1
|
Karmakar S, Chatterjee M, Basu M, Ghosh MK. CK2: The master regulator in tumor immune-microenvironment - A crucial target in oncotherapy. Eur J Pharmacol 2025; 994:177376. [PMID: 39952582 DOI: 10.1016/j.ejphar.2025.177376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/22/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
A constitutively active serine/threonine kinase, casein kinase 2 (CK2) is involved in several physiological functions, such as DNA repair, apoptosis, and cell cycle control. New research emphasizes how critical CK2 is to the immune system's dysregulation in the tumor immune-microenvironment (TIME). The inhibition of immunological responses, including the downregulation of immune effector cells and the elevation of immunosuppressive proteins that aid in the development of tumor and immune evasion, has been linked to CK2 overexpression. CK2 maintains an immunosuppressive milieu that impedes anti-tumor immunity by encouraging the expressions and activities of immune checkpoint markers, regulating cytokines release, and boosting immune-suppressive cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) to maintain immune evasion. It is a promising target for cancer treatment due to its complex role in immune regulation and oncogenic pathways. In this study, we address the therapeutic perspectives of targeting CK2 in oncotherapy and investigate the mechanisms by which it controls immunological responses in the TME. This review, comprehending the function of CK2 in immune suppression can facilitate the creation of innovative treatment approaches aimed at augmenting anti-tumor immunity and enhancing immunotherapy effectiveness.
Collapse
Affiliation(s)
- Subhajit Karmakar
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata, 700032, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India; 4, Raja S.C, Mullick Road, Jadavpur, Kolkata, 700032, India
| | - Mouli Chatterjee
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata, 700032, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India; 4, Raja S.C, Mullick Road, Jadavpur, Kolkata, 700032, India
| | - Malini Basu
- Department of Microbiology, Dhruba Chand Halder College, University of Calcutta, Dakshin Barasat, WB, India
| | - Mrinal K Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata, 700032, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, 201002, India; 4, Raja S.C, Mullick Road, Jadavpur, Kolkata, 700032, India.
| |
Collapse
|
2
|
Abd El Hadi SR, Eldinary MA, Ghith A, Haffez H, Salman A, Sayed GA. Unravelling the potency of the 4-oxo-2-thioxo-1,2,3,4-tetrahydropyrimidine-5-carbonitrile scaffold with S-arylamide hybrids as PIM-1 kinase inhibitors: synthesis, biological activity and in silico studies. RSC Med Chem 2025:d5md00021a. [PMID: 40162200 PMCID: PMC11951167 DOI: 10.1039/d5md00021a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
PIM-1 is a type of serine/threonine kinase that plays a crucial role in controlling several vital processes, including proliferation and apoptosis. New synthetic S-amide tetrahydropyrimidinone derivatives were designed and synthesized as PIM-1 inhibitors with potential anticancer activity. Several biochemical assays were performed for anticancer assessment, including PIM-1 inhibitory assays, MTT, apoptosis and cell cycle, gene expression analysis, c-MYC analysis, and ATPase inhibitory assays. Compounds (8c, 8d, 8g, 8h, 8k, and 8l) exhibited strong in vitro broad antiproliferative activity against MCF-7, DU-145, and PC-3, with a relatively higher SI index suggesting minimal cytotoxicity to normal cells. Furthermore, these compounds induced mixed late apoptosis and necrosis with cell cycle arrest at the G2/M phase. Moreover, compounds 8b, 8f, 8g, 8k, and 8l showed potent inhibitory action against PIM-1 kinase, with corresponding IC50 values of 660, 909, 373, 518, and 501 nM. In silico prediction studies of physiochemical properties, molecular dynamics, and induced fit docking studies were performed for these compounds to explain their potent biological activity. In conclusion, new pyrimidinone compounds (8c, 8d, 8g, 8h, 8k, and 8l) exhibit potential PIM-1 inhibitory activity and can be used as promising scaffolds for further optimization of new leads with selective PIM-inhibitors and anticancer activity.
Collapse
Affiliation(s)
- Soha R Abd El Hadi
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City Cairo 11829 Egypt
| | - Manar A Eldinary
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City Cairo 11829 Egypt
| | - Amna Ghith
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital Woodville South SA 5011 Australia
- Robinson Research Institute, University of Adelaide Adelaide SA 5006 Australia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Future University in Egypt Cairo 11835 Egypt
| | - Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University P.O. Box 11795 Cairo Egypt
- Center of Scientific Excellence "Helwan Structural Biology Research (HSBR)", Helwan University Cairo 11795 Egypt
| | - Aya Salman
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City 11829 Cairo Egypt
| | - Ghadir A Sayed
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City 11829 Cairo Egypt
| |
Collapse
|
3
|
Marminon C, Werner C, Gast A, Herfindal L, Charles J, Lindenblatt D, Aichele D, Mularoni A, Døskeland SO, Jose J, Niefind K, Le Borgne M. Exploring the biological potential of the brominated indenoindole MC11 and its interaction with protein kinase CK2. Biol Chem 2025:hsz-2024-0160. [PMID: 40116007 DOI: 10.1515/hsz-2024-0160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 02/05/2025] [Indexed: 03/23/2025]
Abstract
Protein kinase CK2 is a promising therapeutic target, especially in oncology. Over the years, various inhibitors have been developed, with polyhalogenated scaffolds emerging as a particularly effective class. Halogens like bromine and chlorine enhance inhibitor stability by forming additional interactions within the ATP pocket. Among halogenated scaffolds, benzotriazole and benzimidazole have led to potent molecules such as 4,5,6,7-tetrabromo-1H-benzotriazole (IC50 = 300 nM) and 4,5,6,7-tetrabromo-2-(dimethylamino)benzimidazole (IC50 = 140 nM). Modifications, including 4,5,6-tribromo-7-ethyl-1H-benzotriazole (IC50 = 160 nM), further improved activity. Changing scaffolds while retaining halogens has enabled design of new inhibitors. Flavonols, dibenzofuranones, and the indeno[1,2-b]indole scaffold are key examples. Halogenation of the reference molecule 5-isopropyl-5,6,7,8-tetrahydroindeno[1,2-b]indole-9,10-dione (4b, IC50 = 360 nM) significantly boosted potency. The study focused on introducing four halogens, yielding to the compound 1,2,3,4-tetrabromo-5-isopropyl-5,6,7,8-tetrahydroindeno[1,2-b]indole-9,10-dione (MC11), with an IC50 of 16 nM. Co-crystallography revealed how bromine atoms enhance binding, and MC11 demonstrated strong in cellulo activity, particularly against leukemic cell lines like IPC-Bcl2.
Collapse
Affiliation(s)
- Christelle Marminon
- Small Molecules for Biological Targets Team, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, University of Lyon, F-69373 Lyon, France
| | - Christian Werner
- Department of Chemistry and Biochemistry, Institute of Biochemistry, University of Cologne, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Alexander Gast
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, PharmaCampus, Corrensstraße 48, D-48149 Münster, Germany
| | - Lars Herfindal
- Department of Clinical Science, University of Bergen, N-5009 Bergen, Norway
| | - Johana Charles
- Small Molecules for Biological Targets Team, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, University of Lyon, F-69373 Lyon, France
| | - Dirk Lindenblatt
- Department of Chemistry and Biochemistry, Institute of Biochemistry, University of Cologne, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Dagmar Aichele
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, PharmaCampus, Corrensstraße 48, D-48149 Münster, Germany
| | - Angélique Mularoni
- Small Molecules for Biological Targets Team, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, University of Lyon, F-69373 Lyon, France
| | | | - Joachim Jose
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, PharmaCampus, Corrensstraße 48, D-48149 Münster, Germany
| | - Karsten Niefind
- Department of Chemistry and Biochemistry, Institute of Biochemistry, University of Cologne, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Marc Le Borgne
- Small Molecules for Biological Targets Team, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, University of Lyon, F-69373 Lyon, France
| |
Collapse
|
4
|
Götz C, Montenarh M. Protein kinase CK2 contributes to glucose homeostasis. Biol Chem 2025:hsz-2024-0158. [PMID: 39910713 DOI: 10.1515/hsz-2024-0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/22/2025] [Indexed: 02/07/2025]
Abstract
In the early days of CK2 research, it was already published that the affinity of CK2 for its substrate casein was affected by insulin. Subsequent to the discovery of inhibitors of CK2 kinase activity, it was shown that CK2 has an influence on hormones that regulate glucose homeostasis and on enzymes that influence glucose metabolism in pancreatic islet cells as well as in hormone-sensitive target cells. This regulation includes the influence on transcription factors and thereby, gene expression, as well as direct modulation of the catalytic activity. The used CK2 inhibitors, especially the older ones, show a broad range of specificity, selectivity and off-target effects. Recently applied methods to down-regulate the expression of individual CK2 subunits using siRNA or CRISPR/Cas9 technology have contributed to the improvement of specificity. It was shown that inhibition of CK2 kinase activity or knock-down or knock-out of CK2α leads to an elevated synthesis and secretion of insulin in pancreatic β-cells and a down-regulation of the synthesis and secretion of glucagon from pancreatic α-cells. In the present review CK2-dependent molecular mechanisms will be addressed which contribute to the maintenance of glucose homeostasis.
Collapse
Affiliation(s)
- Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University, Building 44, D-66421 Homburg, Germany
| | - Mathias Montenarh
- Medical Biochemistry and Molecular Biology, Saarland University, Building 44, D-66421 Homburg, Germany
| |
Collapse
|
5
|
Khalifa H, ElHady AK, Liu T, Elgaher WAM, Filhol-Cochet O, Cochet C, Abadi AH, Hamed MM, Abdel-Halim M, Engel M. Discovery of a novel, selective CK2 inhibitor class with an unusual basic scaffold. Eur J Med Chem 2025; 282:117048. [PMID: 39566243 DOI: 10.1016/j.ejmech.2024.117048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
CK2 is a Ser/Thr-protein kinase playing a crucial role in promoting cell growth and survival, hence it is considered a promising target for anti-cancer drugs. However, many previously reported CK2 inhibitors lack selectivity. In search of novel scaffolds for selective CK2 inhibition, we identified a dihydropyrido-thieno[2,3-d]pyrimidine derivative displaying submicromolar inhibitory activity against CK2α. This scaffold captured our interest because of the basic secondary amine, a rather unusual motif for CK2 inhibitors. Our optimization strategy comprised the incorporation of a 4-piperazinyl moiety as a linker group and introduction of varying substituents on the pendant phenyl ring. All resulting compounds exhibited potent CK2α inhibition, with IC50 values in the nanomolar range. Compound 10b demonstrated the most balanced activity profile with a cell-free IC50 value of 36.7 nM and a notable cellular activity with a GI50 of 7.3 μM and 7.5 μM against 786-O renal cell carcinoma and U937 lymphoma cells, respectively. 10b displayed excellent selectivity when screened against a challenging kinase selectivity profiling panel. Moreover, 10b inhibited CK2 in the cells, albeit less potently than CX-4945, but induced cell death more strongly than CX-4945. Altogether, we have identified a novel CK2 inhibitory scaffold with drug-like physicochemical properties in a favorable basic pKa range.
Collapse
Affiliation(s)
- Hend Khalifa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt
| | - Ahmed K ElHady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt; School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, New Administrative Capital, Cairo, Egypt
| | - Ting Liu
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, 66123, Saarbrücken, Germany
| | - Walid A M Elgaher
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123, Saarbrücken, Germany
| | - Odile Filhol-Cochet
- University Grenoble Alpes, INSERM 1292, CEA, UMR Biosante, 38000, Grenoble, France
| | - Claude Cochet
- University Grenoble Alpes, INSERM 1292, CEA, UMR Biosante, 38000, Grenoble, France
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt
| | - Mostafa M Hamed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123, Saarbrücken, Germany
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt.
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, 66123, Saarbrücken, Germany.
| |
Collapse
|
6
|
Bova V, Mannino D, Capra AP, Lanza M, Palermo N, Filippone A, Esposito E. CK and LRRK2 Involvement in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:11661. [PMID: 39519213 PMCID: PMC11546471 DOI: 10.3390/ijms252111661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/21/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Neurodegenerative diseases (NDDs) are currently the most widespread neuronal pathologies in the world. Among these, the most widespread are Alzheimer's disease (AD), dementia, Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD)-all characterized by a progressive loss of neurons in specific regions of the brain leading to varied clinical symptoms. At the basis of neurodegenerative diseases, an emerging role is played by genetic mutations in the leucine-rich repeat kinase 2 (LRRK2) gene that cause increased LRRK2 activity with consequent alteration of neuronal autophagy pathways. LRRK2 kinase activity requires GTPase activity which functions independently of kinase activity and is required for neurotoxicity and to potentiate neuronal death. Important in the neurodegeneration process is the upregulation of casein kinase (CK), which causes the alteration of the AMPK pathway by enhancing the phosphorylation of α-synuclein and huntingtin proteins, known to be involved in PD and HD, and increasing the accumulation of the amyloid-β protein (Aβ) for AD. Recent research has identified CK of the kinases upstream of LRRK2 as a regulator of the stability of the LRRK2 protein. Based on this evidence, this review aims to understand the direct involvement of individual kinases in NDDs and how their crosstalk may impact the pathogenesis and early onset of neurodegenerative diseases.
