Scoping Pleiotropy of CK2 in Musculoskeletal Disorders for a Novel Targeting Approach
Abstract
:1. Introduction
2. Implication of CK2 in Musculoskeletal Disorders
2.1. Rheumatoid Arthritis
2.2. Osteoarthritis
2.3. Bone Fracture
2.4. Osteoporosis
3. Inhibition of CK2
3.1. Molecular Function of CK2
3.1.1. Structure of CK2 and Substrate Recognition
3.1.2. Function of Subunits
3.2. Small Molecular Inhibitors of CK2 and Their Mechanism of Action
4. Targeting CK2 Interactions with Its Substrates–Novel Approach
Disease-Modifying Peptide Drugs-CK2.1 and CK2.3
5. Discussion
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- El Batawi, M.A. Work-related diseases. A new program of the World Health Organization. Scand. J. Work. Environ. Health 1984, 10, 341–346. [Google Scholar] [CrossRef]
- Khan, M.A.; Zubair, H.; Anand, S.; Srivastava, S.K.; Singh, S.; Singh, A.P. Dysregulation of metabolic enzymes in tumor and stromal cells: Role in oncogenesis and therapeutic opportunities. Cancer Lett. 2020, 473, 176–185. [Google Scholar] [CrossRef]
- Egami, R.; Kokaji, T.; Hatano, A.; Yugi, K.; Eto, M.; Morita, K.; Ohno, S.; Fujii, M.; Hironaka, K.-I.; Uematsu, S.; et al. Trans-omic analysis reveals obesity-associated dysregulation of inter-organ metabolic cycles between the liver and skeletal muscle. iScience 2021, 24, 102217. [Google Scholar] [CrossRef]
- Comertpay, B.; Gov, E. Immune cell-specific and common molecular signatures in rheumatoid arthritis through molecular network approaches. Biosystems 2023, 234, 105063. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.; Li, S.; Cao, N.; Wang, Q.; Liu, Y.; Xu, Q.; Zhang, L.; Sun, C.; Xiao, X.; Yao, J. Intestinal flora, intestinal metabolism, and intestinal immunity changes in complete Freud’s adjuvant-rheumatoid arthritis C57BL/6 mice. Int. Immunopharmacol. 2023, 125 Pt A, 111090. [Google Scholar] [CrossRef]
- Mobasheri, A.; Rayman, M.P.; Gualillo, O.; Sellam, J.; van der Kraan, P.; Fearon, U. The role of metabolism in the pathogenesis of osteoarthritis. Nat. Rev. Rheumatol. 2017, 13, 302–311. [Google Scholar] [CrossRef]
- Cho, Y.; Jeong, S.; Kim, H.; Kang, D.; Lee, J.; Kang, S.-B.; Kim, J.-H. Disease-modifying therapeutic strategies in osteoarthritis: Current status and future directions. Exp. Mol. Med. 2021, 53, 1689–1696. [Google Scholar] [CrossRef]
- Liao, Z.; Han, X.; Wang, Y.; Shi, J.; Zhang, Y.; Zhao, H.; Zhang, L.; Jiang, M.; Liu, M. Differential Metabolites in Osteoarthritis: A Systematic Review and Meta-Analysis. Nutrients 2023, 15, 4191. [Google Scholar] [CrossRef] [PubMed]
- Su, Z.; Zong, Z.; Deng, J.; Huang, J.; Liu, G.; Wei, B.; Cui, L.; Li, G.; Zhong, H.; Lin, S. Lipid Metabolism in Cartilage Development, Degeneration, and Regeneration. Nutrients 2022, 14, 3984. [Google Scholar] [CrossRef] [PubMed]
- Xi, Y.; Shen, J.; Li, X.; Bao, Y.; Zhao, T.; Li, B.; Qin, D. Regulatory Effects of Quercetin on Bone Homeostasis: Research Updates and Future Perspectives. Am. J. Chin. Med. 2023, 51, 2077–2094. [Google Scholar] [CrossRef]
- Robin, F.; Chappard, D.; Leroyer, P.; Latour, C.; Mabilleau, G.; Monbet, V.; Cavey, T.; Horeau, M.; Derbré, F.; Roth, M.; et al. Differences in bone microarchitecture between genetic and secondary iron-overload mouse models suggest a role for hepcidin deficiency in iron-related osteoporosis. FASEB J. 2023, 37, e23245. [Google Scholar] [CrossRef] [PubMed]
- Alghadir, A.H.; Gabr, S.A.; Iqbal, A. Hand grip strength, vitamin D status, and diets as predictors of bone health in 6–12 years old school children. BMC Musculoskelet. Disord. 2023, 24, 830. [Google Scholar] [CrossRef] [PubMed]
- Ribeiro, M.S.; Venturini, L.G.; Speck-Hernandez, C.A.; Alabarse, P.V.; Xavier, T.; Taira, T.M.; Duffles, L.F.; Cunha, F.Q.; Fukada, S.Y. AMPKα1 negatively regulates osteoclastogenesis and mitigates pathological bone loss. J. Biol. Chem. 2023, 299, 105379. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Z.; Liu, M.; Zhang, Y.; Wu, J.; Gao, M.; Lei, T.; Huang, F.; Chen, H.; Wu, M. Risk factors for the comorbidity of osteoporosis/osteopenia and kidney stones: A cross-sectional study. Arch. Osteoporos. 2023, 18, 128. [Google Scholar] [CrossRef] [PubMed]
- Yoon, H.; Sung, E.; Kang, J.-H.; Kim, C.-H.; Shin, H.; Yoo, E.; Kim, M.; Lee, M.Y.; Shin, S. Association between body fat and bone mineral density in Korean adults: A cohort study. Sci. Rep. 2023, 13, 17462. [Google Scholar] [CrossRef] [PubMed]
- Claes, L.; Recknagel, S.; Ignatius, A. Fracture healing under healthy and inflammatory conditions. Nat. Rev. Rheumatol. 2012, 8, 133–143. [Google Scholar] [CrossRef] [PubMed]
- Khajuria, D.K.; Reider, I.; Kamal, F.; Norbury, C.C.; Elbarbary, R.A. Distinct defects in early innate and late adaptive immune responses typify impaired fracture healing in diet-induced obesity. Front. Immunol. 2023, 14, 1250309. [Google Scholar] [CrossRef] [PubMed]
- Borgo, C.; D’amore, C.; Cesaro, L.; Sarno, S.; Pinna, L.A.; Ruzzene, M.; Salvi, M. How can a traffic light properly work if it is always green? The paradox of CK2 signaling. Crit. Rev. Biochem. Mol. Biol. 2021, 56, 321–359. [Google Scholar] [CrossRef]
- Ruzzene, M.; Pinna, L.A. Addiction to protein kinase CK2: A common denominator of diverse cancer cells? Biochim. Biophys. Acta 2010, 1804, 499–504. [Google Scholar] [CrossRef]
