1A are auto-activated and phosphorylate the negative elongation factor SPT5, the transcription factor c-Myc, and the C-terminal domain of RNA polymerase II, suggesting a direct function in transcriptional regulation. Based on a database search, we identified abemaciclib, an FDA-approved Cdk4/Cdk6 inhibitor used for the treatment of metastatic breast cancer, as potent inhibitor of HIPK2, HIPK3, and DYRK1A. We determined the crystal structures of HIPK3 and DYRK1A bound to abemaciclib, showing a similar binding mode to the hinge region of the kinase as observed for Cdk6. Remarkably, DYRK1A is inhibited by abemaciclib to the same extent as Cdk4/Cdk6 in vitro, raising the question of whether targeting of DYRK1A contributes to the transcriptional inhibition and therapeutic activity of abemaciclib.
Collapse
Affiliation(s)
| | - Kanchan Anand
- Institute of Structural Biology, University of Bonn, Bonn, Germany
| | - Jonas Moecking
- Institute of Structural Biology, University of Bonn, Bonn, Germany
| | - Robert Düster
- Institute of Structural Biology, University of Bonn, Bonn, Germany
| | - Jinhua Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Nathanael S Gray
- Department of Chemical and Systems Biology, Chem-H and the Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | - Matthias Geyer
- Institute of Structural Biology, University of Bonn, Bonn, Germany.
| |
Collapse
|
15
|
Caescu CI, Hansen J, Crockett B, Xiao W, Arnaud P, Spronck B, Weinberg A, Hashimoto T, Murtada SI, Borkar R, Gallo JM, Jondeau G, Boileau C, Humphrey JD, He JC, Iyengar R, Ramirez F. Inhibition of HIPK2 Alleviates Thoracic Aortic Disease in Mice With Progressively Severe Marfan Syndrome. Arterioscler Thromb Vasc Biol 2021; 41:2483-2493. [PMID: 34320838 PMCID: PMC8530207 DOI: 10.1161/atvbaha.121.316464] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective Despite considerable research, the goal of finding nonsurgical remedies against thoracic aortic aneurysm and acute aortic dissection remains elusive. We sought to identify a novel aortic PK (protein kinase) that can be pharmacologically targeted to mitigate aneurysmal disease in a well-established mouse model of early-onset progressively severe Marfan syndrome (MFS). Approach and Results Computational analyses of transcriptomic data derived from the ascending aorta of MFS mice predicted a probable association between thoracic aortic aneurysm and acute aortic dissection development and the multifunctional, stress-activated HIPK2 (homeodomain-interacting protein kinase 2). Consistent with this prediction, Hipk2 gene inactivation significantly extended the survival of MFS mice by slowing aneurysm growth and delaying transmural rupture. HIPK2 also ranked among the top predicted PKs in computational analyses of DEGs (differentially expressed genes) in the dilated aorta of 3 MFS patients, which strengthened the clinical relevance of the experimental finding. Additional in silico analyses of the human and mouse data sets identified the TGF (transforming growth factor)-β/Smad3 signaling pathway as a potential target of HIPK2 in the MFS aorta. Chronic treatment of MFS mice with an allosteric inhibitor of HIPK2-mediated stimulation of Smad3 signaling validated this prediction by mitigating thoracic aortic aneurysm and acute aortic dissection pathology and partially improving aortic material stiffness. Conclusions HIPK2 is a previously unrecognized determinant of aneurysmal disease and an attractive new target for antithoracic aortic aneurysm and acute aortic dissection multidrug therapy.
Collapse
MESH Headings
- Adult
- Aortic Dissection/enzymology
- Aortic Dissection/genetics
- Aortic Dissection/pathology
- Aortic Dissection/prevention & control
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/enzymology
- Aorta, Thoracic/pathology
- Aortic Aneurysm, Thoracic/enzymology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/prevention & control
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Dilatation, Pathologic
- Disease Models, Animal
- Disease Progression
- Fibrillin-1/genetics
- Humans
- Male
- Marfan Syndrome/complications
- Marfan Syndrome/genetics
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Protein Kinase Inhibitors/pharmacology
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Severity of Illness Index
- Signal Transduction
- Smad3 Protein/metabolism
- Vascular Remodeling/drug effects
- Mice
Collapse
Affiliation(s)
- Cristina I Caescu
- Department of Pharmacological Sciences, Institute for Systems Biomedicine (C.I.C., J.H., B.C., T.H., R.I., F.R.), Icahn School of Medicine at Mount Sinai, New York
| | - Jens Hansen
- Department of Pharmacological Sciences, Institute for Systems Biomedicine (C.I.C., J.H., B.C., T.H., R.I., F.R.), Icahn School of Medicine at Mount Sinai, New York
| | - Brittany Crockett
- Department of Pharmacological Sciences, Institute for Systems Biomedicine (C.I.C., J.H., B.C., T.H., R.I., F.R.), Icahn School of Medicine at Mount Sinai, New York
| | - Wenzhen Xiao
- Division of Nephrology, Department of Medicine (W.X., J.C.H.), Icahn School of Medicine at Mount Sinai, New York
| | - Pauline Arnaud
- Département de Génétique et Centre de Référence Maladies Rares Syndrome de Marfan et Pathologies Apparentées, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, France (P.A., G.J., C.B.)