Collapse
Affiliation(s)
- Valentina Bova
- Department of Chemical, Biological, Pharmaceuticals and Environmental Sciences, University of Messina, Viale Stagno d’Alcontres, 98166 Messina, Italy; (V.B.); (D.M.); (A.P.C.); (M.L.); (E.E.)
| | - Deborah Mannino
- Department of Chemical, Biological, Pharmaceuticals and Environmental Sciences, University of Messina, Viale Stagno d’Alcontres, 98166 Messina, Italy; (V.B.); (D.M.); (A.P.C.); (M.L.); (E.E.)
| | - Anna Paola Capra
- Department of Chemical, Biological, Pharmaceuticals and Environmental Sciences, University of Messina, Viale Stagno d’Alcontres, 98166 Messina, Italy; (V.B.); (D.M.); (A.P.C.); (M.L.); (E.E.)
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceuticals and Environmental Sciences, University of Messina, Viale Stagno d’Alcontres, 98166 Messina, Italy; (V.B.); (D.M.); (A.P.C.); (M.L.); (E.E.)
| | - Nicoletta Palermo
- Department of Biochemical, Dental, Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmaceuticals and Environmental Sciences, University of Messina, Viale Stagno d’Alcontres, 98166 Messina, Italy; (V.B.); (D.M.); (A.P.C.); (M.L.); (E.E.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceuticals and Environmental Sciences, University of Messina, Viale Stagno d’Alcontres, 98166 Messina, Italy; (V.B.); (D.M.); (A.P.C.); (M.L.); (E.E.)
| |
Collapse
|
7
|
Latosińska M, Latosińska JN. Serine/Threonine Protein Kinases as Attractive Targets for Anti-Cancer Drugs-An Innovative Approach to Ligand Tuning Using Combined Quantum Chemical Calculations, Molecular Docking, Molecular Dynamic Simulations, and Network-like Similarity Graphs. Molecules 2024; 29:3199. [PMID: 38999151 PMCID: PMC11243552 DOI: 10.3390/molecules29133199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024] Open
Abstract
Serine/threonine protein kinases (CK2, PIM-1, RIO1) are constitutively active, highly conserved, pleiotropic, and multifunctional kinases, which control several signaling pathways and regulate many cellular functions, such as cell activity, survival, proliferation, and apoptosis. Over the past decades, they have gained increasing attention as potential therapeutic targets, ranging from various cancers and neurological, inflammation, and autoimmune disorders to viral diseases, including COVID-19. Despite the accumulation of a vast amount of experimental data, there is still no "recipe" that would facilitate the search for new effective kinase inhibitors. The aim of our study was to develop an effective screening method that would be useful for this purpose. A combination of Density Functional Theory calculations and molecular docking, supplemented with newly developed quantitative methods for the comparison of the binding modes, provided deep insight into the set of desirable properties responsible for their inhibition. The mathematical metrics helped assess the distance between the binding modes, while heatmaps revealed the locations in the ligand that should be modified according to binding site requirements. The Structure-Binding Affinity Index and Structural-Binding Affinity Landscape proposed in this paper helped to measure the extent to which binding affinity is gained or lost in response to a relatively small change in the ligand's structure. The combination of the physico-chemical profile with the aforementioned factors enabled the identification of both "dead" and "promising" search directions. Tests carried out on experimental data have validated and demonstrated the high efficiency of the proposed innovative approach. Our method for quantifying differences between the ligands and their binding capabilities holds promise for guiding future research on new anti-cancer agents.
Collapse
Affiliation(s)
- Magdalena Latosińska
- Faculty of Physics, Adam Mickiewicz University, Uniwersytetu Poznańskiego 2, 61-814 Poznań, Poland
| | | |
Collapse
|
8
|
Franci L, Vallini G, Bertolino FM, Cicaloni V, Inzalaco G, Cicogni M, Tinti L, Calabrese L, Barone V, Salvini L, Rubegni P, Galvagni F, Chiariello M. MAPK15 controls cellular responses to oxidative stress by regulating NRF2 activity and expression of its downstream target genes. Redox Biol 2024; 72:103131. [PMID: 38555711 PMCID: PMC10998232 DOI: 10.1016/j.redox.2024.103131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024] Open
Abstract
Oxidation processes in mitochondria and different environmental insults contribute to unwarranted accumulation of reactive oxygen species (ROS). These, in turn, rapidly damage intracellular lipids, proteins, and DNA, ultimately causing aging and several human diseases. Cells have developed different and very effective systems to control ROS levels. Among these, removal of excessive amounts is guaranteed by upregulated expression of various antioxidant enzymes, through activation of the NF-E2-Related Factor 2 (NRF2) protein. Here, we show that Mitogen Activated Protein Kinase 15 (MAPK15) controls the transactivating potential of NRF2 and, in turn, the expression of its downstream target genes. Specifically, upon oxidative stress, MAPK15 is necessary to increase NRF2 expression and nuclear translocation, by inducing its activating phosphorylation, ultimately supporting transactivation of cytoprotective antioxidant genes. Lungs are continuously exposed to oxidative damages induced by environmental insults such as air pollutants and cigarette smoke. Interestingly, we demonstrate that MAPK15 is very effective in supporting NRF2-dependent antioxidant transcriptional response to cigarette smoke of epithelial lung cells. Oxidative damage induced by cigarette smoke indeed represents a leading cause of disability and death worldwide by contributing to the pathogenesis of different chronic respiratory diseases and lung cancer. Therefore, the development of novel therapeutic strategies able to modulate cellular responses to oxidative stress would be highly beneficial. Our data contribute to the necessary understanding of the molecular mechanisms behind such responses and identify new potentially actionable targets.
Collapse
Affiliation(s)
- Lorenzo Franci
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale Delle Ricerche (CNR), Siena, Italy; Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Siena, Italy.
| | - Giulia Vallini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy; Section of Dermatology, Department of Medical, Surgical and Neurological Science, University of Siena, Italy.
| | - Franca Maria Bertolino
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale Delle Ricerche (CNR), Siena, Italy; Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Siena, Italy; Department of Medical Biotechnologies, University of Siena, Siena, Italy.
| | | | - Giovanni Inzalaco
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale Delle Ricerche (CNR), Siena, Italy; Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Siena, Italy; Department of Medical Biotechnologies, University of Siena, Siena, Italy.
| | | | - Laura Tinti
- Toscana Life Sciences Foundation, Siena, Italy.
| | - Laura Calabrese
- Section of Dermatology, Department of Medical, Surgical and Neurological Science, University of Siena, Italy.
| | - Virginia Barone
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy.
| | | | - Pietro Rubegni
- Section of Dermatology, Department of Medical, Surgical and Neurological Science, University of Siena, Italy.
| | - Federico Galvagni
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Italy.
| | - Mario Chiariello
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale Delle Ricerche (CNR), Siena, Italy; Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Siena, Italy.
| |
Collapse
|
9
|
Chesnokova LS, Mosher BS, Fulkerson HL, Nam HW, Shakya AK, Yurochko AD. Distinct early role of PTEN regulation during HCMV infection of monocytes. Proc Natl Acad Sci U S A 2024; 121:e2312290121. [PMID: 38483999 PMCID: PMC10962971 DOI: 10.1073/pnas.2312290121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/01/2023] [Indexed: 03/19/2024] Open
Abstract
Human cytomegalovirus (HCMV) infection of monocytes is essential for viral dissemination and persistence. We previously identified that HCMV entry/internalization and subsequent productive infection of this clinically relevant cell type is distinct when compared to other infected cells. We showed that internalization and productive infection required activation of epidermal growth factor receptor (EGFR) and integrin/c-Src, via binding of viral glycoprotein B to EGFR, and the pentamer complex to β1/β3 integrins. To understand how virus attachment drives entry, we compared infection of monocytes with viruses containing the pentamer vs. those without the pentamer and then used a phosphoproteomic screen to identify potential phosphorylated proteins that influence HCMV entry and trafficking. The screen revealed that the most prominent pentamer-biased phosphorylated protein was the lipid- and protein-phosphatase phosphatase and tensin homolog (PTEN). PTEN knockdown with siRNA or PTEN inhibition with a PTEN inhibitor decreased pentamer-mediated HCMV entry, without affecting trimer-mediated entry. Inhibition of PTEN activity affected lipid metabolism and interfered with the onset of the endocytic processes required for HCMV entry. PTEN inactivation was sufficient to rescue pentamer-null HCMV from lysosomal degradation. We next examined dephosphorylation of a PTEN substrate Rab7, a regulator of endosomal maturation. Inhibition of PTEN activity prevented dephosphorylation of Rab7. Phosphorylated Rab7, in turn, blocked early endosome to late endosome maturation and promoted nuclear localization of the virus and productive infection.
Collapse
Affiliation(s)
- Liudmila S. Chesnokova
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA71103
- Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA71103
| | - Bailey S. Mosher
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA71103
- Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA71103
| | - Heather L. Fulkerson
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA71103
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA71103
| | - Hyung W. Nam
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA71103
| | - Akhalesh K. Shakya
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA71103
| | - Andrew D. Yurochko
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA71103
- Center for Applied Immunology and Pathological Processes, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA71103
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA71103
- Feist-Weller Cancer Center, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA 71103, Shreveport, LA71103
- Center for Excellence in Arthritis and Rheumatology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA71103
- Center of Excellence for Emerging Viral Threats, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA71103
| |
Collapse
|
10
|
Papp B, Le Borgne M, Perret F, Marminon C, Józsa L, Pető Á, Kósa D, Nagy L, Kéki S, Ujhelyi Z, Pallér Á, Budai I, Bácskay I, Fehér P. Formulation and Investigation of CK2 Inhibitor-Loaded Alginate Microbeads with Different Excipients. Pharmaceutics 2023; 15:2701. [PMID: 38140042 PMCID: PMC10748227 DOI: 10.3390/pharmaceutics15122701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
The aim of this study was to formulate and characterize CK2 inhibitor-loaded alginate microbeads via the polymerization method. Different excipients were used in the formulation to improve the penetration of an active agent and to stabilize our preparations. Transcutol® HP was added to the drug-sodium alginate mixture and polyvinylpyrrolidone (PVP) was added to the hardening solution, alone and in combination. To characterize the formulations, mean particle size, scanning electron microscopy analysis, encapsulation efficiency, swelling behavior, an enzymatic stability test and an in vitro dissolution study were performed. The cell viability assay and permeability test were also carried out on the Caco-2 cell line. The anti-oxidant and anti-inflammatory effects of the formulations were finally evaluated. The combination of Transcutol® HP and PVP in the formulation of sodium alginate microbeads could improve the stability, in vitro permeability, anti-oxidant and anti-inflammatory effects of the CK2 inhibitor.
Collapse
Affiliation(s)
- Boglárka Papp
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary; (B.P.); (L.J.); (Á.P.); (D.K.); (Z.U.); (Á.P.); (I.B.)
- Institute of Healthcare Industry, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary
| | - Marc Le Borgne
- Small Molecules for Biological Targets Team, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, Univ Lyon, 69373 Lyon, France; (M.L.B.); (C.M.)
| | - Florent Perret
- Univ Lyon, Université Lyon 1, CNRS, INSA, CPE, ICBMS, 69622 Lyon, France;
| | - Christelle Marminon
- Small Molecules for Biological Targets Team, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, Univ Lyon, 69373 Lyon, France; (M.L.B.); (C.M.)
| | - Liza Józsa
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary; (B.P.); (L.J.); (Á.P.); (D.K.); (Z.U.); (Á.P.); (I.B.)
- Institute of Healthcare Industry, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary
| | - Ágota Pető
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary; (B.P.); (L.J.); (Á.P.); (D.K.); (Z.U.); (Á.P.); (I.B.)
- Institute of Healthcare Industry, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary
| | - Dóra Kósa
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary; (B.P.); (L.J.); (Á.P.); (D.K.); (Z.U.); (Á.P.); (I.B.)
- Institute of Healthcare Industry, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary
| | - Lajos Nagy
- Department of Applied Chemistry, Faculty of Science and Technology, Institute of Chemistry, University of Debrecen, Egyetem Tér 1, H-4032 Debrecen, Hungary; (L.N.); (S.K.)
| | - Sándor Kéki
- Department of Applied Chemistry, Faculty of Science and Technology, Institute of Chemistry, University of Debrecen, Egyetem Tér 1, H-4032 Debrecen, Hungary; (L.N.); (S.K.)
| | - Zoltán Ujhelyi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary; (B.P.); (L.J.); (Á.P.); (D.K.); (Z.U.); (Á.P.); (I.B.)