- St-Denis, N.A.; Litchfield, D.W. Protein kinase CK2 in health and disease: From birth to death: The role of protein kinase CK2 in the regulation of cell proliferation and survival. Cell Mol. Life Sci. 2009, 66, 1817–1829. [Google Scholar] [CrossRef]
- Halloran, D.; Pandit, V.; Nohe, A. The Role of Protein Kinase CK2 in Development and Disease Progression: A Critical Review. J. Dev. Biol. 2022, 10, 31. [Google Scholar] [CrossRef] [PubMed]
- Borgo, C.; D’amore, C.; Sarno, S.; Salvi, M.; Ruzzene, M. Protein kinase CK2: A potential therapeutic target for diverse human diseases. Signal Transduct. Target. Ther. 2021, 6, 183. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Lu, C.; Wu, X.; Zhang, Z.; Li, J.; Guo, B.; Li, D.; Liang, C.; Dang, L.; Pan, X.; et al. Targeting osteoblastic casein kinase-2 interacting protein-1 to enhance Smad-dependent BMP signaling and reverse bone formation reduction in glucocorticoid-induced osteoporosis. Sci. Rep. 2017, 7, 41295. [Google Scholar] [CrossRef] [PubMed]
- Rea, I.M.; Gibson, D.S.; McGilligan, V.; McNerlan, S.E.; Alexander, H.D.; Ross, O.A. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front. Immunol. 2018, 9, 586. [Google Scholar] [CrossRef] [PubMed]
- McGarry, T.; Biniecka, M.; Veale, D.J.; Fearon, U. Hypoxia, oxidative stress and inflammation. Free Radic. Biol. Med. 2018, 125, 15–24. [Google Scholar] [CrossRef]
- Song, J.; Bae, Y.S. CK2 Down-Regulation Increases the Expression of Senescence-Associated Secretory Phenotype Factors through NF-κB Activation. Int. J. Mol. Sci. 2021, 22, 406. [Google Scholar] [CrossRef]
- Pagano, M.A.; Andrzejewska, M.; Ruzzene, M.; Sarno, S.; Cesaro, L.; Bain, J.; Elliott, M.; Meggio, F.; Kazimierczuk, Z.; Pinna, L.A. Optimization of Protein Kinase CK2 Inhibitors Derived from 4,5,6,7-Tetrabromobenzimidazole. J. Med. Chem. 2004, 47, 6239–6247. [Google Scholar] [CrossRef]
- Sarno, S.; Pinna, L.A. Protein kinase CK2 as a druggable target. Mol. Biosyst. 2008, 4, 889–894. [Google Scholar] [CrossRef]
- Borgo, C.; Ruzzene, M. Protein kinase CK2 inhibition as a pharmacological strategy. Adv. Protein Chem. Struct. Biol. 2021, 124, 23–46. [Google Scholar] [PubMed]
- Meggio, F.; Pinna, L.A. One-thousand-and-one substrates of protein kinase CK2? FASEB J. 2003, 17, 349–368. [Google Scholar] [CrossRef] [PubMed]
- Franchin, C.; Borgo, C.; Cesaro, L.; Zaramella, S.; Vilardell, J.; Salvi, M.; Arrigoni, G.; Pinna, L.A. Re-evaluation of protein kinase CK2 pleiotropy: New insights provided by a phosphoproteomics analysis of CK2 knockout cells. Cell. Mol. Life Sci. 2018, 75, 2011–2026. [Google Scholar] [CrossRef] [PubMed]
- Salvi, M.; Cesaro, L.; Pinna, L.A. Variable contribution of protein kinases to the generation of the human phosphoproteome: A global weblogo analysis. Biomol. Concepts 2010, 1, 185–195. [Google Scholar] [CrossRef] [PubMed]
- Olsen, B.B.; Guerra, B.; Niefind, K.; Issinger, O.G. Structural basis of the constitutive activity of protein kinase CK2. Methods Enzymol. 2010, 484, 515–529. [Google Scholar] [PubMed]
- Pinna, L.A. Protein kinase CK2: A challenge to canons. J. Cell Sci. 2002, 115 Pt 20, 3873–3878. [Google Scholar] [CrossRef] [PubMed]
- Pinna, L.A. Protein kinase CK2. Int. J. Biochem. Cell Biol. 1997, 29, 551–554. [Google Scholar] [CrossRef]
- Filhol, O.; Cochet, C. Protein kinase CK2 in health and disease: Cellular functions of protein kinase CK2: A dynamic affair. Cell Mol. Life Sci. 2009, 66, 1830–1839. [Google Scholar] [CrossRef] [PubMed]
- Niefind, K.; Guerra, B.; Ermakowa, I.; Issinger, O. Crystal structure of human protein kinase CK2: Insights into basic properties of the CK2 holoenzyme. EMBO J. 2001, 20, 5320–5331. [Google Scholar] [CrossRef]
- Olsten, M.E.; Litchfield, D.W. Order or chaos? An evaluation of the regulation of protein kinase CK2. Biochem. Cell Biol. 2004, 82, 681–693. [Google Scholar] [CrossRef]
- Montenarh, M.; Götz, C. Protein Kinase CK2α’, More than a Backup of CK2α. Cells 2023, 12, 2834. [Google Scholar] [CrossRef]
- Pierre, F.; Chua, P.C.; O’Brien, S.E.; Siddiqui-Jain, A.; Bourbon, P.; Haddach, M.; Michaux, J.; Nagasawa, J.; Schwaebe, M.K.; Stefan, E.; et al. Discovery and SAR of 5-(3-Chlorophenylamino)benzo[c][2,6]naphthyridine-8-carboxylic Acid (CX-4945), the First Clinical Stage Inhibitor of Protein Kinase CK2 for the Treatment of Cancer. J. Med. Chem. 2011, 54, 635–654. [Google Scholar] [CrossRef] [PubMed]
- Zanin, S.; Sandre, M.; Cozza, G.; Ottaviani, D.; Marin, O.; Pinna, L.A.; Ruzzene, M. Chimeric peptides as modulators of CK2-dependent signaling: Mechanism of action and off-target effects. Biochim. Biophys. Acta 2015, 1854 Pt B, 1694–1707. [Google Scholar] [CrossRef]
- Perea, S.E.; Baladrón, I.; Valenzuela, C.; Perera, Y. CIGB-300: A peptide-based drug that impairs the Protein Kinase CK2-mediated phosphorylation. Semin. Oncol. 2018, 45, 58–67. [Google Scholar] [CrossRef] [PubMed]
- Pérez, G.V.; Rosales, M.; Ramón, A.C.; Rodríguez-Ulloa, A.; Besada, V.; González, L.J.; Aguilar, D.; Vázquez-Blomquist, D.; Falcón, V.; Caballero, E.; et al. CIGB-300 Anticancer Peptide Differentially Interacts with CK2 Subunits and Regulates Specific Signaling Mediators in a Highly Sensitive Large Cell Lung Carcinoma Cell Model. Biomedicines 2022, 11, 43. [Google Scholar] [CrossRef] [PubMed]