- LVTS, INSERM U1148, Université de Paris, Hôpital Bichat, France (P.A., G.J., C.B.)
| | - Bart Spronck
- Department of Biomedical Engineering, Yale University, New Haven, CT (B.S., S.-I.M., J.D.H.)
| | - Alan Weinberg
- Department of Population Health Science and Policy (A.W.), Icahn School of Medicine at Mount Sinai, New York
| | - Takeshi Hashimoto
- Department of Pharmacological Sciences, Institute for Systems Biomedicine (C.I.C., J.H., B.C., T.H., R.I., F.R.), Icahn School of Medicine at Mount Sinai, New York
| | - Sae-Il Murtada
- Department of Biomedical Engineering, Yale University, New Haven, CT (B.S., S.-I.M., J.D.H.)
| | - Roshan Borkar
- Department of Pharmaceutical Sciences, State University of New York, Buffalo (R.B., J.M.G.)
| | - James M Gallo
- Department of Pharmaceutical Sciences, State University of New York, Buffalo (R.B., J.M.G.)
| | - Guillaume Jondeau
- Département de Génétique et Centre de Référence Maladies Rares Syndrome de Marfan et Pathologies Apparentées, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, France (P.A., G.J., C.B.)
- LVTS, INSERM U1148, Université de Paris, Hôpital Bichat, France (P.A., G.J., C.B.)
| | - Catherine Boileau
- Département de Génétique et Centre de Référence Maladies Rares Syndrome de Marfan et Pathologies Apparentées, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, France (P.A., G.J., C.B.)
- LVTS, INSERM U1148, Université de Paris, Hôpital Bichat, France (P.A., G.J., C.B.)
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT (B.S., S.-I.M., J.D.H.)
| | - John Cijiang He
- Division of Nephrology, Department of Medicine (W.X., J.C.H.), Icahn School of Medicine at Mount Sinai, New York
| | - Ravi Iyengar
- Department of Pharmacological Sciences, Institute for Systems Biomedicine (C.I.C., J.H., B.C., T.H., R.I., F.R.), Icahn School of Medicine at Mount Sinai, New York
| | - Francesco Ramirez
- Department of Pharmacological Sciences, Institute for Systems Biomedicine (C.I.C., J.H., B.C., T.H., R.I., F.R.), Icahn School of Medicine at Mount Sinai, New York
| |
Collapse
|
16
|
Němec V, Maier L, Berger BT, Chaikuad A, Drápela S, Souček K, Knapp S, Paruch K. Highly selective inhibitors of protein kinases CLK and HIPK with the furo[3,2-b]pyridine core. Eur J Med Chem 2021; 215:113299. [PMID: 33636538 DOI: 10.1016/j.ejmech.2021.113299] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/12/2021] [Accepted: 02/12/2021] [Indexed: 12/18/2022]
Abstract
The furo [3,2-b]pyridine motif represents a relatively underexplored central pharmacophore in the area of kinase inhibitors. Herein, we report flexible synthesis of 3,5-disubstituted furo [3,2-b]pyridines that relies on chemoselective couplings of newly prepared 5-chloro-3-iodofuro [3,2-b]pyridine. This methodology allowed efficient second-generation synthesis of the state-of-the-art chemical biology probe for CLK1/2/4 MU1210, and identification of the highly selective inhibitors of HIPKs MU135 and MU1787 which are presented and characterized in this study, including the X-ray crystal structure of MU135 in HIPK2. chemical biology probe.