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary
| | - Ádám Pallér
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary; (B.P.); (L.J.); (Á.P.); (D.K.); (Z.U.); (Á.P.); (I.B.)
| | - István Budai
- Faculty of Engineering, University of Debrecen, Ótemető Utca 2–4, H-4028 Debrecen, Hungary;
| | - Ildikó Bácskay
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary; (B.P.); (L.J.); (Á.P.); (D.K.); (Z.U.); (Á.P.); (I.B.)
- Institute of Healthcare Industry, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary
| | - Pálma Fehér
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary; (B.P.); (L.J.); (Á.P.); (D.K.); (Z.U.); (Á.P.); (I.B.)
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary
| |
Collapse
|
11
|
Patel S, Vyas VK, Sharma M, Ghate M. Structure-guided discovery of adenosine triphosphate-competitive casein kinase 2 inhibitors. Future Med Chem 2023; 15:987-1014. [PMID: 37307219 DOI: 10.4155/fmc-2023-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023] Open
Abstract
Casein kinase 2 (CK2) is a ubiquitous, highly pleiotropic serine-threonine kinase. CK2 has been identified as a potential drug target for the treatment of cancer and related disorders. Several adenosine triphosphate-competitive CK2 inhibitors have been identified and have progressed at different levels of clinical trials. This review presents details of CK2 protein, structural insights into adenosine triphosphate binding pocket, current clinical trial candidates and their analogues. Further, it includes the emerging structure-based drug design approaches, chemistry, structure-activity relationship and biological screening of potent and selective CK2 inhibitors. The authors tabulated the details of CK2 co-crystal structures because these co-crystal structures facilitated the structure-guided discovery of CK2 inhibitors. The narrow hinge pocket compared with related kinases provides useful insights into the discovery of CK2 inhibitors.
Collapse
Affiliation(s)
- Shivani Patel
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Manmohan Sharma
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Manjunath Ghate
- School of Pharmacy, National Forensic Science University, Gandhinagar, Gujarat, 382007, India
| |
Collapse
|
12
|
Patel S, Patel S, Tulsian K, Kumar P, Vyas VK, Ghate M. Design of 2-amino-6-methyl-pyrimidine benzoic acids as ATP competitive casein kinase-2 (CK2) inhibitors using structure- and fragment-based design, docking and molecular dynamic simulation studies. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2023; 34:211-230. [PMID: 37051759 DOI: 10.1080/1062936x.2023.2196091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Overexpression of casein kinase-2 (CK2) has been implicated in several carcinomas, mainly lung, prostate and acute myeloid leukaemia. The smaller nucleotide pocket compared to related kinases provides a great opportunity to discover newer ATP-competitive CK2 inhibitors. In this study, we have employed an integrated structure- and fragment-based design strategy to design 2-amino-6-methyl-pyrimidine benzoic acids as ATP-competitive CK2 inhibitors. A statistically significant four features-based E-pharmacophore (ARRR) model was used to screen 780,092 molecules. Further, the retrieved hits were considered for molecular docking study to identify essential binding interactions. At the same time, fragment-based virtual screening was performed using a dataset of 1,542,397 fragments. The identified hits and fragments were used as structure templates to rationalize the design of 2-amino-6-methyl-pyrimidine benzoic acids as newer CK2 inhibitors. Finally, the binding interactions of the designed hits were identified using an induced fit docking (IFD) study, and their stability was estimated by a molecular dynamics (MD) simulation study of 100 ns.
Collapse
Affiliation(s)
- S Patel
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - S Patel
- Department of Botany, Bioinformatics and Climate Change Impacts Management, Gujarat University, Ahmedabad, India
| | - K Tulsian
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - P Kumar
- Department of Botany, Bioinformatics and Climate Change Impacts Management, Gujarat University, Ahmedabad, India
| | - V K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - M Ghate
- School of Pharmacy, National Forensic Science University, Gandhinagar, India
| |
Collapse
|
13
|
Yadav S, Ahamad S, Gupta D, Mathur P. Lead optimization, pharmacophore development and scaffold design of protein kinase CK2 inhibitors as potential COVID-19 therapeutics. J Biomol Struct Dyn 2023; 41:1811-1827. [PMID: 35014595 DOI: 10.1080/07391102.2021.2024449] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Therapeutic agents being designed against COVID-19 have targeted either the virus directly or the host cellular machinery. A particularly attractive host target is the ubiquitous and constitutively active serine-threonine kinase, Protein kinase CK2 (CK2). CK2 enhances viral protein synthesis by inhibiting the sequestration of host translational machinery as stress granules and assists in viral egression via association with the N-protein at filopodial protrusions of the infected cell. CK2 inhibitors such as Silmitasertib have been proposed as possible therapeutic candidates in COVID-19 infections. The present study aims to optimize Silmitasertib, develop pharmacophore models and design unique scaffolds to modulate CK2. The lead optimization phase involved the generation of compounds structurally similar to Silmitasertib via bioisostere replacement followed by a multi-stage docking approach to identify drug-like candidates. Molecular dynamics (MD) simulations were performed for two promising candidates (ZINC-43206125 and PC-57664175) to estimate their binding stability and interaction. Top scoring candidates from the lead optimization phase were utilized to build ligand-based pharmacophore models. These models were then merged with structure-based pharmacophores (e-pharmacophores) to build a hybrid hypothesis. This hybrid hypothesis was validated against a decoy set and used to screen a diverse kinase inhibitors library to identify favored chemical features in the retrieved actives. These chemical features include; an anion, an aromatic ring and an H-bond acceptor. Based on the knowledge of these features; de-novo scaffold design was carried out which identified phenindiones, carboxylated steroids, macrocycles and peptides as novel scaffolds with the potential to modulate CK2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Siddharth Yadav
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Shahzaib Ahamad
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Puniti Mathur
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| |
Collapse
|
14
|
Thus YJ, De Rooij MFM, Swier N, Beijersbergen RL, Guikema JEJ, Kersten MJ, Eldering E, Pals ST, Kater AP, Spaargaren M. Inhibition of casein kinase 2 sensitizes mantle cell lymphoma to venetoclax through MCL-1 downregulation. Haematologica 2023; 108:797-810. [PMID: 36226498 PMCID: PMC9973496 DOI: 10.3324/haematol.2022.281668] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Indexed: 11/09/2022] Open
Abstract
BCL-2 family proteins are frequently aberrantly expressed in mantle cell lymphoma (MCL). Recently, the BCL-2-specific inhibitor venetoclax has been approved by the US Food and Drug Administration for chronic lymphocytic leukemia (CLL) and acute myeloid leukemia (AML). In MCL, venetoclax has shown promising efficacy in early clinical trials; however, a significant subset of patients is resistant. By conducting a kinome-centered CRISPR-Cas9 knockout sensitizer screen, we identified casein kinase 2 (CK2) as a major regulator of venetoclax resistance in MCL. Interestingly, CK2 is over-expressed in MCL and high CK2 expression is associated with poor patient survival. Targeting of CK2, either by inducible short hairpin RNA (shRNA)-mediated knockdown of CK2 or by the CK2-inhibitor silmitasertib, did not affect cell viability by itself, but strongly synergized with venetoclax in both MCL cell lines and primary samples, also if combined with ibrutinib. Furthermore, targeting of CK2 reduced MCL-1 levels, which involved impaired MCL-1 translation by inhibition of eIF4F complex assembly, without affecting BCL-2 and BCL-XL expression. Combined, this results in enhanced BCL-2 dependence and, consequently, venetoclax sensitization. In cocultures, targeting of CK2 overcame stroma-mediated venetoclax resistance of MCL cells. Taken together, our findings indicate that targeting of CK2 sensitizes MCL cells to venetoclax through downregulation of MCL-1. These novel insights provide a strong rationale for combining venetoclax with CK2 inhibition as therapeutic strategy for MCL patients.
Collapse
Affiliation(s)
- Yvonne J Thus
- Department of Pathology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands; Cancer Center Amsterdam (CCA), Cancer Biology and Immunology - Target and Therapy Discovery, Amsterdam
| | - Martin F M De Rooij
- Department of Pathology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands; Cancer Center Amsterdam (CCA), Cancer Biology and Immunology - Target and Therapy Discovery, Amsterdam
| | - Nathalie Swier
- Department of Pathology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands; Cancer Center Amsterdam (CCA), Cancer Biology and Immunology - Target and Therapy Discovery, Amsterdam
| | - Roderick L Beijersbergen
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands; The NKI Robotics and Screening Center, Netherlands Cancer Institute, Amsterdam
| | - Jeroen E J Guikema
- Department of Pathology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands; Cancer Center Amsterdam (CCA), Cancer Biology and Immunology - Target and Therapy Discovery, Amsterdam
| | - Marie-José Kersten
- Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands; Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam
| | - Eric Eldering
- Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands; Cancer Center Amsterdam (CCA), Cancer Biology and Immunology - Target and Therapy Discovery, Amsterdam, The Netherlands; Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam
| | - Steven T Pals
- Department of Pathology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands; Cancer Center Amsterdam (CCA), Cancer Biology and Immunology - Target and Therapy Discovery, Amsterdam
| | - Arnon P Kater
- Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands; Cancer Center Amsterdam (CCA), Cancer Biology and Immunology - Target and Therapy Discovery, Amsterdam, The Netherlands; Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam
| | - Marcel Spaargaren
- Department of Pathology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands; Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam, The Netherlands; Cancer Center Amsterdam (CCA), Cancer Biology and Immunology - Target and Therapy Discovery, Amsterdam.
| |
Collapse
|
15
|
Latosińska JN, Latosińska M, Orzeszko A, Maurin JK. Synthesis and Crystal Structure of Adamantylated 4,5,6,7-Tetrahalogeno-1 H-benzimidazoles Novel Multi-Target Ligands (Potential CK2, M2 and SARS-CoV-2 Inhibitors); X-ray/DFT/QTAIM/Hirshfeld Surfaces/Molecular Docking Study. Molecules 2022; 28:147. [PMID: 36615341 PMCID: PMC9822452 DOI: 10.3390/molecules28010147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
A series of new congeners, 1-[2-(1-adamantyl)ethyl]-1H-benzimidazole (AB) and 1-[2-(1-adamantyl)ethyl]-4,5,6,7-tetrahalogeno-1H-benzimidazole (Hal=Cl, Br, I; tClAB, tBrAB, tIAB), have been synthesized and studied. These novel multi-target ligands combine a benzimidazole ring known to show antitumor activity and an adamantyl moiety showing anti-influenza activity. Their crystal structures were determined by X-ray, while intermolecular interactions were studied using topological Bader's Quantum Theory of Atoms in Molecules, Hirshfeld Surfaces, CLP and PIXEL approaches. The newly synthesized compounds crystallize within two different space groups, P-1 (AB and tIAB) and P21/c (tClAB and tBrAB). A number of intramolecular hydrogen bonds, C-H⋯Hal (Hal=Cl, Br, I), were found in all halogen-containing congeners studied, but the intermolecular C-H⋯N hydrogen bond was detected only in AB and tIAB, while C-Hal⋯π only in tClAB and tBrAB. The interplay between C-H⋯N and C-H⋯Hal hydrogen bonds and a shift from the strong (C-H⋯Cl) to the very weak (C-H⋯I) attractive interactions upon Hal exchange, supplemented with Hal⋯Hal overlapping, determines the differences in the symmetry of crystalline packing and is crucial from the biological point of view. The hypothesis about the potential dual inhibitor role of the newly synthesized congeners was verified using molecular docking and the congeners were found to be pharmaceutically attractive as Human Casein Kinase 2, CK2, inhibitors, Membrane Matrix 2 Protein, M2, blockers and Severe Acute Respiratory Syndrome Coronavirus 2, SARS-CoV-2, inhibitors. The addition of adamantyl moiety seems to broaden and modify the therapeutic indices of the 4,5,6,7-tetrahalogeno-1H-benzimidazoles.
Collapse
Affiliation(s)
| | - Magdalena Latosińska
- Faculty of Physics, Adam Mickiewicz University, Uniwersytetu Poznańskiego 2, 61-614 Poznań, Poland
| | - Andrzej Orzeszko
- Institute of Chemistry, Warsaw University of Life Sciences, 159C Nowoursynowska St., 02-787 Warsaw, Poland
| | - Jan Krzysztof Maurin
- National Medicines Institute, Chełmska 30/34, 00-750 Warsaw, Poland
- National Centre for Nuclear Research, Andrzeja Sołtana 7, 05-400 Otwock-Świerk, Poland
| |
Collapse
|
16
|
Baier A, Szyszka R. CK2 and protein kinases of the CK1 superfamily as targets for neurodegenerative disorders. Front Mol Biosci 2022; 9:916063. [PMID: 36275622 PMCID: PMC9582958 DOI: 10.3389/fmolb.2022.916063] [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: 04/08/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Casein kinases are involved in a variety of signaling pathways, and also in inflammation, cancer, and neurological diseases. Therefore, they are regarded as potential therapeutic targets for drug design. Recent studies have highlighted the importance of the casein kinase 1 superfamily as well as protein kinase CK2 in the development of several neurodegenerative pathologies, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. CK1 kinases and their closely related tau tubulin kinases as well as CK2 are found to be overexpressed in the mammalian brain. Numerous substrates have been detected which play crucial roles in neuronal and synaptic network functions and activities. The development of new substances for the treatment of these pathologies is in high demand. The impact of these kinases in the progress of neurodegenerative disorders, their bona fide substrates, and numerous natural and synthetic compounds which are able to inhibit CK1, TTBK, and CK2 are discussed in this review.