- Cirigliano, S.M.; Bessone, M.I.D.; Berardi, D.E.; Flumian, C.; Bal de Kier Joffé, E.D.; Perea, S.E.; Farina, H.G.; Todaro, L.B.; Urtreger, A.J. The synthetic peptide CIGB-300 modulates CK2-dependent signaling pathways affecting the survival and chemoresistance of non-small cell lung cancer cell lines. Cancer Cell Int. 2017, 17, 42. [Google Scholar] [CrossRef] [PubMed]
- Sarduy, M.R.; García, I.; Coca, M.A.; Perera, A.; Torres, L.A.; Valenzuela, C.M.; Baladrón, I.; Solares, M.; Reyes, V.; Hernández, I.; et al. Optimizing CIGB-300 intralesional delivery in locally advanced cervical cancer. Br. J. Cancer 2015, 112, 1636–1643. [Google Scholar] [CrossRef] [PubMed]
- Vázquez-Blomquist, D.; Ramón, A.C.; Rosales, M.; Pérez, G.V.; Rosales, A.; Palenzuela, D.; Perera, Y.; Perea, S.E. Gene expression profiling unveils the temporal dynamics of CIGB-300-regulated transcriptome in AML cell lines. BMC Genom. 2023, 24, 373. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez-Merchan, E.C. The Current Role of Disease-modifying Osteoarthritis Drugs. Arch. Bone Jt. Surg. 2023, 11, 11–22. [Google Scholar]
- Song, Z.; Li, Y.; Shang, C.; Shang, G.; Kou, H.; Li, J.; Chen, S.; Liu, H. Sprifermin: Effects on Cartilage Homeostasis and Therapeutic Prospects in Cartilage-Related Diseases. Front. Cell Dev. Biol. 2021, 9, 786546. [Google Scholar] [CrossRef]
- Maksimovic, V.; Pavlovic-Popovic, Z.; Vukmirovic, S.; Cvejic, J.; Mooranian, A.; Al-Salami, H.; Mikov, M.; Golocorbin-Kon, S. Molecular mechanism of action and pharmacokinetic properties of methotrexate. Mol. Biol. Rep. 2020, 47, 4699–4708. [Google Scholar] [CrossRef]
- Chua, M.M.J.; Lee, M.; Dominguez, I. Cancer-type dependent expression of CK2 transcripts. PLoS ONE 2017, 12, e0188854. [Google Scholar] [CrossRef]
- Sun, D.; Gao, W.; Hu, H.; Zhou, S. Why 90% of clinical drug development fails and how to improve it? Acta Pharm. Sin B 2022, 12, 3049–3062. [Google Scholar] [CrossRef]
- Lee, D.M.; Weinblatt, M.E. Rheumatoid arthritis. Lancet 2001, 358, 903–911. [Google Scholar] [CrossRef]
- Gibofsky, A. Epidemiology, pathophysiology, and diagnosis of rheumatoid arthritis: A Synopsis. Am. J. Manag. Care 2014, 20, S128–S135. [Google Scholar]
- Huang, J.; Fu, X.; Chen, X.; Li, Z.; Huang, Y.; Liang, C. Promising Therapeutic Targets for Treatment of Rheumatoid Arthritis. Front. Immunol. 2021, 12, 686155. [Google Scholar] [CrossRef]
- Mcinnes, I.B.; Schett, G. The pathogenesis of rheumatoid arthritis. N. Engl. J. Med. 2011, 365, 2205–2219. [Google Scholar] [CrossRef] [PubMed]
- Radu, A.; Bungao, S.G. Management of Rheumatoid Arthritis: An Overview. Cells 2021, 10, 2857. [Google Scholar] [CrossRef] [PubMed]
- Luo, Y.; Lei, Y.; Guo, X.; Zhu, D.; Zhang, H.; Guo, Z.; Xu, Z.; Zhao, H.; Xi, Y.; Peng, X.; et al. CX-4945 inhibits fibroblast-like synoviocytes functions through the CK2-p53 axis to reduce rheumatoid arthritis disease severity. Int. Immunopharmacol. 2023, 119, 110163. [Google Scholar] [CrossRef] [PubMed]
- Bartok, B.; Firestein, G.S. Fibroblast-like synoviocytes: Key effector cells in rheumatoid arthritis. Immunol. Rev. 2009, 1, 233–255. [Google Scholar] [CrossRef] [PubMed]
- Yang, W.; Gibson, S.A.; Yan, Z. Protein kinase 2 (CK2) controls CD4(+) T cell effector function in the pathogenesis of colitis. Mucosal Immunol. 2020, 13, 788–798. [Google Scholar] [CrossRef] [PubMed]
- Ulges, A.; Klein, M.; Reuters, S. Protein kinase CK2 enables regulatory T cells to suppress excessive TH2 responses in vivo. Nat. Immunol. 2015, 16, 267–275. [Google Scholar] [CrossRef] [PubMed]
- Zheng, L.; Zhang, Z.; Sheng, P.; Mobasheri, A. The role of metabolism in chondrocyte dysfunction and the progression of osteoarthritis. Ageing Res. Rev. 2021, 66, 101249. [Google Scholar] [CrossRef] [PubMed]
- Henrotin, Y.; Kurz, B.; Aigner, T. Oxygen and reactive oxygen species in cartilage degradation: Friends or foes? Osteoarthr. Cartil. 2005, 13, 643–654. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.W.; Song, Y.S.; Lee, S.Y.; Yoon, Y.G.; Lee, S.H.; Park, B.S.; Yun, I.; Choi, H.; Kim, K.; Chung, W.T.; et al. Downregulation of protein kinase CK2 activity facilitates tumor necrosis factor-α-mediated chondrocyte death through apoptosis and autophagy. PLoS ONE 2011, 6, e19163. [Google Scholar] [CrossRef] [PubMed]
- Nishimori, S.; Lai, F.; Shiraishi, M.; Kobayashi, T.; Kozhemyakina, E.; Yao, T.-P.; Lassar, A.B.; Kronenberg, H.M. PTHrP targets HDAC4 and HDAC5 to repress chondrocyte hypertrophy. J. Clin. Investig. 2019, 4, e97903. [Google Scholar] [CrossRef] [PubMed]
- Hui, W.; A Young, D.; Rowan, A.D.; Xu, X.; E Cawston, T.; Proctor, C.J. Oxidative changes and signalling pathways are pivotal in initiating age-related changes in articular cartilage. Ann. Rheum. Dis. 2016, 75, 449–458. [Google Scholar] [CrossRef] [PubMed]
- Librizzi, M.; Naselli, F.; Abruscato, G.; Luparello, C.; Caradonna, F. Parathyroid Hormone Related Protein (PTHrP)-Associated Molecular Signatures in Tissue Differentiation and Non-Tumoral Diseases. Biology 2023, 12, 950. [Google Scholar] [CrossRef]
- Aarts, M.M.; Levy, D.; He, B.; Stregger, S.; Chen, T.; Richard, S.; Henderson, J.E. Parathyroid Hormone-related Protein Interacts with RNA. J. Biol. Chem. 1999, 274, 4832–4838. [Google Scholar] [CrossRef]
- Okoumassoun, L.E.; Russo, C.; Denizeau, F.; Averill-Bates, D.; Henderson, J.E. Parathyroid hormone-related protein (PTHrP) inhibits mitochondrial-dependent apoptosis through CK2. J. Cell. Physiol. 2007, 212, 591–599. [Google Scholar] [CrossRef]