Collapse
Affiliation(s)
- Václav Němec
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic; International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Pekařská 53, 656 91, Brno, Czech Republic
| | - Lukáš Maier
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic; International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Pekařská 53, 656 91, Brno, Czech Republic
| | - Benedict-Tilman Berger
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences (BMLS), Goethe University Frankfurt am Main, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany; Institut für Pharmazeutische Chemie, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Apirat Chaikuad
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences (BMLS), Goethe University Frankfurt am Main, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany; Institut für Pharmazeutische Chemie, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Stanislav Drápela
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Pekařská 53, 656 91, Brno, Czech Republic; Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 65, Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Kotlářská 267/2, 611 37, Brno, Czech Republic
| | - Karel Souček
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Pekařská 53, 656 91, Brno, Czech Republic; Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 65, Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Kotlářská 267/2, 611 37, Brno, Czech Republic
| | - Stefan Knapp
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences (BMLS), Goethe University Frankfurt am Main, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany; Institut für Pharmazeutische Chemie, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Kamil Paruch
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic; International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne's University Hospital in Brno, Pekařská 53, 656 91, Brno, Czech Republic.
| |
Collapse
|
17
|
Dai Y, Kyoyama H, Yang YL, Wang Y, Liu S, Wang Y, Mao JH, Xu Z, Uematsu K, Jablons DM, You L. A novel isoform of Homeodomain-interacting protein kinase-2 promotes YAP/TEAD transcriptional activity in NSCLC cells. Oncotarget 2021; 12:173-184. [PMID: 33613845 PMCID: PMC7869571 DOI: 10.18632/oncotarget.27871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 04/03/2020] [Indexed: 11/25/2022] Open
Abstract
Homeodomain-interacting protein kinase-2 (HIPK2) can either promote or inhibit transcription depending on cellular context. In this study, we show that a new HIPK2 isoform increases TEAD reporter activity in NSCLC cells. We detected HIPK2 copy number gain in 5/6 (83.3%) NSCLC cell lines. In NSCLC patients with high HIPK2 mRNA expression in the Human Protein Atlas, the five-year survival rate is significantly lower than in patients with low expression (38% vs 47%; p = 0.047). We also found that 70/78 (89.7%) of NSCLC tissues have moderate to strong expression of the N-terminal HIPK2 protein. We detected and cloned a novel HIPK2 isoform 3 and found that its forced overexpression promotes TEAD reporter activity in NSCLC cells. Expressing HIPK2 isoform 3_K228A kinase-dead plasmid failed to increase TEAD reporter activity in NSCLC cells. Next, we showed that two siRNAs targeting HIPK2 decreased HIPK2 isoform 3 and YAP protein levels in NSCLC cells. Degradation of the YAP protein was accelerated after HIPK2 knockdown in NSCLC cells. Inhibition of HIPK2 isoform 3 decreased the mRNA expression of YAP downstream gene CTGF. The specific HIPK2 kinase inhibitor TBID decreased TEAD reporter activity, reduced cancer side populations, and inhibited tumorsphere formation of NSCLC cells. In summary, this study indicates that HIPK2 isoform 3, the main HIPK2 isoform expressed in NSCLC, promotes YAP/TEAD transcriptional activity in NSCLC cells. Our results suggest that HIPK2 isoform 3 may be a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Yuyuan Dai
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Model Animal Research Center of Nanjing University, Nanjing, Jiangsu, China
- These authors contributed equally to this work
| | - Hiroyuki Kyoyama
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Department of Pulmonary Medicine, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
- These authors contributed equally to this work
| | - Yi-Lin Yang
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- These authors contributed equally to this work
| | - Yucheng Wang
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Shu Liu
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Yinghao Wang
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Zhidong Xu
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Kazutsugu Uematsu
- Department of Pulmonary Medicine, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
| | - David M. Jablons
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Liang You
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| |
Collapse
|
18
|
Poston RG, Murphy L, Rejepova A, Ghaninejad-Esfahani M, Segales J, Mulligan K, Saha RN. Certain ortho-hydroxylated brominated ethers are promiscuous kinase inhibitors that impair neuronal signaling and neurodevelopmental processes. J Biol Chem 2020; 295:6120-6137. [PMID: 32229587 PMCID: PMC7196656 DOI: 10.1074/jbc.ra119.011138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
The developing nervous system is remarkably sensitive to environmental signals, including disruptive toxins, such as polybrominated diphenyl ethers (PBDEs). PBDEs are an environmentally pervasive class of brominated flame retardants whose neurodevelopmental toxicity mechanisms remain largely unclear. Using dissociated cortical neurons from embryonic Rattus norvegicus, we found here that chronic exposure to 6-OH-BDE-47, one of the most prevalent hydroxylated PBDE metabolites, suppresses both spontaneous and evoked neuronal electrical activity. On the basis of our previous work on mitogen-activated protein kinase (MAPK)/extracellular signal-related kinase (ERK) (MEK) biology and our observation that 6-OH-BDE-47 is structurally similar to kinase inhibitors, we hypothesized that certain hydroxylated PBDEs mediate neurotoxicity, at least in part, by impairing the MEK-ERK axis of MAPK signal transduction. We tested this hypothesis on three experimental platforms: 1) in silico, where modeling ligand-protein docking suggested that 6-OH-BDE-47 is a promiscuous ATP-competitive kinase inhibitor; 2) in vitro in dissociated neurons, where 6-OH-BDE-47 and another specific hydroxylated BDE metabolite similarly impaired phosphorylation of MEK/ERK1/2 and activity-induced transcription of a neuronal immediate early gene; and 3) in vivo in Drosophila melanogaster, where developmental exposures to 6-OH-BDE-47 and a MAPK inhibitor resulted in offspring displaying similarly increased frequency of mushroom-body β-lobe midline crossing, a metric of axonal guidance. Taken together, our results support that certain ortho-hydroxylated PBDE metabolites are promiscuous kinase inhibitors and can cause disruptions of critical neurodevelopmental processes, including neuronal electrical activity, pre-synaptic functions, MEK-ERK signaling, and axonal guidance.