Collapse
Affiliation(s)
- Andrea Baier
- Institute of Biological Sciences, The John Paul II Catholic University of Lublin, Lublin, Poland
| | - Ryszard Szyszka
- Institute of Biological Sciences, The John Paul II Catholic University of Lublin, Lublin, Poland
| |
Collapse
|
17
|
Trembley JH, Kren BT, Afzal M, Scaria GA, Klein MA, Ahmed K. Protein kinase CK2 – diverse roles in cancer cell biology and therapeutic promise. Mol Cell Biochem 2022; 478:899-926. [PMID: 36114992 PMCID: PMC9483426 DOI: 10.1007/s11010-022-04558-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022]
Abstract
The association of protein kinase CK2 (formerly casein kinase II or 2) with cell growth and proliferation in cells was apparent at early stages of its investigation. A cancer-specific role for CK2 remained unclear until it was determined that CK2 was also a potent suppressor of cell death (apoptosis); the latter characteristic differentiated its function in normal versus malignant cells because dysregulation of both cell growth and cell death is a universal feature of cancer cells. Over time, it became evident that CK2 exerts its influence on a diverse range of cell functions in normal as well as in transformed cells. As such, CK2 and its substrates are localized in various compartments of the cell. The dysregulation of CK2 is documented in a wide range of malignancies; notably, by increased CK2 protein and activity levels with relatively moderate change in its RNA abundance. High levels of CK2 are associated with poor prognosis in multiple cancer types, and CK2 is a target for active research and testing for cancer therapy. Aspects of CK2 cellular roles and targeting in cancer are discussed in the present review, with focus on nuclear and mitochondrial functions and prostate, breast and head and neck malignancies.
Collapse
Affiliation(s)
- Janeen H Trembley
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA.
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Betsy T Kren
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA
| | - Muhammad Afzal
- Department of Biochemistry, Riphah International University, Islamabad, Pakistan
| | - George A Scaria
- Hematology/Oncology Section, Primary Care Service Line, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA
| | - Mark A Klein
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
- Hematology/Oncology Section, Primary Care Service Line, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Khalil Ahmed
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA.
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
- Department of Urology, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
18
|
CK2 Inhibition and Antitumor Activity of 4,7-Dihydro-6-nitroazolo[1,5-a]pyrimidines. Molecules 2022; 27:molecules27165239. [PMID: 36014483 PMCID: PMC9415015 DOI: 10.3390/molecules27165239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/04/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
Today, cancer is one of the most widespread and dangerous human diseases with a high mortality rate. Nevertheless, the search and application of new low-toxic and effective drugs, combined with the timely diagnosis of diseases, makes it possible to cure most types of tumors at an early stage. In this work, the range of new polysubstituted 4,7-dihydro-6-nitroazolo[1,5-a]pyrimidines was extended. The structure of all the obtained compounds was confirmed by the data of 1H, 13C NMR spectroscopy, IR spectroscopy, and elemental analysis. These compounds were evaluated against human recombinant CK2 using the ADP-GloTM assay. In addition, the IC50 parameters were calculated based on the results of the MTT test against glioblastoma (A-172), embryonic rhabdomyosarcoma (Rd), osteosarcoma (Hos), and human embryonic kidney (Hek-293) cells. Compounds 5f, 5h, and 5k showed a CK2 inhibitory activity close to the reference molecule (staurosporine). The most potential compound in the MTT test was 5m with an IC50 from 13 to 27 µM. Thus, our results demonstrate that 4,7-dihydro-6-nitroazolo[1,5-a]pyrimidines are promising for further investigation of their antitumor properties.
Collapse
|
19
|
White A, McGlone A, Gomez-Pastor R. Protein Kinase CK2 and Its Potential Role as a Therapeutic Target in Huntington's Disease. Biomedicines 2022; 10:1979. [PMID: 36009526 PMCID: PMC9406209 DOI: 10.3390/biomedicines10081979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Huntington's Disease (HD) is a devastating neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the HTT gene, for which no disease modifying therapies are currently available. Much of the recent research has focused on developing therapies to directly lower HTT expression, and while promising, these therapies have presented several challenges regarding administration and efficacy. Another promising therapeutic approach is the modulation of HTT post-translational modifications (PTMs) that are dysregulated in disease and have shown to play a key role in HTT toxicity. Among all PTMs, modulation of HTT phosphorylation has been proposed as an attractive therapeutic option due to the possibility of orally administering specific kinase effectors. One of the kinases described to participate in HTT phosphorylation is Protein Kinase CK2. CK2 has recently emerged as a target for the treatment of several neurological and psychiatric disorders, although its role in HD remains controversial. While pharmacological studies in vitro inhibiting CK2 resulted in reduced HTT phosphorylation and increased toxicity, genetic approaches in mouse models of HD have provided beneficial effects. In this review we discuss potential therapeutic approaches related to the manipulation of HTT-PTMs with special emphasis on the role of CK2 as a therapeutic target in HD.
Collapse
Affiliation(s)
| | | | - Rocio Gomez-Pastor
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
20
|
Nipun VB, Amin KA. Recent Advances in Protein Kinase CK2, a Potential Therapeutic Target in Cancer. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022; 48:919-931. [DOI: 10.1134/s1068162022050144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- V. B. Nipun
- Cancer Research Center, Shantou University Medical Collage, Shantou, Guangdong, 515041, PR China
- Department of Chemistry, Faculty of Science, University of Imam Abdulrahman Bin Faisal university, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - K. A. Amin
- Department of Chemistry, Faculty of Science, University of Imam Abdulrahman Bin Faisal university, P.O. Box 1982, Dammam, 31441, Saudi Arabia
- Basic and Applied Scientific Research Center, Imam Abdulrahman Bin Faisal university, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| |
Collapse
|
21
|
Huang Y, Winklbauer R. Cell cortex regulation by the planar cell polarity protein Prickle1. J Cell Biol 2022; 221:e202008116. [PMID: 35512799 PMCID: PMC9082893 DOI: 10.1083/jcb.202008116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/18/2022] [Accepted: 04/09/2022] [Indexed: 01/07/2023] Open
Abstract
The planar cell polarity pathway regulates cell polarity, adhesion, and rearrangement. Its cytoplasmic core components Prickle (Pk) and Dishevelled (Dvl) often localize as dense puncta at cell membranes to form antagonizing complexes and establish cell asymmetry. In vertebrates, Pk and Dvl have been implicated in actomyosin cortex regulation, but the mechanism of how these proteins control cell mechanics is unclear. Here we demonstrate that in Xenopus prechordal mesoderm cells, diffusely distributed, cytoplasmic Pk1 up-regulates the F-actin content of the cortex. This counteracts cortex down-regulation by Dvl2. Both factors act upstream of casein kinase II to increase or decrease cortical tension. Thus, cortex modulation by Pk1 and Dvl2 is translated into mechanical force and affects cell migration and rearrangement during radial intercalation in the prechordal mesoderm. Pk1 also forms puncta and plaques, which are associated with localized depletion of cortical F-actin, suggesting opposite roles for diffuse and punctate Pk1.
Collapse
Affiliation(s)
- Yunyun Huang
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Rudolf Winklbauer
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Shahroz MM, Sharma HK, Altamimi ASA, Alamri MA, Ali A, Ali A, Alqahtani S, Altharawi A, Alabbas AB, Alossaimi MA, Riadi Y, Firoz A, Afzal O. Novel and Potential Small Molecule Scaffolds as DYRK1A Inhibitors by Integrated Molecular Docking-Based Virtual Screening and Dynamics Simulation Study. Molecules 2022; 27:1159. [PMID: 35208955 PMCID: PMC8875901 DOI: 10.3390/molecules27041159] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 12/16/2022] Open
Abstract
The dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) is a novel, promising and emerging biological target for therapeutic intervention in neurodegenerative diseases, especially in Alzheimer's disease (AD). The molMall database, comprising rare, diverse and unique compounds, was explored for molecular docking-based virtual screening against the DYRK1A protein, in order to find out potential inhibitors. Ligands exhibiting hydrogen bond interactions with key amino acid residues such as Ile165, Lys188 (catalytic), Glu239 (gk+1), Leu241 (gk+3), Ser242, Asn244, and Asp307, of the target protein, were considered potential ligands. Hydrogen bond interactions with Leu241 (gk+3) were considered key determinants for the selection. High scoring structures were also docked by Glide XP docking in the active sites of twelve DYRK1A related protein kinases, viz. DYRK1B, DYRK2, CDK5/p25, CK1, CLK1, CLK3, GSK3β, MAPK2, MAPK10, PIM1, PKA, and PKCα, in order to find selective DYRK1A inhibitors. MM/GBSA binding free energies of selected ligand-protein complexes were also calculated in order to remove false positive hits. Physicochemical and pharmacokinetic properties of the selected six hit ligands were also computed and related with the proposed limits for orally active CNS drugs. The computational toxicity webserver ProTox-II was used to predict the toxicity profile of selected six hits (molmall IDs 9539, 11352, 15938, 19037, 21830 and 21878). The selected six docked ligand-protein systems were exposed to 100 ns molecular dynamics (MD) simulations to validate their mechanism of interactions and stability in the ATP pocket of human DYRK1A kinase. All six ligands were found to be stable in the ATP binding pocket of DYRK1A kinase.
Collapse
Affiliation(s)
- Mir Mohammad Shahroz
- Department of Pharmaceutical Chemistry, College of Pharmacy, Sri Satya Sai University of Technology and Medical Sciences, Sehore 466001, Madhya Pradesh, India;
| | - Hemant Kumar Sharma
- Department of Pharmaceutical Chemistry, College of Pharmacy, Sri Satya Sai University of Technology and Medical Sciences, Sehore 466001, Madhya Pradesh, India;
| | - Abdulmalik S. A. Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al Kharj 11942, Saudi Arabia; (A.S.A.A.); (M.A.A.); (S.A.); (A.A.); (A.B.A.); (M.A.A.); (Y.R.)
| | - Mubarak A. Alamri
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al Kharj 11942, Saudi Arabia; (A.S.A.A.); (M.A.A.); (S.A.); (A.A.); (A.B.A.); (M.A.A.); (Y.R.)
| | - Abuzer Ali
- Department of Pharmacognosy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Amena Ali
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Safar Alqahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al Kharj 11942, Saudi Arabia; (A.S.A.A.); (M.A.A.); (S.A.); (A.A.); (A.B.A.); (M.A.A.); (Y.R.)
| | - Ali Altharawi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al Kharj 11942, Saudi Arabia; (A.S.A.A.); (M.A.A.); (S.A.); (A.A.); (A.B.A.); (M.A.A.); (Y.R.)
| | - Alhumaidi B. Alabbas
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al Kharj 11942, Saudi Arabia; (A.S.A.A.); (M.A.A.); (S.A.); (A.A.); (A.B.A.); (M.A.A.); (Y.R.)
| | - Manal A. Alossaimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al Kharj 11942, Saudi Arabia; (A.S.A.A.); (M.A.A.); (S.A.); (A.A.); (A.B.A.); (M.A.A.); (Y.R.)
| | - Yassine Riadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al Kharj 11942, Saudi Arabia; (A.S.A.A.); (M.A.A.); (S.A.); (A.A.); (A.B.A.); (M.A.A.); (Y.R.)
| | - Ahmad Firoz
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80200, Jeddah 21589, Saudi Arabia;
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al Kharj 11942, Saudi Arabia; (A.S.A.A.); (M.A.A.); (S.A.); (A.A.); (A.B.A.); (M.A.A.); (Y.R.)
| |
Collapse
|
23
|
Pucko EB, Ostrowski RP. Inhibiting CK2 among Promising Therapeutic Strategies for Gliomas and Several Other Neoplasms. Pharmaceutics 2022; 14:331. [PMID: 35214064 PMCID: PMC8877581 DOI: 10.3390/pharmaceutics14020331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
In gliomas, casein kinase 2 (CK2) plays a dominant role in cell survival and tumour invasiveness and is upregulated in many brain tumours. Among CK2 inhibitors, benzimidazole and isothiourea derivatives hold a dominant position. While targeting glioma tumour cells, they show limited toxicity towards normal cells. Research in recent years has shown that these compounds can be suitable as components of combined therapies with hyperbaric oxygenation. Such a combination increases the susceptibility of glioma tumour cells to cell death via apoptosis. Moreover, researchers planning on using any other antiglioma investigational pharmaceutics may want to consider using these agents in combination with CK2 inhibitors. However, different compounds are not equally effective when in such combination. More research is needed to elucidate the mechanism of treatment and optimize the treatment regimen. In addition, the role of CK2 in gliomagenesis and maintenance seems to have been challenged recently, as some compounds structurally similar to CK2 inhibitors do not inhibit CK2 while still being effective at reducing glioma viability and invasion. Furthermore, some newly developed inhibitors specific for CK2 do not appear to have strong anticancer properties. Further experimental and clinical studies of these inhibitors and combined therapies are warranted.