- Saha, S.; Buttari, B.; Panieri, E.; Profumo, E.; Saso, L. An Overview of Nrf2 Signaling Pathway and Its Role in Inflammation. Molecules 2020, 25, 5474. [Google Scholar] [CrossRef]
- Soares, M.P.; Seldon, M.P.; Gregoire, I.P.; Vassilevskaia, T.; Berberat, P.O.; Yu, J.; Tsui, T.-Y.; Bach, F.H. Heme Oxygenase-1 Modulates the Expression of Adhesion Molecules Associated with Endothelial Cell Activation. J. Immunol. 2004, 172, 3553–3563. [Google Scholar] [CrossRef]
- Park, Y.C.; Kim, K.M.; Song, J.D.; Chung, H.T. Protein kinase CK2 mediates peroxynitrite-induced heme oxygenase-1 expression in articular chondrocytes. Int. J. Mol. Med. 2012, 29, 1039–1044. [Google Scholar] [CrossRef]
- Kim, K.M.; Sohn, D.H.; Kim, K.; Park, Y.C. Inhibition of protein kinase CK2 facilitates cellular senescence by inhibiting the expression of HO-1 in articular chondrocytes. Int. J. Mol. Med. 2019, 43, 1033–1040. [Google Scholar] [CrossRef]
- 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. 2022, 65, 1302–1312. [Google Scholar] [CrossRef]
- Lee, S.W.; Rho, J.H.; Lee, S.Y.; Yoo, S.H.; Kim, H.Y.; Chung, W.T.; Yoo, Y.H. Alpha B-Crystallin Protects Rat Articular Chondrocytes against Casein Kinase II Inhibition-Induced Apoptosis. PLoS ONE 2016, 11, e0166450. [Google Scholar] [CrossRef]
- Muire, P.J.; Mangum, L.H.; Wenke, J.C. Time Course of Immune Response and Immunomodulation During Normal and Delayed Healing of Musculoskeletal Wounds. Front. Immunol. 2020, 11, 1056. [Google Scholar] [CrossRef]
- Prein, C.; Beier, F. ECM signaling in cartilage development and endochondral ossification. Curr. Top. Dev. Biol. 2019, 133, 25–47. [Google Scholar] [PubMed]
- PPajarinen, J.; Lin, T.; Gibon, E.; Kohno, Y.; Maruyama, M.; Nathan, K.; Lu, L.; Yao, Z.; Goodman, S.B. Mesenchymal stem cell-macrophage crosstalk and bone healing. Biomaterials 2019, 196, 80–89. [Google Scholar] [CrossRef] [PubMed]
- Recknagel, S.D.; Bindl, R.; Brochhausen, C.; Göckelmann, M.D.; Wehner, T.; Schoengraf, P.; Huber-Lang, M.; Claes, L.; Ignatius, A.D. Systemic inflammation induced by a thoracic trauma alters the cellular composition of the early fracture callus. J. Trauma Inj. Infect. Crit. Care 2013, 74, 531–537. [Google Scholar] [CrossRef] [PubMed]
- Saul, D.; Khosla, S. Fracture Healing in the Setting of Endocrine Diseases, Aging, and Cellular Senescence. Endocr. Rev. 2022, 43, 984–1002. [Google Scholar] [CrossRef] [PubMed]
- Takayama, H.; La Rochelle, W.J.; Anver, M.; E Bockman, D.; Merlino, G. Scatter factor/hepatocyte growth factor as a regulator of skeletal muscle and neural crest development. Proc. Natl. Acad. Sci. USA 1996, 93, 5866–5871. [Google Scholar] [CrossRef]
- Majidinia, M.; Sadeghpour, A.; Yousefi, B. The roles of signaling pathways in bone repair and regeneration. J. Cell Physiol. 2018, 233, 2937–2948. [Google Scholar] [CrossRef] [PubMed]
- Dumic-Cule, I.; Peric, M.; Kucko, L.; Grgurevic, L.; Pecina, M.; Vukicevic, S. Bone morphogenetic proteins in fracture repair. Int. Orthop. 2018, 42, 2619–2626. [Google Scholar] [CrossRef] [PubMed]
- Cho, J.H.; Lee, J.H.; Lee, K.M.; Lee, C.-K.; Shin, D.-M. BMP-2 Induced Signaling Pathways and Phenotypes: Comparisons Between Senescent and Non-senescent Bone Marrow Mesenchymal Stem Cells. Calcif. Tissue Int. 2021, 110, 489–503. [Google Scholar] [CrossRef] [PubMed]
- Bragdon, B.; Thinakaran, S.; Moseychuk, O.; King, D.; Young, K.; Litchfield, D.W.; Petersen, N.O.; Nohe, A. Casein kinase 2 beta-subunit is a regulator of bone morphogenetic protein 2 signaling. Biophys. J. 2010, 99, 897–904. [Google Scholar] [CrossRef] [PubMed]
- Yu, B.; Huo, L.; Liu, Y.; Deng, P.; Szymanski, J.; Li, J.; Luo, X.; Hong, C.; Lin, J.; Wang, C.Y. PGC-1α Controls Skeletal Stem Cell Fate and Bone-Fat Balance in Osteoporosis and Skeletal Aging by Inducing TAZ. Cell Stem Cell 2018, 23, 193–209.e5. [Google Scholar] [CrossRef]
- Nie, J.; Liu, L.; He, F.; Fu, X.; Han, W.; Zhang, L. CKIP-1: A scaffold protein and potential therapeutic target integrating multiple signaling pathways and physiological functions. Ageing Res. Rev. 2013, 12, 276–281. [Google Scholar] [CrossRef]
- Fu, L.; Zhang, L. Physiological functions of CKIP-1: From molecular mechanisms to therapy implications. Ageing Res. Rev. 2019, 53, 100908. [Google Scholar] [CrossRef]
- Yuan, Y.; Sun, J.; Zhou, H.; Wang, S.; He, C.; Chen, T.; Fang, M.; Li, S.; Kang, S.; Huang, X.; et al. The effect of QiangGuYin on osteoporosis through the AKT/mTOR/autophagy signaling pathway mediated by CKIP-1. Aging 2022, 14, 892–906. [Google Scholar] [CrossRef]
- Peng, X.; Wu, X.; Zhang, J.; Zhang, G.; Li, G.; Pan, X. The role of CKIP-1 in osteoporosis development and treatment. Bone Jt. Res. 2018, 7, 173–178. [Google Scholar] [CrossRef]
- Litchfield, D.W. Protein kinase CK2: Structure, regulation and role in cellular decisions of life and death. Biochem. J. 2003, 369 Pt 1, 1–15. [Google Scholar] [CrossRef]
- Salvi, M.; Sarno, S.; Cesaro, L.; Nakamura, H.; Pinna, L.A. Extraordinary pleiotropy of protein kinase CK2 revealed by weblogo phosphoproteome analysis. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2009, 1793, 847–859. [Google Scholar] [CrossRef]
- Franchin, C.; Borgo, C.; Zaramella, S.; Cesaro, L.; Arrigoni, G.; Salvi, M.; Pinna, L.A. Exploring the CK2 Paradox: Restless, Dangerous, Dispensable. Pharmaceuticals 2017, 10, 11. [Google Scholar] [CrossRef]