Collapse
Affiliation(s)
- Robert G Poston
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Lillian Murphy
- Department of Biological Sciences, Center for Interdisciplinary Molecular Biology: Education, Research and Advancement (CIMERA), California State University, Sacramento, California 95819
| | - Ayna Rejepova
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Mina Ghaninejad-Esfahani
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Joshua Segales
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Kimberly Mulligan
- Department of Biological Sciences, Center for Interdisciplinary Molecular Biology: Education, Research and Advancement (CIMERA), California State University, Sacramento, California 95819
| | - Ramendra N Saha
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343.
| |
Collapse
|
19
|
Yeung W, Ruan Z, Kannan N. Emerging roles of the αC-β4 loop in protein kinase structure, function, evolution, and disease. IUBMB Life 2020; 72:1189-1202. [PMID: 32101380 DOI: 10.1002/iub.2253] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/07/2020] [Indexed: 12/11/2022]
Abstract
The faithful propagation of cellular signals in most organisms relies on the coordinated functions of a large family of protein kinases that share a conserved catalytic domain. The catalytic domain is a dynamic scaffold that undergoes large conformational changes upon activation. Most of these conformational changes, such as movement of the regulatory αC-helix from an "out" to "in" conformation, hinge on a conserved, but understudied, loop termed the αC-β4 loop, which mediates conserved interactions to tether flexible structural elements to the kinase core. We previously showed that the αC-β4 loop is a unique feature of eukaryotic protein kinases. Here, we review the emerging roles of this loop in kinase structure, function, regulation, and diseases. Through a kinome-wide analysis, we define the boundaries of the loop for the first time and show that sequence and structural variation in the loop correlate with conformational and regulatory variation. Many recurrent disease mutations map to the αC-β4 loop and contribute to drug resistance and abnormal kinase activation by relieving key auto-inhibitory interactions associated with αC-helix and inter-lobe movement. The αC-β4 loop is a hotspot for post-translational modifications, protein-protein interaction, and Hsp90 mediated folding. Our kinome-wide analysis provides insights for hypothesis-driven characterization of understudied kinases and the development of allosteric protein kinase inhibitors.
Collapse
Affiliation(s)
- Wayland Yeung
- Institute of Bioinformatics, University of Georgia, Athens, Georgia
| | - Zheng Ruan
- Institute of Bioinformatics, University of Georgia, Athens, Georgia
| | - Natarajan Kannan
- Institute of Bioinformatics, University of Georgia, Athens, Georgia.,Department of Biochemistry & Molecular Biology, University of Georgia, Athens, Georgia
| |
Collapse
|
20
|
Abstract
Homeodomain-interacting protein kinases (HIPKs) are kinases that phosphorylate transcription factors involved in cell proliferation, differentiation, and apoptosis. Their structures have been long sought because of their potential as drug targets in cancers and fibrosis. Agnew and colleagues present the first crystal structure of the HIPK2 kinase domain, complexed with the small-molecule inhibitor CX-4945, revealing important structural differences from related protein kinases of the DYRK family. This structure provides a starting point to exploit HIPK2's distinct structural features to develop selective small-molecule inhibitors of this kinase.
Collapse
Affiliation(s)
- James M Murphy
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|