Collapse
Affiliation(s)
| | - Robert P. Ostrowski
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| |
Collapse
|
24
|
Aboushady Y, Gabr M, ElHady AK, Salah M, Abadi AH, Wilms G, Becker W, Abdel-Halim M, Engel M. Discovery of Hydroxybenzothiazole Urea Compounds as Multitargeted Agents Suppressing Major Cytotoxic Mechanisms in Neurodegenerative Diseases. ACS Chem Neurosci 2021; 12:4302-4318. [PMID: 34726394 DOI: 10.1021/acschemneuro.1c00475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Multiple factors are causally responsible and/or contribute to the progression of Alzheimer's and Parkinson's diseases. The protein kinase Dyrk1A was identified as a promising target as it phosphorylates tau protein, α-synuclein, and parkin. The first goal of our study was to optimize our previously identified Dyrk1A inhibitors of the 6-hydroxy benzothiazole urea chemotype in terms of potency and selectivity. Our efforts led to the development of the 3-fluorobenzyl amide derivative 16b, which displayed the highest potency against Dyrk1A (IC50 = 9.4 nM). In general, the diversification of the benzylamide moiety led to an enhanced selectivity over the most homologous isoform, Dyrk1B, which was a meaningful indicator, as the high selectivity could be confirmed in an extended selectivity profiling of 3b and 16b. Eventually, we identified the novel phenethyl amide derivative 24b as a triple inhibitor of Dyrk1A kinase activity (IC50 = 119 nM) and the aggregation of tau and α-syn oligomers. We provide evidence that the novel combination of selective Dyrk1A inhibition and suppression of tau and α-syn aggregations of our new lead compound confers efficacy in several established cellular models of neurotoxic mechanisms relevant to neurodegenerative diseases, including α-syn- and 6-hydroxydopamine-induced cytotoxicities.
Collapse
Affiliation(s)
- Youssef Aboushady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Moustafa Gabr
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Ahmed K. ElHady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
- School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, New Administrative Capital, Cairo 11311, Egypt
| | - Mohamed Salah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, October University for Modern Sciences and Arts, Cairo 12451, Egypt
| | - Ashraf H. Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Gerrit Wilms
- Institute of Pharmacology and Toxicology, Medical Faculty of the RWTH Aachen University, Wendlingweg 2, Aachen 52074, Germany
| | - Walter Becker
- Institute of Pharmacology and Toxicology, Medical Faculty of the RWTH Aachen University, Wendlingweg 2, Aachen 52074, Germany
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3 Saarbrücken D-66123, Germany
| |
Collapse
|
25
|
Tran NNQ, Chun KH. ROCK2-Specific Inhibitor KD025 Suppresses Adipocyte Differentiation by Inhibiting Casein Kinase 2. Molecules 2021; 26:4747. [PMID: 34443331 PMCID: PMC8401933 DOI: 10.3390/molecules26164747] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 11/16/2022] Open
Abstract
KD025, a ROCK2 isoform-specific inhibitor, has an anti-adipogenic activity which is not mediated by ROCK2 inhibition. To identify the target, we searched binding targets of KD025 by using the KINOMEscanTM screening platform, and we identified casein kinase 2 (CK2) as a novel target. KD025 showed comparable binding affinity to CK2α (Kd = 128 nM). By contrast, CK2 inhibitor CX-4945 and ROCK inhibitor fasudil did not show such cross-reactivity. In addition, KD025 effectively inhibited CK2 at a nanomolar concentration (IC50 = 50 nM). We examined if the inhibitory effect of KD025 on adipocyte differentiation is through the inhibition of CK2. Both CX-4945 and KD025 suppressed the generation of lipid droplets and the expression of proadipogenic genes Pparg and Cebpa in 3T3-L1 cells during adipocyte differentiation. Fasudil exerted no significant effect on the quantity of lipid droplets, but another ROCK inhibitor Y-27632 increased the expression of Pparg and Cebpa. Both CX-4945 and KD025 acted specifically in the middle stage (days 1-3) but were ineffective when treated at days 0-1 or the late stages, indicating that CX-4945 and KD025 may regulate the same target, CK2. The mRNA and protein levels of CK2α and CK2β generally decreased in 3T3-L1 cells at day 2 but recovered thereafter. Other well-known CK2 inhibitors DMAT and quinalizarin inhibited effectively the differentiation of 3T3-L1 cells. Taken together, the results of this study confirmed that KD025 inhibits ROCK2 and CK2, and that the inhibitory effect on adipocyte differentiation is through the inhibition of CK2.
Collapse
Affiliation(s)
| | - Kwang-Hoon Chun
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon 21936, Korea;
| |
Collapse
|
26
|
Lindenblatt D, Applegate V, Nickelsen A, Klußmann M, Neundorf I, Götz C, Jose J, Niefind K. Molecular Plasticity of Crystalline CK2α' Leads to KN2, a Bivalent Inhibitor of Protein Kinase CK2 with Extraordinary Selectivity. J Med Chem 2021; 65:1302-1312. [PMID: 34323071 DOI: 10.1021/acs.jmedchem.1c00063] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
CK2α and CK2α' are paralogous catalytic subunits of CK2, which belongs to the eukaryotic protein kinases. CK2 promotes tumorigenesis and the spread of pathogenic viruses like SARS-CoV-2 and is thus an attractive drug target. Efforts to develop selective CK2 inhibitors binding offside the ATP site had disclosed the αD pocket in CK2α; its occupation requires large conformational adaptations of the helix αD. As shown here, the αD pocket is accessible also in CK2α', where the necessary structural plasticity can be triggered with suitable ligands even in the crystalline state. A CK2α' structure with an ATP site and an αD pocket ligand guided the design of the bivalent CK2 inhibitor KN2. It binds to CK2 with low nanomolar affinity, is cell-permeable, and suppresses the intracellular phosphorylation of typical CK2 substrates. Kinase profiling revealed a high selectivity of KN2 for CK2 and emphasizes the selectivity-promoting potential of the αD pocket.
Collapse
Affiliation(s)
- Dirk Lindenblatt
- Department für Chemie, Institut für Biochemie, Universität zu Köln, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Violetta Applegate
- Department für Chemie, Institut für Biochemie, Universität zu Köln, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Anna Nickelsen
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, PharmaCampus, Corrensstr. 48, D-48149 Münster, Germany
| | - Merlin Klußmann
- Department für Chemie, Institut für Biochemie, Universität zu Köln, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Ines Neundorf
- Department für Chemie, Institut für Biochemie, Universität zu Köln, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Claudia Götz
- Medizinische Biochemie und Molekularbiologie, Universität des Saarlandes, Kirrberger Str., Geb. 44, D-66421 Homburg/Saar, Germany
| | - Joachim Jose
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, PharmaCampus, Corrensstr. 48, D-48149 Münster, Germany
| | - Karsten Niefind
- Department für Chemie, Institut für Biochemie, Universität zu Köln, Zülpicher Str. 47, D-50674 Köln, Germany
| |
Collapse
|
27
|
Regulation of stability and inhibitory activity of the tumor suppressor SEF through casein-kinase II-mediated phosphorylation. Cell Signal 2021; 86:110085. [PMID: 34280495 DOI: 10.1016/j.cellsig.2021.110085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 11/23/2022]
Abstract
Inflammation and cancer are intimately linked. A key mediator of inflammation is the transcription-factor NF-κB/RelA:p50. SEF (also known as IL-17RD) is a feedback antagonist of NF-κB/RelA:p50 that is emerging as an important link between inflammation and cancer. SEF acts as a buffer to prevent excessive NF-κB activity by sequestering NF-κB/RelA:p50 in the cytoplasm of unstimulated cells, and consequently attenuating the NF-κB response upon pro-inflammatory cytokine stimulation. SEF contributes to cancer progression also via modulating other signaling pathways, including those triggered by growth-factors. Despite its important role in human physiology and pathology, mechanisms that regulate SEF biochemical properties and inhibitory activity are unknown. Here we show that human SEF is an intrinsically labile protein that is stabilized via CK2-mediated phosphorylation, and identified the residues whom phosphorylation by CK2 stabilizes hSEF. Unlike endogenous SEF, ectopic SEF was rapidly degraded when overexpressed but was stabilized in the presence of excess CK2, suggesting a mechanism for limiting SEF levels depending upon CK2 processivity. Additionally, phosphorylation by CK2 potentiated hSef interaction with NF-κB in cell-free binding assays. Most importantly, we identified a CK2 phosphorylation site that was indispensable for SEF inhibition of pro-inflammatory cytokine signaling but was not required for SEF inhibition of growth-factor signaling. To our knowledge, this is the first demonstration of post-translational modifications that regulate SEF at multiple levels to optimize its inhibitory activity in a specific signaling context. These findings may facilitate the design of SEF variants for treating cytokine-dependent pathologies, including cancer and chronic inflammation.
Collapse
|
28
|
Zwicker F, Hauswald H, Weber KJ, Debus JÜ, Huber PE. In Vivo Evaluation of Combined CK2 Inhibition and Irradiation in Human WiDr Tumours. In Vivo 2021; 35:111-117. [PMID: 33402456 DOI: 10.21873/invivo.12238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/17/2020] [Accepted: 10/21/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Casein kinase 2 (CK2) which sustains multiple pro-survival functions in cellular DNA-damage response, is strictly regulated in normal cells but elevated in cancer. CK2 is considered as a potential therapeutic target, and its inhibition has been associated with radiosensitization in mammalian cells in vitro. Here, we investigated potential radiosensitization by CK2 inhibition in vivo. MATERIALS AND METHODS The effect of CK2 inhibition in vivo was investigated in human WiDr-xenograft tumours grown subcutaneously on BALB/c nu/nu mice with and without fractionated irradiation. CK2 inhibition was performed using the specific inhibitor tetra-bromobenzotriazole (TBB). Histological examinations included staining for apoptosis and double-strand breaks. RESULTS Both TBB treatment alone and radiation alone significantly reduced tumour growth, which was reflected by increased apoptosis rates. However, TBB treatment did not boost radiation-induced tumour growth suppression in combined treatment, although the apoptosis rate increased and repair of double-strand breaks was reduced. This was in stark contrast to previous data on in vitro radiosensitization. CONCLUSION The absence of radiosensitization by CK2 inhibition should be investigated in different tumour models.
Collapse
Affiliation(s)
- Felix Zwicker
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; .,Clinical Cooperation Unit Molecular Radiation Oncology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Henrik Hauswald
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Klaus-Josef Weber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - JÜrgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Molecular Radiation Oncology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Molecular Radiation Oncology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| |
Collapse
|
29
|
Protein kinase CK2: a potential therapeutic target for diverse human diseases. Signal Transduct Target Ther 2021; 6:183. [PMID: 33994545 PMCID: PMC8126563 DOI: 10.1038/s41392-021-00567-7] [Citation(s) in RCA: 200] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/04/2023] Open
Abstract
CK2 is a constitutively active Ser/Thr protein kinase, which phosphorylates hundreds of substrates, controls several signaling pathways, and is implicated in a plethora of human diseases. Its best documented role is in cancer, where it regulates practically all malignant hallmarks. Other well-known functions of CK2 are in human infections; in particular, several viruses exploit host cell CK2 for their life cycle. Very recently, also SARS-CoV-2, the virus responsible for the COVID-19 pandemic, has been found to enhance CK2 activity and to induce the phosphorylation of several CK2 substrates (either viral and host proteins). CK2 is also considered an emerging target for neurological diseases, inflammation and autoimmune disorders, diverse ophthalmic pathologies, diabetes, and obesity. In addition, CK2 activity has been associated with cardiovascular diseases, as cardiac ischemia-reperfusion injury, atherosclerosis, and cardiac hypertrophy. The hypothesis of considering CK2 inhibition for cystic fibrosis therapies has been also entertained for many years. Moreover, psychiatric disorders and syndromes due to CK2 mutations have been recently identified. On these bases, CK2 is emerging as an increasingly attractive target in various fields of human medicine, with the advantage that several very specific and effective inhibitors are already available. Here, we review the literature on CK2 implication in different human pathologies and evaluate its potential as a pharmacological target in the light of the most recent findings.