- Chojnowski, J.E.; Mcmillan, E.A.; Strochlic, T.I. Identification of Novel CK2 Kinase Substrates Using a Versatile Biochemical Approach. J. Vis. Exp. 2019, 21, e59037. [Google Scholar]
- Borgo, C.; Cesaro, L.; Hirota, T.; Kuwata, K.; D’amore, C.; Ruppert, T.; Blatnik, R.; Salvi, M.; Pinna, L.A. Comparing the efficacy and selectivity of Ck2 inhibitors a phosphoproteomics approach. Eur. J. Med. Chem. 2021, 214, 113217. [Google Scholar] [CrossRef] [PubMed]
- Gyenis, L.; Menyhart, D.; Cruise, E.S.; Jurcic, K.; Roffey, S.E.; Chai, D.B.; Trifoi, F.; Fess, S.R.; Desormeaux, P.J.; Díaz, T.N.d.V.; et al. Chemical Genetic Validation of CSNK2 Substrates Using an Inhibitor-Resistant Mutant in Combination with Triple SILAC Quantitative Phosphoproteomics. Front. Mol. Biosci. 2022, 9, 909711. [Google Scholar] [CrossRef]
- Borgo, C.; Cesaro, L.; Hirota, T.; Kuwata, K.; D’Amore, C.; Ruppert, T.; Blatnik, R.; Salvi, M.; Pinna, L.A. Analysis of the phosphoproteome of CK2α((-/-))/Δα’ C2C12 myoblasts compared to the wild-type cells. Open Biol. 2023, 13, 220220. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.; Choi, K.; Kang, H.; Lee, S.-Y.; Chi, S.-W.; Lee, M.-S.; Song, J.; Im, D.; Choi, Y.; Cho, S. Identification of a Novel Function of CX-4945 as a Splicing Regulator. PLoS ONE 2014, 9, e94978. [Google Scholar] [CrossRef] [PubMed]
- Merholz, M.; Jian, Y.; Wimberg, J.; Gessler, L.; Hashemolhosseini, S. In Skeletal Muscle Fibers, Protein Kinase Subunit CSNK2A1/CK2α Is Required for Proper Muscle Homeostasis and Structure and Function of Neuromuscular Junctions. Cells 2022, 11, 3962. [Google Scholar] [CrossRef]
- Salvi, M.; Borgo, C.; Pinna, L.A.; Ruzzene, M. Targeting CK2 in cancer: A valuable strategy or a waste of time? Cell Death Discov. 2021, 7, 325. [Google Scholar] [CrossRef] [PubMed]
- Trembley, J.H.; Kren, B.T.; Afzal, M.; Scaria, G.A.; Klein, M.A.; Ahmed, K. Protein kinase CK2—Diverse roles in cancer cell biology and therapeutic promise. Mol. Cell. Biochem. 2022, 478, 899–926. [Google Scholar] [CrossRef]
- Jiang, Q.; Yang, G.; Liu, Q.; Wang, S.; Cui, D. Function and Role of Regulatory T Cells in Rheumatoid Arthritis. Front. Immunol. 2021, 12, 626193. [Google Scholar] [CrossRef]
- Ye, H.; Fu, D.; Fang, X.; Xie, Y.; Zheng, X.; Fan, W.; Hu, F.; Li, Z. Casein Kinase II exacerbates rheumatoid arthritis via promoting Th1 and Th17 cell inflammatory responses. Expert. Opin. Ther. Targets 2021, 25, 1017–1024. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.Y.; Yun, J.-S.; Kim, W.-K.; Chun, H.-S.; Jin, H.; Cho, S.; Chang, J.H. Structural Basis for the Selective Inhibition of Cdc2-Like Kinases by CX-4945. BioMed Res. Int. 2019, 2019, 6125068. [Google Scholar] [CrossRef]
- Ma, Y.; Sender, S.; Sekora, A.; Kong, W.; Bauer, P.; Ameziane, N.; Krake, S.; Radefeldt, M.; Al-Ali, R.; Weiss, F.U.; et al. Inhibitory Response to CK II Inhibitor Silmitasertib and CDKs Inhibitor Dinaciclib Is Related to Genetic Differences in Pancreatic Ductal Adenocarcinoma Cell Lines. Int. J. Mol. Sci. 2022, 23, 4409. [Google Scholar] [CrossRef]
- Lertsuwan, J.; Lertsuwan, K.; Sawasdichai, A.; Tasnawijitwong, N.; Lee, K.Y.; Kitchen, P.; Afford, S.; Gaston, K.; Jayaraman, P.S.; Satayavivad, J. CX-4945 Induces Methuosis in Cholangiocarcinoma Cell Lines by a CK2-Independent Mechanism. Cancers 2018, 10, 283. [Google Scholar] [CrossRef]
- Silva-Pavez, E.; Villar, P.; Trigo, C.; Caamaño, E.; Niechi, I.; Pérez, P.; Muñoz, J.P.; Aguayo, F.; Burzio, V.A.; Varas-Godoy, M.; et al. CK2 inhibition with silmitasertib promotes methuosis-like cell death associated to catastrophic massive vacuolization of colorectal cancer cells. Cell Death Dis. 2019, 10, 73. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Son, Y.H.; Moon, S.H.; Kim, J. The Protein Kinase 2 Inhibitor CX-4945 Regulates Osteoclast and Osteoblast Differentiation In Vitro. Mol. Cells 2013, 36, 417–423. [Google Scholar] [CrossRef]
- Chen, X.; Li, C.; Wang, D.; Chen, Y.; Zhang, N. Recent Advances in the Discovery of CK2 Allosteric Inhibitors: From Traditional Screening to Structure-Based Design. Molecules 2020, 25, 870. [Google Scholar] [CrossRef]
- Moucadel, V.; Prudent, R.; Sautel, C.F.; Teillet, F.; Barette, C.; Lafanechere, L.; Receveur-Brechot, V.; Cochet, C. Antitumoral activity of allosteric inhibitors of protein kinase CK2. Oncotarget 2011, 2, 997–1010. [Google Scholar] [CrossRef]
- Li, C.; Zhang, X.; Zhang, N.; Zhou, Y.; Sun, G.; Zhao, L.; Zhong, R. Identification and Biological Evaluation of CK2 Allosteric Fragments through Structure-Based Virtual Screening. Molecules 2020, 25, 237. [Google Scholar] [CrossRef]
- Brear, P.; North, A.; Iegre, J.; Georgiou, K.H.; Lubin, A.; Carro, L.; Green, W.; Sore, H.F.; Hyvönen, M.; Spring, D.R. Novel non-ATP competitive small molecules targeting the CK2 α/β interface. Bioorg Med. Chem. 2018, 26, 3016–3020. [Google Scholar] [CrossRef] [PubMed]
- Iegre, J.; Brear, P.; De Fusco, C.; Yoshida, M.; Mitchell, S.L.; Rossmann, M.; Carro, L.; Sore, H.F.; Hyvönen, M.; Spring, D.R. Second-generation CK2α inhibitors targeting the αD pocket. Chem. Sci. 2018, 9, 3041–3049. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.-H.; Zhang, X.; Attarian, D.; Kraus, V.B. Synergistic roles of CBX4 chromo and SIM domains in regulating senescence of primary human osteoarthritic chondrocytes. Arthritis Res. Ther. 2023, 25, 197. [Google Scholar] [CrossRef] [PubMed]