Collapse
|
30
|
Baier A, Kokel A, Horton W, Gizińska E, Pandey G, Szyszka R, Török B, Török M. Organofluorine Hydrazone Derivatives as Multifunctional Anti-Alzheimer's Agents with CK2 Inhibitory and Antioxidant Features. ChemMedChem 2021; 16:1927-1932. [PMID: 33713036 DOI: 10.1002/cmdc.202100047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Indexed: 11/10/2022]
Abstract
A set of novel hydrazone derivatives were synthesized and analyzed for their biological activities. The compounds were tested for their inhibitory effect on the phosphorylating activity of the protein kinase CK2, and their antioxidant activity was also determined in three commonly used assays. The hydrazones were evaluated for their radical scavenging against the DPPH, ABTS and peroxyl radicals. Several compounds have been identified as good antioxidants as well as potent protein kinase CK2 inhibitors. Most hydrazones containing a 4-N(CH3 )2 residue or perfluorinated phenyl rings showed high activity in the radical-scavenging assays and possess nanomolar IC50 values in the kinase assays.
Collapse
Affiliation(s)
- Andrea Baier
- Department of Animal Physiology and Toxicology, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Anne Kokel
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA, 02125, USA
| | - William Horton
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA, 02125, USA
| | - Ewa Gizińska
- Department of Molecular Biology, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Garima Pandey
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA, 02125, USA
| | - Ryszard Szyszka
- Department of Molecular Biology, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Béla Török
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA, 02125, USA
| | - Marianna Török
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA, 02125, USA
| |
Collapse
|
31
|
Wang Y, Wang X, Xu G, Gou S. Novel CK2-Specific Pt(II) Compound Reverses Cisplatin-Induced Resistance by Inhibiting Cancer Cell Stemness and Suppressing DNA Damage Repair in Non-small Cell Lung Cancer Treatments. J Med Chem 2021; 64:4163-4178. [PMID: 33784109 DOI: 10.1021/acs.jmedchem.1c00079] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cancer stem cells (CSCs) have a pivotal impact in drug resistance, tumor metastasis, and progression of various cancer entities, including in non-small cell lung cancer (NSCLC). A CK2 inhibitor HY1 was found to show potent CSC inhibitory effects in A549 cells. By taking advantage of inherent CK2 specificity and CSC inhibition of HY1, a Pt(II) agent (HY1-Pt) was developed by conjugation of HY1 with an active Pt(II) unit to reverse cisplatin-induced resistance in A549/cDDP cell treatment. In vitro biological studies indicated that HY1-Pt can target CK2, suppress DNA damage repair, reinforce cellular accumulation of platinum, and reverse resistance apart from effectively inhibiting CSCs through Wnt/β-catenin signal pathway in A549/cDDP cells. Significantly, HY1-Pt presented an acceptable pharmacokinetic behavior and exhibited higher tumor growth inhibitory efficacy than cisplatin either in A549 or A549/cDDP xenograft models with low toxicity. Overall, HY1-Pt is a promising drug candidate for NSCLC treatment.
Collapse
Affiliation(s)
- Yuanjiang Wang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Xinyi Wang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Gang Xu
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Shaohua Gou
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| |
Collapse
|
32
|
Atkinson EL, Iegre J, Brear PD, Zhabina EA, Hyvönen M, Spring DR. Downfalls of Chemical Probes Acting at the Kinase ATP-Site: CK2 as a Case Study. Molecules 2021; 26:1977. [PMID: 33807474 PMCID: PMC8037657 DOI: 10.3390/molecules26071977] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Protein kinases are a large class of enzymes with numerous biological roles and many have been implicated in a vast array of diseases, including cancer and the novel coronavirus infection COVID-19. Thus, the development of chemical probes to selectively target each kinase is of great interest. Inhibition of protein kinases with ATP-competitive inhibitors has historically been the most widely used method. However, due to the highly conserved structures of ATP-sites, the identification of truly selective chemical probes is challenging. In this review, we use the Ser/Thr kinase CK2 as an example to highlight the historical challenges in effective and selective chemical probe development, alongside recent advances in the field and alternative strategies aiming to overcome these problems. The methods utilised for CK2 can be applied to an array of protein kinases to aid in the discovery of chemical probes to further understand each kinase's biology, with wide-reaching implications for drug development.
Collapse
Affiliation(s)
- Eleanor L. Atkinson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; (E.L.A.); (J.I.)
| | - Jessica Iegre
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; (E.L.A.); (J.I.)
| | - Paul D. Brear
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK; (P.D.B.); (E.A.Z.); (M.H.)
| | - Elizabeth A. Zhabina
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK; (P.D.B.); (E.A.Z.); (M.H.)
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK; (P.D.B.); (E.A.Z.); (M.H.)
| | - David R. Spring
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; (E.L.A.); (J.I.)
| |
Collapse
|
33
|
Bansal Y, Minhas R, Singhal A, Arora RK, Bansal G. Benzimidazole: A Multifacted Nucelus for Anticancer Agents. CURR ORG CHEM 2021. [DOI: 10.2174/1385272825666210208141107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cancer is characterized by an uncontrolled proliferation of cells, dedifferentiation,
invasiveness and metastasis. Endothelial growth factor (eGF), insulin-like growth factor
(IGF), platelet-derived growth factor (PDGF), Fibroblast growth factor (FGF), Vascular endothelial
growth factor (VEGF), checkpoint kinase 1 & 2 ( Chk1 & Chk2), aurora kinases,
topoisomerases, histone deacetylators (HDAC), poly(ADP-Ribose)polymerase (PARP), farnesyl
transferases, RAS-MAPK pathway and PI3K-Akt-mTOR pathway, are some of the
prominent mediators implicated in the proliferation of tumor cells. Huge artillery of natural
and synthetic compounds as anticancer, which act by inhibiting one or more of the enzymes
and/or pathways responsible for the progression of tumor cells, is reported in the literature.
The major limitations of anticancer agents used in clinics as well as of those under development
in literature are normal cell toxicity and other side effects due to lack of specificity.
Hence, medicinal chemists across the globe have been working for decades to develop potent and safe anticancer
agents from natural sources as well as from different classes of heterocycles. Benzimidazole is one of the most important
and explored heteronucelus because of their versatility in biological actions as well as synthetic applications
in medicinal chemistry. The structural similarity of amino derivatives of benzimidazole with purines makes it a fascinating
nucleus for the development of anticancer, antimicrobial and anti-HIV agents. This review article is an attempt
to critically analyze various reports on benzimidazole derivatives acting on different targets to act as anticancer so as
to understand the structural requirements around benzimidazole nucleus for each target and enable medicinal chemists
to promote rational development of antitumor agents.
Collapse
Affiliation(s)
- Yogita Bansal
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala - 147002, India
| | - Richa Minhas
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala - 147002, India
| | - Ankit Singhal
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala - 147002, India
| | - Radhey Krishan Arora
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala - 147002, India
| | - Gulshan Bansal
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala - 147002, India
| |
Collapse
|
34
|
Alves ALV, Gomes INF, Carloni AC, Rosa MN, da Silva LS, Evangelista AF, Reis RM, Silva VAO. Role of glioblastoma stem cells in cancer therapeutic resistance: a perspective on antineoplastic agents from natural sources and chemical derivatives. Stem Cell Res Ther 2021; 12:206. [PMID: 33762015 PMCID: PMC7992331 DOI: 10.1186/s13287-021-02231-x] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 02/15/2021] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma (GBM) is the highest-grade form of glioma, as well as one of the most aggressive types of cancer, exhibiting rapid cellular growth and highly invasive behavior. Despite significant advances in diagnosis and therapy in recent decades, the outcomes for high-grade gliomas (WHO grades III-IV) remain unfavorable, with a median overall survival time of 15–18 months. The concept of cancer stem cells (CSCs) has emerged and provided new insight into GBM resistance and management. CSCs can self-renew and initiate tumor growth and are also responsible for tumor cell heterogeneity and the induction of systemic immunosuppression. The idea that GBM resistance could be dependent on innate differences in the sensitivity of clonogenic glial stem cells (GSCs) to chemotherapeutic drugs/radiation prompted the scientific community to rethink the understanding of GBM growth and therapies directed at eliminating these cells or modulating their stemness. This review aims to describe major intrinsic and extrinsic mechanisms that mediate chemoradioresistant GSCs and therapies based on antineoplastic agents from natural sources, derivatives, and synthetics used alone or in synergistic combination with conventional treatment. We will also address ongoing clinical trials focused on these promising targets. Although the development of effective therapy for GBM remains a major challenge in molecular oncology, GSC knowledge can offer new directions for a promising future.
Collapse
Affiliation(s)
- Ana Laura V Alves
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, São Paulo, Brazil
| | - Izabela N F Gomes
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, São Paulo, Brazil
| | - Adriana C Carloni
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, São Paulo, Brazil
| | - Marcela N Rosa
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, São Paulo, Brazil
| | - Luciane S da Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, São Paulo, Brazil
| | - Adriane F Evangelista
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, São Paulo, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, São Paulo, Brazil.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, 4806-909, Braga, Portugal
| | - Viviane Aline O Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, São Paulo, Brazil.
| |
Collapse
|
35
|
Wells CI, Drewry DH, Pickett JE, Tjaden A, Krämer A, Müller S, Gyenis L, Menyhart D, Litchfield DW, Knapp S, Axtman AD. Development of a potent and selective chemical probe for the pleiotropic kinase CK2. Cell Chem Biol 2021; 28:546-558.e10. [PMID: 33484635 DOI: 10.1016/j.chembiol.2020.12.013] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/30/2020] [Accepted: 12/22/2020] [Indexed: 12/18/2022]
Abstract
Building on the pyrazolopyrimidine CK2 (casein kinase 2) inhibitor scaffold, we designed a small targeted library. Through comprehensive evaluation of inhibitor selectivity, we identified inhibitor 24 (SGC-CK2-1) as a highly potent and cell-active CK2 chemical probe with exclusive selectivity for both human CK2 isoforms. Remarkably, despite years of research pointing to CK2 as a key driver in cancer, our chemical probe did not elicit a broad antiproliferative phenotype in >90% of >140 cell lines when tested in dose-response. While many publications have reported CK2 functions, CK2 biology is complex and an available high-quality chemical tool such as SGC-CK2-1 will be indispensable in deciphering the relationships between CK2 function and phenotypes.
Collapse
Affiliation(s)
- Carrow I Wells
- Structural Genomics Consortium (SGC), UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, NC 27599, USA; Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, UNC-CH, Chapel Hill, NC 27599, USA
| | - David H Drewry
- Structural Genomics Consortium (SGC), UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, NC 27599, USA; Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, UNC-CH, Chapel Hill, NC 27599, USA
| | - Julie E Pickett
- Structural Genomics Consortium (SGC), UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, NC 27599, USA; Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, UNC-CH, Chapel Hill, NC 27599, USA
| | - Amelie Tjaden
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany; Structural Genomics Consortium, Buchman Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Andreas Krämer
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany; Structural Genomics Consortium, Buchman Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Susanne Müller
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany; Structural Genomics Consortium, Buchman Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Laszlo Gyenis
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Daniel Menyhart
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - David W Litchfield
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada; Department of Oncology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Stefan Knapp
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany; Structural Genomics Consortium, Buchman Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Alison D Axtman
- Structural Genomics Consortium (SGC), UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, NC 27599, USA; Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, UNC-CH, Chapel Hill, NC 27599, USA.
| |
Collapse
|
36
|
Protein kinase CK2 inhibition as a pharmacological strategy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 124:23-46. [PMID: 33632467 DOI: 10.1016/bs.apcsb.2020.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
CK2 is a constitutively active Ser/Thr protein kinase which phosphorylates hundreds of substrates. Since they are primarily related to survival and proliferation pathways, the best-known pathological roles of CK2 are in cancer, where its targeting is currently being considered as a possible therapy. However, CK2 activity has been found instrumental in many other human pathologies, and its inhibition will expectably be extended to different purposes in the near future. Here, after a description of CK2 features and implications in diseases, we analyze the different inhibitors and strategies available to target CK2, and update the results so far obtained by their in vivo application.