- Oshima, T.; Niwa, Y.; Kuwata, K.; Srivastava, A.; Hyoda, T.; Tsuchiya, Y.; Kumagai, M.; Tsuyuguchi, M.; Tamaru, T.; Sugiyama, A.; et al. Cell-based screen identifies a new potent and highly selective CK2 inhibitor for modulation of circadian rhythms and cancer cell growth. Sci. Adv. 2019, 5, eaau9060. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Wang, Y.; Wang, J.; Zhou, Z.; Cao, S.; Zhang, J. Strategies of Targeting CK2 in Drug Discovery: Challenges, Opportunities, and Emerging Prospects. J. Med. Chem. 2023, 66, 2257–2281. [Google Scholar] [CrossRef] [PubMed]
- Ferguson, A.D.; Sheth, P.R.; Basso, A.D.; Paliwal, S.; Gray, K.; Fischmann, T.O.; Le, H.V. Structural basis of CX-4945 binding to human protein kinase CK2. FEBS Lett. 2011, 585, 104–110. [Google Scholar] [CrossRef] [PubMed]
- Siddiqui-Jain, A.; Drygin, D.; Streiner, N.; Chua, P.; Pierre, F.; O’Brien, S.E.; Bliesath, J.; Omori, M.; Huser, N.; Ho, C.; et al. CX-4945, an orally bioavailable selective inhibitor of protein kinase CK2, inhibits prosurvival and angiogenic signaling and exhibits antitumor efficacy. Cancer Res. 2010, 70, 10288–10298. [Google Scholar] [CrossRef] [PubMed]
- Atkinson, E.L.; Iegre, J.; Brear, P.; A Zhabina, E.; Hyvönen, M.; Spring, D. Downfalls of Chemical Probes Acting at the Kinase ATP-Site: CK2 as a Case Study. Molecules 2021, 26, 1977. [Google Scholar] [CrossRef]
- Schneider, C.C.; Hessenauer, A.; Götz, C.; Montenarh, M. DMAT, an inhibitor of protein kinase CK2 induces reactive oxygen species and DNA double strand breaks. Oncol. Rep. 2009, 21, 1593–1597. [Google Scholar]
- Pagano, M.A.; Meggio, F.; Ruzzene, M.; Andrzejewska, M.; Kazimierczuk, Z.; Pinna, L.A. 2-Dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole: A novel powerful and selective inhibitor of protein kinase CK2. Biochem. Biophys. Res. Commun. 2004, 321, 1040–1044. [Google Scholar] [CrossRef] [PubMed]
- Yim, H.; Lee, Y.H.; Lee, C.H.; Lee, S.K. Emodin, an Anthraquinone Derivative Isolated from the Rhizomes of Rheum palmatum, Selectively Inhibits the Activity of Casein Kinase II as a Competitive Inhibitor. Planta Medica 1999, 65, 9–13. [Google Scholar] [CrossRef] [PubMed]
- Feng, Y.; Huang, S.L.; Dou, W.; Zhang, S.; Chen, J.H.; Shen, Y.; Shen, J.H.; Leng, Y. Emodin, a natural product, selectively inhibits 11beta-hydroxysteroid dehydrogenase type 1 and ameliorates metabolic disorder in diet-induced obese mice. Br. J. Pharmacol. 2010, 161, 113–126. [Google Scholar] [CrossRef]
- Sarno, S.; Moro, S.; Meggio, F.; Zagotto, G.; Ben, D.D.; Ghisellini, P.; Battistutta, R.; Zanotti, G.; Pinna, L.A. Toward the rational design of protein kinase casein kinase-2 inhibitors. Pharmacol. Ther. 2002, 93, 159–168. [Google Scholar] [CrossRef]
- Cozza, G.; Mazzorana, M.; Papinutto, E.; Bain, J.; Elliott, M.; di Maira, G.; Gianoncelli, A.; Pagano, M.A.; Sarno, S.; Ruzzene, M.; et al. Quinalizarin as a potent, selective and cell-permeable inhibitor of protein kinase CK2. Biochem. J. 2009, 421, 387–395. [Google Scholar] [CrossRef]
- Franchin, C.; Cesaro, L.; Salvi, M.; Millioni, R.; Iori, E.; Cifani, P.; James, P.; Arrigoni, G.; Pinna, L. Quantitative analysis of a phosphoproteome readily altered by the protein kinase CK2 inhibitor quinalizarin in HEK-293T cells. Biochim. Biophys. Acta (BBA)—Proteins Proteom. 2014, 1854, 609–623. [Google Scholar] [CrossRef] [PubMed]
- Bullock, A.N.; Debreczeni, J.; Fedorov, O.Y.; Nelson, A.; Marsden, B.D.; Knapp, S. Structural Basis of Inhibitor Specificity of the Human Protooncogene Proviral Insertion Site in Moloney Murine Leukemia Virus (PIM-1) Kinase. J. Med. Chem. 2005, 48, 7604–7614. [Google Scholar] [CrossRef]
- Mathison, C.J.N.; Chianelli, D.; Rucker, P.V.; Nelson, J.; Roland, J.; Huang, Z.; Yang, Y.; Jiang, J.; Xie, Y.F.; Epple, R.; et al. Efficacy and Tolerability of Pyrazolo [1,5-a]pyrimidine RET Kinase Inhibitors for the Treatment of Lung Adenocarcinoma. ACS Med. Chem. Lett. 2020, 11, 558–565. [Google Scholar] [CrossRef]
- Sacks, D.; Baxter, B.; Campbell, B.C.; Carpenter, J.S.; Cognard, C.; Dippel, D.; Eesa, M.; Fischer, U.; Hausegger, K.; Hirsch, J.A. Multisociety Consensus Quality Improvement Revised Consensus Statement for Endovascular Therapy of Acute Ischemic Stroke. Int. J. Stroke 2018, 13, 612–632. [Google Scholar] [CrossRef]
- Krämer, A.; Kurz, C.G.; Berger, B.-T.; Celik, I.E.; Tjaden, A.; Greco, F.A.; Knapp, S.; Hanke, T. Optimization of pyrazolo[1,5-a]pyrimidines lead to the identification of a highly selective casein kinase 2 inhibitor. Eur. J. Med. Chem. 2020, 208, 112770. [Google Scholar] [CrossRef]
- Licciardello, M.P.; Workman, P. A New Chemical Probe Challenges the Broad Cancer Essentiality of CK2. Trends Pharmacol. Sci. 2021, 42, 313–315. [Google Scholar] [CrossRef]
- Wells, C.I.; Drewry, D.H.; Pickett, J.E.; Tjaden, A.; Krämer, A.; Müller, S.; Gyenis, L.; Menyhart, D.; Litchfield, D.W.; Knapp, S.; et al. Development of a potent and selective chemical probe for the pleiotropic kinase CK2. Cell Chem. Biol. 2021, 28, 546–558.e10. [Google Scholar] [CrossRef]
- Yao, K.; Youn, H.; Gao, X.; Huang, B.; Zhou, F.; Li, B.; Han, H. Casein kinase 2 inhibition attenuates androgen receptor function and cell proliferation in prostate cancer cells. Prostate 2012, 72, 1423–1430. [Google Scholar] [CrossRef] [PubMed]