Collapse
|
37
|
Tandon V, de la Vega L, Banerjee S. Emerging roles of DYRK2 in cancer. J Biol Chem 2021; 296:100233. [PMID: 33376136 PMCID: PMC7948649 DOI: 10.1074/jbc.rev120.015217] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022] Open
Abstract
Over the last decade, the CMGC kinase DYRK2 has been reported as a tumor suppressor across various cancers triggering major antitumor and proapoptotic signals in breast, colon, liver, ovary, brain, and lung cancers, with lower DYRK2 expression correlated with poorer prognosis in patients. Contrary to this, various medicinal chemistry studies reported robust antiproliferative properties of DYRK2 inhibitors, whereas unbiased 'omics' and genome-wide association study-based studies identified DYRK2 as a highly overexpressed kinase in various patient tumor samples. A major paradigm shift occurred in the last 4 years when DYRK2 was found to regulate proteostasis in cancer via a two-pronged mechanism. DYRK2 phosphorylated and activated the 26S proteasome to enhance degradation of misfolded/tumor-suppressor proteins while also promoting the nuclear stability and transcriptional activity of its substrate, heat-shock factor 1 triggering protein folding. Together, DYRK2 regulates proteostasis and promotes protumorigenic survival for specific cancers. Indeed, potent and selective small-molecule inhibitors of DYRK2 exhibit in vitro and in vivo anti-tumor activity in triple-negative breast cancer and myeloma models. However, with conflicting and contradictory reports across different cancers, the overarching role of DYRK2 remains enigmatic. Specific cancer (sub)types coupled to spatiotemporal interactions with substrates could decide the procancer or anticancer role of DYRK2. The current review aims to provide a balanced and critical appreciation of the literature to date, highlighting top substrates such as p53, c-Myc, c-Jun, heat-shock factor 1, proteasome, or NOTCH1, to discuss DYRK2 inhibitors available to the scientific community and to shed light on this duality of protumorigenic and antitumorigenic roles of DYRK2.
Collapse
Affiliation(s)
- Vasudha Tandon
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Laureano de la Vega
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Sourav Banerjee
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom.
| |
Collapse
|
38
|
Larson SR, Bortell N, Illies A, Crisler WJ, Matsuda JL, Lenz LL. Myeloid Cell CK2 Regulates Inflammation and Resistance to Bacterial Infection. Front Immunol 2020; 11:590266. [PMID: 33363536 PMCID: PMC7752951 DOI: 10.3389/fimmu.2020.590266] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/05/2020] [Indexed: 12/27/2022] Open
Abstract
Kinase activity plays an essential role in the regulation of immune cell defenses against pathogens. The protein kinase CK2 (formerly casein kinase II) is an evolutionarily conserved kinase with hundreds of identified substrates. CK2 is ubiquitously expressed in somatic and immune cells, but the roles of CK2 in regulation of immune cell function remain largely elusive. This reflects the essential role of CK2 in organismal development and limited prior work with conditional CK2 mutant murine models. Here, we generated mice with a conditional (floxed) allele of Csnk2a, which encodes the catalytic CK2α subunit of CK2. When crossed to Lyz2-cre mice, excision of Csnk2a sequence impaired CK2α expression in myeloid cells but failed to detectably alter myeloid cell development. By contrast, deficiency for CK2α increased inflammatory myeloid cell recruitment, activation, and resistance following systemic Listeria monocytogenes (Lm) infection. Results from mixed chimera experiments indicated that CK2α deficiency in only a subset of myeloid cells was not sufficient to reduce bacterial burdens. Nor did cell-intrinsic deficiency for CK2α suffice to alter accumulation or activation of monocytes and neutrophils in infected tissues. These data suggest that CK2α expression by Lyz2-expressing cells promotes inflammatory and anti-bacterial responses through effects in trans. Our results highlight previously undescribed suppressive effects of CK2 activity on inflammatory myeloid cell responses and illustrate that cell-extrinsic effects of CK2 can shape inflammatory and protective innate immune responses.
Collapse
Affiliation(s)
- Sandy R. Larson
- Immunology and Microbiology Department, University of Colorado School of Medicine, Aurora, CO, United States
| | - Nikki Bortell
- Immunology and Microbiology Department, University of Colorado School of Medicine, Aurora, CO, United States
| | - Alysha Illies
- Immunology and Microbiology Department, University of Colorado School of Medicine, Aurora, CO, United States
| | - William J. Crisler
- Immunology and Microbiology Department, University of Colorado School of Medicine, Aurora, CO, United States
| | - Jennifer L. Matsuda
- Department of Biomedical Research, National Jewish Health, Denver, CO, United States
| | - Laurel L. Lenz
- Immunology and Microbiology Department, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Biomedical Research, National Jewish Health, Denver, CO, United States
| |
Collapse
|
39
|
Chojnacki K, Lindenblatt D, Wińska P, Wielechowska M, Toelzer C, Niefind K, Bretner M. Synthesis, biological properties and structural study of new halogenated azolo[4,5-b]pyridines as inhibitors of CK2 kinase. Bioorg Chem 2020; 106:104502. [PMID: 33317841 DOI: 10.1016/j.bioorg.2020.104502] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/06/2020] [Accepted: 11/19/2020] [Indexed: 11/26/2022]
Abstract
The new halogenated 1H-triazolo[4,5-b]pyridines and 1H-imidazo[4,5-b]pyridines were synthesised as analogues of known CK2 inhibitors: 4,5,6,7-tetrabromo-1H-benzotriazole (TBBt) and 4,5,6,7-tetrabromo-1H-benzimidazole (TBBi). Their influence on the activity of recombinant human CK2α, CK2α' and PIM1 kinases was determined. The most active inhibitors were di- and trihalogenated 1H-triazolo[4,5-b]pyridines (4a, 5a and 10a) with IC50 values 2.56, 3.82 and 3.26 μM respectively for CK2α. Furthermore, effect on viability of cancer cell lines MCF-7 (human breast adenocarcinoma) and CCRF-CEM (T lymphoblast leukemia) of all final compounds was evaluated. Finally, three crystal structures of complexes of CK2α1-335 with inhibitors 4a, 5a and 10a were obtained. In addition, new protocol was used to obtain high-resolution crystal structures of CK2α'Cys336Ser in complex with four inhibitors (4a, 5a, 5b, 10a).
Collapse
Affiliation(s)
- K Chojnacki
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland.
| | - D Lindenblatt
- Department für Chemie, Institut für Biochemie, Universtät zu Köln, Zülpicher Straße 47, D-50674 Köln, Germany
| | - P Wińska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| | - M Wielechowska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| | - C Toelzer
- Department für Chemie, Institut für Biochemie, Universtät zu Köln, Zülpicher Straße 47, D-50674 Köln, Germany
| | - K Niefind
- Department für Chemie, Institut für Biochemie, Universtät zu Köln, Zülpicher Straße 47, D-50674 Köln, Germany
| | - M Bretner
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| |
Collapse
|
40
|
Husain K, Williamson TT, Nelson N, Ghansah T. Protein kinase 2 (CK2): a potential regulator of immune cell development and function in cancer. Immunol Med 2020; 44:159-174. [PMID: 33164702 DOI: 10.1080/25785826.2020.1843267] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Protein kinase CK2, formally known as casein kinase II, is ubiquitously expressed and highly conserved serine/threonine or tyrosine kinase enzyme that regulates diverse signaling pathways responsible for cellular processes (i.e., cell proliferation and apoptosis) via interactions with over 500 known substrates. The enzyme's physiological interactions and cellular functions have been widely studied, most notably in the blood and solid malignancies. CK2 has intrinsic role in carcinogenesis as overexpression of CK2 subunits (α, α`, and β) and deregulation of its activity have been linked to various forms of cancers. CK2 also has extrinsic role in cancer stroma or in the tumor microenvironment (TME) including the immune cells. However, very few research studies have focused on extrinsic role of CK2 in regulating immune responses as a therapeutic alternative for cancer. The following review discusses CK2's regulation of key signaling events [Nuclear factor kappa B (NF-κB), Janus kinase/signal transducer and activators of transcription (JAK/STAT), Hypoxia inducible factor-1alpha (HIF-1α), Cyclooygenase-2 (COX-2), Extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK), Notch, Protein kinase B/AKT, Ikaros and Wnt] that can influence the development and function of immune cells in cancer. Potential clinical trials using potent CK2 inhibitors will facilitate and improve the treatment of human malignancies.
Collapse
Affiliation(s)
- Kazim Husain
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA
| | - Tanika T Williamson
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA
| | - Nadine Nelson
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA
| | - Tomar Ghansah
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
41
|
Montenarh M, Götz C. Protein kinase CK2 and ion channels (Review). Biomed Rep 2020; 13:55. [PMID: 33082952 PMCID: PMC7560519 DOI: 10.3892/br.2020.1362] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
Protein kinase CK2 appears as a tetramer or higher molecular weight oligomer composed of catalytic CK2α, CK2α' subunits and non-catalytic regulatory CK2β subunits or as individual subunits. It is implicated in a variety of different regulatory processes, such as Akt signalling, splicing and DNA repair within eukaryotic cells. The present review evaluates the influence of CK2 on ion channels in the plasma membrane. CK2 phosphorylates platform proteins such as calmodulin and ankyrin G, which bind to channel proteins for a physiological transport to and positioning into the membrane. In addition, CK2 directly phosphorylates a variety of channel proteins directly to regulate opening and closing of the channels. Thus, modulation of CK2 activities by specific inhibitors, by siRNA technology or by CRISPR/Cas technology has an influence on intracellular ion concentrations and thereby on cellular signalling. The physiological regulation of the intracellular ion concentration is important for cell survival and correct intracellular signalling. Disturbance of this regulation results in a variety of different diseases including epilepsy, heart failure, cystic fibrosis and diabetes. Therefore, these effects should be considered when using CK2 inhibition as a treatment option for cancer.
Collapse
Affiliation(s)
- Mathias Montenarh
- Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Saarland, Germany
| | - Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Saarland, Germany
| |
Collapse
|
42
|
Miller S, Hirota T. Pharmacological Interventions to Circadian Clocks and Their Molecular Bases. J Mol Biol 2020; 432:3498-3514. [DOI: 10.1016/j.jmb.2020.01.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/30/2019] [Accepted: 01/02/2020] [Indexed: 12/31/2022]
|
43
|
Schulze F, Müller S, Guli X, Schumann L, Brehme H, Riffert T, Rohde M, Goerss D, Rackow S, Einsle A, Kirschstein T, Köhling R. CK2 Inhibition Prior to Status Epilepticus Persistently Enhances K Ca2 Function in CA1 Which Slows Down Disease Progression. Front Cell Neurosci 2020; 14:33. [PMID: 32174814 PMCID: PMC7054465 DOI: 10.3389/fncel.2020.00033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 02/04/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose Epilepsy therapy is currently based on anti-seizure drugs that do not modify the course of the disease, i.e., they are not anti-epileptogenic in nature. Previously, we observed that in vivo casein kinase 2 (CK2) inhibition with 4,5,6,7-tetrabromotriazole (TBB) had anti-epileptogenic effects in the acute epilepsy slice model. Methods Here, we pretreated rats with TBB in vivo prior to the establishment of a pilocarpine-induced status epilepticus (SE) in order to analyze the long-term sequelae of such a preventive TBB administration. Results We found that TBB pretreatment delayed onset of seizures after pilocarpine and slowed down disease progression during epileptogenesis. This was accompanied with a reduced proportion of burst firing neurons in the CA1 area. Western blot analyses demonstrated that CA1 tissue from TBB-pretreated epileptic animals contained significantly less CK2 than TBB-pretreated controls. On the transcriptional level, TBB pretreatment led to differential gene expression changes of KCa2.2, but also of HCN1 and HCN3 channels. Thus, in the presence of the HCN channel blocker ZD7288, pretreatment with TBB rescued the afterhyperpolarizing potential (AHP) as well as spike frequency adaptation in epileptic animals, both of which are prominent functions of KCa2 channels. Conclusion These data indicate that TBB pretreatment prior to SE slows down disease progression during epileptogenesis involving increased KCa2 function, probably due to a persistently decreased CK2 protein expression.
Collapse
Affiliation(s)
- Felix Schulze
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Steffen Müller
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Xiati Guli
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Lukas Schumann
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Hannes Brehme
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Till Riffert
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Marco Rohde
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Doreen Goerss
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Simone Rackow
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Anne Einsle
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Center of Transdisciplinary Neurosciences Rostock, University of Rostock, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Center of Transdisciplinary Neurosciences Rostock, University of Rostock, Rostock, Germany
| |
Collapse
|
44
|
Marzec E, Poznański J, Paprocki D. Thermodynamic contribution of iodine atom to the binding of heterogeneously polyhalogenated benzotriazoles by the catalytic subunit of human protein kinase CK2. IUBMB Life 2020; 72:1203-1210. [PMID: 32083806 DOI: 10.1002/iub.2257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/09/2020] [Indexed: 12/29/2022]
Abstract
A series of novel benzotriazole derivatives containing iodine atom(s) were synthesized. The binding of these compounds to the catalytic subunit of human protein kinase CK2 was evaluated using differential scanning fluorimetry. The obtained thermodynamic data were compared with those determined previously for the brominated and chlorinated benzotriazole analogues to get a deeper insight into the thermodynamic contribution of iodine substitution to the free energy of ligand binding. We have shown that iodine atom(s) attached to the benzene ring of benzotriazole enhance(s) its binding by the target protein. This effect is the strongest when two iodine atoms are attached at positions peripheral to the triazole ring, which according to the structures deposited in protein data bank may be indicative for the formation of the halogen bond between iodine and carbonyl groups of residues located in the hinge region of the protein. Finally, quantitative structure-activity relationship analysis pointed the solute hydrophobicity as the main factor contributing to the binding affinity.