- Pagano, M.A.; Poletto, G.; Di Maira, G.; Cozza, G.; Ruzzene, M.; Sarno, S.; Bain, J.; Elliott, M.; Moro, S.; Zagotto, G.; et al. Tetrabromocinnamic Acid (TBCA) and Related Compounds Represent a New Class of Specific Protein Kinase CK2 Inhibitors. ChemBioChem 2007, 8, 129–139. [Google Scholar] [CrossRef] [PubMed]
- Golub, A.G.; Bdzhola, V.G.; Kyshenia, Y.V.; Sapelkin, V.M.; Prykhod’ko, A.O.; Kukharenko, O.P.; Ostrynska, O.V.; Yarmoluk, S.M. Structure-based discovery of novel flavonol inhibitors of human protein kinase CK2. Mol. Cell. Biochem. 2011, 356, 107–115. [Google Scholar] [CrossRef]
- Guerra, B.; Issinger, O.-G. Protein Kinase CK2 in Human Diseases. Curr. Med. Chem. 2008, 15, 1870–1886. [Google Scholar] [CrossRef] [PubMed]
- Meggio, F.; Pagano, M.A.; Moro, S.; Zagotto, G.; Ruzzene, M.; Sarno, S.; Cozza, G.; Bain, J.; Elliott, M.; Deana, A.D.; et al. Inhibition of Protein Kinase CK2 by Condensed Polyphenolic Derivatives. An in Vitro and in Vivo Study. Biochemistry 2004, 43, 12931–12936. [Google Scholar] [CrossRef] [PubMed]
- Brear, P.; De Fusco, C.; Georgiou, K.H.; Francis-Newton, N.J.; Stubbs, C.J.; Sore, H.F.; Venkitaraman, A.R.; Abell, C.; Spring, D.R.; Hyvönen, M. Specific inhibition of CK2α from an anchor outside the active site. Chem. Sci. 2016, 7, 6839–6845. [Google Scholar] [CrossRef]
- Wang, X.; Wang, Y.; Gou, S. A platinum(II) complex HY1-Pt overcomes cisplatin-induced resistance and attenuates metastasis of epithelial ovarian cancer by cancer cell stemness inhibition. Int. J. Biochem. Cell Biol. 2023, 157, 106395. [Google Scholar] [CrossRef]
- Zhang, J.; Tang, P.; Zou, L.; Zhang, J.; Chen, J.; Yang, C.; He, G.; Liu, B.; Liu, J.; Chiang, C.M.; et al. Discovery of Novel Dual-Target Inhibitor of Bromodomain-Containing Protein 4/Casein Kinase 2 Inducing Apoptosis and Autophagy-Associated Cell Death for Triple-Negative Breast Cancer Therapy. J. Med. Chem. 2021, 64, 18025–18053. [Google Scholar] [CrossRef]
- Calder, J.; Nagelberg, A.; Luu, J.; Lu, D.; Lockwood, W.W. Resistance to BET inhibitors in lung adenocarcinoma is mediated by casein kinase phosphorylation of BRD4. Oncogenesis 2021, 10, 27. [Google Scholar] [CrossRef] [PubMed]
- Channavajhala, P.; Seldin, D.C. Functional interaction of protein kinase CK2 and c-Myc in lymphomagenesis. Oncogene 2002, 21, 5280–5288. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Kim, J.; Roh, M.; O E Franco, O.; Hayward, S.W.; Wills, M.L.; A Abdulkadir, S. Pim1 kinase synergizes with c-MYC to induce advanced prostate carcinoma. Oncogene 2010, 29, 2477–2487. [Google Scholar] [CrossRef]
- Wińska, P.; Wielechowska, M.; Koronkiewicz, M.; Borowiecki, P. Synthesis and Anticancer Activity of Novel Dual Inhibitors of Human Protein Kinases CK2 and PIM-1. Pharmaceutics 2023, 15, 1991. [Google Scholar] [CrossRef] [PubMed]
- Prudent, R.; Moucadel, V.; Sautel, C.F.; Barette, C.; Lafanechère, L.; Mouawad, L.; Grierson, D.; Schmidt, F.; Filippakopoulos, P.; Bullock, A.N.; et al. New potent dual inhibitors of CK2 and Pim kinases: Discovery and structural insights. FASEB J. 2010, 24, 3171–3185. [Google Scholar]
- Sarno, S.; Mazzorana, M.; Traynor, R.; Ruzzene, M.; Cozza, G.; Pagano, M.A.; Meggio, F.; Zagotto, G.; Battistutta, R.; Pinna, L.A. Structural features underlying the selectivity of the kinase inhibitors NBC and dNBC: Role of a nitro group that discriminates between CK2 and DYRK1A. Cell. Mol. Life Sci. 2011, 69, 449–460. [Google Scholar] [CrossRef] [PubMed]
- Łukowska-Chojnacka, E.; Wińska, P.; Wielechowska, M.; Poprzeczko, M.; Bretner, M. Synthesis of novel polybrominated benzimidazole derivatives-potential CK2 inhibitors with anticancer and proapoptotic activity. Bioorg Med. Chem. 2016, 24, 735–741. [Google Scholar] [CrossRef]
- Morooka, S.; Hoshina, M.; Kii, I.; Okabe, T.; Kojima, H.; Inoue, N.; Okuno, Y.; Denawa, M.; Yoshida, S.; Fukuhara, J.; et al. Identification of a Dual Inhibitor of SRPK1 and CK2 That Attenuates Pathological Angiogenesis of Macular Degeneration in Mice. Mol. Pharmacol. 2015, 88, 316–325. [Google Scholar] [CrossRef]
- Sato, K.; Padgaonkar, A.A.; Baker, S.J.; Cosenza, S.C.; Rechkoblit, O.; Subbaiah, D.R.C.V.; Domingo-Domenech, J.; Bartkowski, A.; Port, E.R.; Aggarwal, A.K.; et al. Simultaneous CK2/TNIK/DYRK1 inhibition by 108600 suppresses triple negative breast cancer stem cells and chemotherapy-resistant disease. Nat. Commun. 2021, 12, 4671. [Google Scholar] [CrossRef]
- Perera, Y.; Ramos, Y.; Padrón, G.; Caballero, E.; Guirola, O.; Caligiuri, L.G.; Lorenzo, N.; Gottardo, F.; Farina, H.G.; Filhol, O.; et al. CIGB-300 anticancer peptide regulates the protein kinase CK2-dependent phosphoproteome. Mol. Cell. Biochem. 2020, 470, 63–75. [Google Scholar] [CrossRef]
- Farina, H.G.; Acero, F.B.; Perera, Y.; Rodríguez, A.; Perea, S.E.; Castro, B.A.; Gomez, R.; Alonso, D.F.; Gomez, D.E. CIGB-300, a proapoptotic peptide, inhibits angiogenesis in vitro and in vivo. Exp. Cell Res. 2011, 317, 1677–1688. [Google Scholar] [CrossRef]
- Gottardo, M.F.; Capobianco, C.S.; Sidabra, J.E.; Garona, J.; Perera, Y.; Perea, S.E.; Alonso, D.F.; Farina, H.G. Preclinical efficacy of CIGB-300, an anti-CK2 peptide, on breast cancer metastasic colonization. Sci. Rep. 2020, 10, 14689. [Google Scholar] [CrossRef]