Collapse
Affiliation(s)
- Ewa Marzec
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Jarosław Poznański
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Daniel Paprocki
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
45
|
Zhang Y, Zhu Z, Ding H, Wan S, Zhang X, Li Y, Ji J, Wang X, Zhang M, Ye SD. β-catenin stimulates Tcf7l1 degradation through recruitment of casein kinase 2 in mouse embryonic stem cells. Biochem Biophys Res Commun 2020; 524:280-287. [PMID: 31987502 DOI: 10.1016/j.bbrc.2020.01.074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/13/2020] [Indexed: 11/18/2022]
Abstract
Activation of the Wnt/β-catenin signaling pathway by the inhibition of glycogen synthase kinase-3 (GSK-3) will induce Tcf7l1 protein degradation to effectively promote embryonic stem cell (ESC) self-renewal. However, the exact mechanism remains unclear. Here, we found that inhibition of casein kinase 2 (Csnk2) by TBB or DMAT was sufficient to block the reduction of the Tcf7l1 protein induced by CHIR99021, a specific inhibitor of GSK-3. Similarly, downregulation of Csnk2 increased the Tcf7l1 level. In contrast, overexpression of Csnk2 significantly decreased Tcf7l1 protein stability in mouse ESCs. Notably, Csnk2α1 controls Tcf7l1 turnover to a greater degree than the other two isoforms of Csnk2, Csnk2α2 and Csnk2β, as Csnk2α1-overexpressing mouse ESCs exhibited the lowest level of Tcf7l1. Csnk2α1 interacted with and phosphorylated Tcf7l1. In addition, the association of Csnk2α1 and Tcf7l1 was enhanced by CHIR99021. Our study demonstrated, for the first time, that Csnk2 is involved in Tcf7l1 turnover mediated by the Wnt/β-catenin signaling pathway. These results expand our understanding of the function and circuit of Wnt/β-catenin signaling pathway in ESCs.
Collapse
Affiliation(s)
- Yan Zhang
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Zhenhua Zhu
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Huiwen Ding
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Shengpeng Wan
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Xinbao Zhang
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Yuting Li
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Junxiang Ji
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Xin Wang
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Meng Zhang
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China
| | - Shou-Dong Ye
- Center for Stem Cell and Translational Medicine, School of Life Sciences & Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, PR China.
| |
Collapse
|
46
|
Ageshina AA, Chesnokov GA, Topchiy MA, Alabugin IV, Nechaev MS, Asachenko AF. Making endo-cyclizations favorable again: a conceptually new synthetic approach to benzotriazoles via azide group directed lithiation/cyclization of 2-azidoaryl bromides. Org Biomol Chem 2019; 17:4523-4534. [PMID: 30994147 DOI: 10.1039/c9ob00615j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Although benzotriazoles are important and ubiquitous, currently there is only one conceptual approach to their synthesis: bridging the two ortho-amino groups with an electrophilic nitrogen atom. Herein, we disclose a new practical alternative - the endo-cyclization of 2-azidoaryl lithiums obtained in situ from 2-azido-aryl bromides. The scope of the reaction is illustrated using twenty-four examples with a variety of alkyl, alkoxy, perfluoroalkyl, and halogen substituents. We found that the directing effect of the azide group allows selective metal-halogen exchange in aryl azides containing several bromine atoms. Furthermore, (2-bromophenyl)diazomethane undergoes similar cyclization to give an indazole. Thus, cyclizations of aryl lithiums containing an ortho-X = Y = Z group emerge as a new general approach for the synthesis of aromatic heterocycles. DFT computations suggested that the observed endo-selectivity applies to the anionic cyclizations of other functionalities that undergo "1,1-additions" (i.e., azides, diazo compounds, and isonitriles). In contrast, cyclizations with the heteroatomic functionalities that follow the "1,2-addition" pattern (cyanates, thiocyanates, isocyanates, isothiocyanates, and nitriles) prefer the exo-cyclization path. Hence, such reactions expand the current understanding of stereoelectronic factors in anionic cyclizations.
Collapse
Affiliation(s)
- Alexandra A Ageshina
- A.V. Topchiev Institute of Petrochemical Synthesis, Russian Academy of Sciences, 119991 Moscow, Leninskiy prospect 29, Russian Federation.
| | | | | | | | | | | |
Collapse
|
47
|
McKay S, Ryan TJ, McQueen J, Indersmitten T, Marwick KFM, Hasel P, Kopanitsa MV, Baxter PS, Martel MA, Kind PC, Wyllie DJA, O'Dell TJ, Grant SGN, Hardingham GE, Komiyama NH. The Developmental Shift of NMDA Receptor Composition Proceeds Independently of GluN2 Subunit-Specific GluN2 C-Terminal Sequences. Cell Rep 2019; 25:841-851.e4. [PMID: 30355491 PMCID: PMC6218242 DOI: 10.1016/j.celrep.2018.09.089] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 07/13/2018] [Accepted: 09/26/2018] [Indexed: 01/06/2023] Open
Abstract
The GluN2 subtype (2A versus 2B) determines biophysical properties and signaling of forebrain NMDA receptors (NMDARs). During development, GluN2A becomes incorporated into previously GluN2B-dominated NMDARs. This “switch” is proposed to be driven by distinct features of GluN2 cytoplasmic C-terminal domains (CTDs), including a unique CaMKII interaction site in GluN2B that drives removal from the synapse. However, these models remain untested in the context of endogenous NMDARs. We show that, although mutating the endogenous GluN2B CaMKII site has secondary effects on GluN2B CTD phosphorylation, the developmental changes in NMDAR composition occur normally and measures of plasticity and synaptogenesis are unaffected. Moreover, the switch proceeds normally in mice that have the GluN2A CTD replaced by that of GluN2B and commences without an observable decline in GluN2B levels but is impaired by GluN2A haploinsufficiency. Thus, GluN2A expression levels, and not GluN2 subtype-specific CTD-driven events, are the overriding factor in the developmental switch in NMDAR composition. Mutating the GluN2B CaMKII site affects phosphorylation of its C-terminal domain The developmental changes in NMDAR composition and synaptogenesis occur normally Changes in NMDAR composition do not require distinct GluN2 C-terminal domains Developmental changes in NMDAR composition are primarily sensitive to GluN2A levels
Collapse
Affiliation(s)
- Sean McKay
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; UK Dementia Research Institute at the University of Edinburgh, Chancellor's Building, Edinburgh Medical School, Edinburgh EH16 4SB, UK
| | - Tomás J Ryan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland; Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - Jamie McQueen
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; UK Dementia Research Institute at the University of Edinburgh, Chancellor's Building, Edinburgh Medical School, Edinburgh EH16 4SB, UK
| | - Tim Indersmitten
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Katie F M Marwick
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Philip Hasel
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; UK Dementia Research Institute at the University of Edinburgh, Chancellor's Building, Edinburgh Medical School, Edinburgh EH16 4SB, UK
| | - Maksym V Kopanitsa
- The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK; UK Dementia Research Institute at Imperial College London, Hammersmith Hospital Campus, Imperial College, London W12 0NN, UK
| | - Paul S Baxter
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; UK Dementia Research Institute at the University of Edinburgh, Chancellor's Building, Edinburgh Medical School, Edinburgh EH16 4SB, UK
| | - Marc-André Martel
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Peter C Kind
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - David J A Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Thomas J O'Dell
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Seth G N Grant
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK; Centre for Clinical Brain Sciences, University of Edinburgh Chancellor's Building, Edinburgh, UK
| | - Giles E Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; UK Dementia Research Institute at the University of Edinburgh, Chancellor's Building, Edinburgh Medical School, Edinburgh EH16 4SB, UK.
| | - Noboru H Komiyama
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; The Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK; Centre for Clinical Brain Sciences, University of Edinburgh Chancellor's Building, Edinburgh, UK.
| |
Collapse
|
48
|
Agnew C, Liu L, Liu S, Xu W, You L, Yeung W, Kannan N, Jablons D, Jura N. The crystal structure of the protein kinase HIPK2 reveals a unique architecture of its CMGC-insert region. J Biol Chem 2019; 294:13545-13559. [PMID: 31341017 PMCID: PMC6746438 DOI: 10.1074/jbc.ra119.009725] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/11/2019] [Indexed: 01/07/2023] Open
Abstract
The homeodomain-interacting protein kinase (HIPK) family is comprised of four nuclear protein kinases, HIPK1-4. HIPK proteins phosphorylate a diverse range of transcription factors involved in cell proliferation, differentiation, and apoptosis. HIPK2, thus far the best-characterized member of this largely understudied family of protein kinases, plays a role in the activation of p53 in response to DNA damage. Despite this tumor-suppressor function, HIPK2 is also found overexpressed in several cancers, and its hyperactivation causes chronic fibrosis. There are currently no structures of HIPK2 or of any other HIPK kinase. Here, we report the crystal structure of HIPK2's kinase domain bound to CX-4945, a casein kinase 2α (CK2α) inhibitor currently in clinical trials against several cancers. The structure, determined at 2.2 Å resolution, revealed that CX-4945 engages the HIPK2 active site in a hybrid binding mode between that seen in structures of CK2α and Pim1 kinases. The HIPK2 kinase domain crystallized in the active conformation, which was stabilized by phosphorylation of the activation loop. We noted that the overall kinase domain fold of HIPK2 closely resembles that of evolutionarily related dual-specificity tyrosine-regulated kinases (DYRKs). Most significant structural differences between HIPK2 and DYRKs included an absence of the regulatory N-terminal domain and a unique conformation of the CMGC-insert region and of a newly defined insert segment in the αC-β4 loop. This first crystal structure of HIPK2 paves the way for characterizing the understudied members of the HIPK family and for developing HIPK2-directed therapies for managing cancer and fibrosis.
Collapse
Affiliation(s)
- Christopher Agnew
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158
| | - Lijun Liu
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158
| | - Shu Liu
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94115
| | - Wei Xu
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94115
| | - Liang You
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94115
| | - Wayland Yeung
- Institute of Bioinformatics and Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| | - Natarajan Kannan
- Institute of Bioinformatics and Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| | - David Jablons
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94115, Supported by the Kazan McClain Partners' Foundation and the H. N. and Frances C. Berger Foundation. To whom correspondence may be addressed:
1600 Divisadero St., A745, San Francisco, CA 94115. Tel.:
415-353-7502; E-mail:
| | - Natalia Jura
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158,Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California 94158, To whom correspondence may be addressed:
555 Mission Bay Blvd. S., Rm. 452W, San Francisco, CA 94158. Tel.:
415-514-1133; E-mail:
| |
Collapse
|
49
|
Phosphorylation of p23-1 cochaperone by protein kinase CK2 affects root development in Arabidopsis. Sci Rep 2019; 9:9846. [PMID: 31285503 PMCID: PMC6614504 DOI: 10.1038/s41598-019-46327-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 06/25/2019] [Indexed: 12/27/2022] Open
Abstract
Root growth is a fundamental process in plants and assures nutrient and water uptake required for efficient photosynthesis and metabolism. Postembryonic development of roots is controlled by the functionality of the meristem. Several hormones and signaling molecules regulate the size of the meristem, and among them, auxins play a major role. Protein kinase CK2, along with the chaperone protein HSP90, has been found to be involved in the regulation of auxin transport. Here, we show that p23-1, a cochaperone of HSP90, is phosphorylated by CK2 in Arabidopsis. We identified Ser201 as the major CK2 target site in p23-1 and demonstrated that phosphorylation of this site is necessary for normal root development. Moreover, we shed light on the nature of CK2 in Arabidopsis, showing that the three catalytic isoforms, CK2 αA, αB and αC, are proteins of approximately 40 kDa. Our results increase knowledge of the connection among HSP90, p23-1 and CK2 in Arabidopsis, suggesting the existence of a possible common root development mechanism controlled by these signaling molecules.
Collapse
|
50
|
Pastor M, Zapico JM, Coderch C, Maslyk M, Panchuk R, de Pascual-Teresa B, Ramos A. From a MMP2/CK2 multitarget approach to the identification of potent and selective MMP13 inhibitors. Org Biomol Chem 2019; 17:916-929. [PMID: 30629065 DOI: 10.1039/c8ob02990c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In this article, we describe our efforts in the search of MMP2/CK2 dual targeting inhibitors. We have followed a rational drug design approach based on our experience in the selective inhibition of these two enzymes. We have successfully obtained highly active MMP2 (10, IC50 = 70 nM; 11, IC50 = 100 nM) and CK2 (16a, IC50 = 500 nM) inhibitors. However, structural fine tuning of these small molecules to simultaneously target both enzymes turned out to be an unattainable goal. Unexpectedly, we were lucky to identify new and selective MMP13 inhibitors (10, IC50 = 3.7 nM and 11, IC50 = 5.6 nM) with a novel TBB-derived scaffold. These compounds constitute an interesting starting point for further optimization.
Collapse
Affiliation(s)
- Miryam Pastor
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28925, Alcorcón, Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|