- Zien, P.; Duncan, J.S.; Skierski, J.; Bretner, M.; Litchfield, D.W.; Shugar, D. Tetrabromobenzotriazole (TBBt) and tetrabromobenzimidazole (TBBz) as selective inhibitors of protein kinase CK2: Evaluation of their effects on cells and different molecular forms of human CK2. Biochim. Biophys. Acta (BBA)—Proteins Proteom. 2005, 1754, 271–280. [Google Scholar] [CrossRef]
- Koo, J.-H.; Yu, H.C.; Nam, S.; Kim, D.-C.; Lee, J.H. Casein Kinase 2 Alpha Inhibition Protects against Sepsis-Induced Acute Kidney Injury. Int. J. Mol. Sci. 2023, 24, 9783. [Google Scholar] [CrossRef]
- Nitta, R.T.; Gholamin, S.; Feroze, A.H.; Agarwal, M.; Cheshier, S.H.; Mitra, S.S.; Li, G. Casein kinase 2α regulates glioblastoma brain tumor-initiating cell growth through the β-catenin pathway. Oncogene 2014, 34, 3688–3699. [Google Scholar] [CrossRef]
- Wang, T.; Yang, N.; Liang, C.; Xu, H.; An, Y.; Xiao, S.; Zheng, M.; Liu, L.; Wang, G.; Nie, L. Detecting Protein-Protein Interaction Based on Protein Fragment Complementation Assay. Curr. Protein Pept. Sci. 2020, 21, 598–610. [Google Scholar] [CrossRef] [PubMed]
- Dai, B.; Bailey-Kellogg, C. Protein interaction interface region prediction by geometric deep learning. Bioinformatics 2021, 37, 2580–2588. [Google Scholar] [CrossRef] [PubMed]
- Apostolopoulos, V.; Bojarska, J.; Chai, T.-T.; Elnagdy, S.; Kaczmarek, K.; Matsoukas, J.; New, R.; Parang, K.; Lopez, O.P.; Parhiz, H.; et al. A Global Review on Short Peptides: Frontiers and Perspectives. Molecules 2021, 26, 430. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Wang, N.; Zhang, W.; Cheng, X.; Yan, Z.; Shao, G.; Wang, X.; Wang, R.; Fu, C. Therapeutic peptides: Current applications and future directions. Signal Transduct. Target. Ther. 2022, 7, 48. [Google Scholar] [CrossRef] [PubMed]
- Berillo, D.; Yeskendir, A.; Zharkinbekov, Z.; Raziyeva, K.; Saparov, A. Peptide-Based Drug Delivery Systems. Medicina 2021, 57, 1209. [Google Scholar] [CrossRef] [PubMed]
- Hao, M.; Zhang, L.; Chen, P. Membrane Internalization Mechanisms and Design Strategies of Arginine-Rich Cell-Penetrating Peptides. Int. J. Mol. Sci. 2022, 23, 9038. [Google Scholar] [CrossRef]
- De Oliveira, E.C.L.; da Costa, K.S.; Taube, P.S.; Lima, A.H.; Junior, C.D.S.D.S. Biological Membrane-Penetrating Peptides: Computational Prediction and Applications. Front. Cell Infect. Microbiol. 2022, 12, 838259. [Google Scholar] [CrossRef]
- Lai, Y.; Fois, G.; Flores, J.R.; Tuvim, M.J.; Zhou, Q.; Yang, K.; Leitz, J.; Peters, J.; Zhang, Y.; Pfuetzner, R.A.; et al. Inhibition of calcium-triggered secretion by hydrocarbon-stapled peptides. Nature 2022, 603, 949–956. [Google Scholar] [CrossRef]
- Dong, C.Z.; Bruzzoni-Giovanelli, H.; Yu, Y.; Dorgham, K.; Parizot, C.; Zini, J.M.; Brossas, J.Y.; Tuffery, P.; Rebollo, A. Identification of peptides interfering with the LRRK2/PP1 interaction. PLoS ONE 2020, 15, e0237110. [Google Scholar] [CrossRef]
- Dergunova, L.V.; Filippenkov, I.B.; Limborska, S.A.; Myasoedov, N.F. Neuroprotective Peptides and New Strategies for Ischemic Stroke Drug Discoveries. Genes 2023, 14, 953. [Google Scholar] [CrossRef]
- Simón-Gracia, L.; Loisel, S.; Sidorenko, V.; Scodeller, P.; Parizot, C.; Savier, E.; Haute, T.; Teesalu, T.; Rebollo, A. Preclinical Validation of Tumor-Penetrating and Interfering Peptides against Chronic Lymphocytic Leukemia. Mol. Pharm. 2022, 19, 895–903. [Google Scholar] [CrossRef]
- Wang, J.; Wu, Y.; Chen, Z.; Chen, Y.; Lin, Q.; Liang, Y. Exogenous Bioactive Peptides Have a Potential Therapeutic Role in Delaying Aging in Rodent Models. Int. J. Mol. Sci. 2022, 23, 1421. [Google Scholar] [CrossRef]
- Bruzzoni-Giovanelli, H.; Alezra, V.; Wolff, N.; Dong, C.Z.; Tuffery, P.; Rebollo, A. Interfering peptides targeting protein-protein interactions: The next generation of drugs? Drug Discov. Today 2018, 23, 272–285. [Google Scholar] [CrossRef] [PubMed]
- Lisberg, A.; Ellis, R.; Nicholson, K.; Moku, P.; Swarup, A.; Dhurjati, P.; Nohe, A. Mathematical modeling of the effects of CK2.3 on mineralization in osteoporotic bone. CPT Pharmacomet. Syst. Pharmacol. 2017, 6, 208–215. [Google Scholar] [CrossRef] [PubMed]
- Bragdon, B.; Thinakaran, S.; Moseychuk, O.; Gurski, L.; Bonor, J.; Price, C.; Wang, L.; Beamer, W.G.; Nohe, A. Casein kinase 2 regulates in vivo bone formation through its interaction with bone morphogenetic protein receptor type Ia. Bone 2011, 49, 944–954. [Google Scholar] [CrossRef] [PubMed]
- Akkiraju, H.; Bonor, J.; Nohe, A. CK2.1, a novel peptide, induces articular cartilage formation in vivo. J. Orthop. Res. 2016, 35, 876–885. [Google Scholar] [CrossRef]
- Akkiraju, H.; Srinivasan, P.P.; Xu, X.; Jia, X.; Safran, C.B.K.; Nohe, A. CK2.1, a bone morphogenetic protein receptor type Ia mimetic peptide, repairs cartilage in mice with destabilized medial meniscus. Stem Cell Res. Ther. 2017, 8, 82. [Google Scholar] [CrossRef]
- Takahashi, K.; Setoguchi, T.; Tsuru, A.; Saitoh, Y.; Nagano, S.; Ishidou, Y.; Maeda, S.; Furukawa, T.; Komiya, S. Inhibition of casein kinase 2 prevents growth of human osteosarcoma. Oncol. Rep. 2016, 37, 1141–1147. [Google Scholar] [CrossRef]
- Halloran, D.; Pandit, V.; MacMurray, C.; Stone, V.; DeGeorge, K.; Eskander, M.; Root, D.; McTague, S.; Pelkey, H.; Nohe, A. Age-Related Low Bone Mineral Density in C57BL/6 Mice Is Reflective of Aberrant Bone Morphogenetic Protein-2 Signaling Observed in Human Patients Diagnosed with Osteoporosis. Int. J. Mol. Sci. 2022, 23, 11205. [Google Scholar] [CrossRef] [PubMed]
- Vrathasha, V.; Weidner, H.; Nohe, A. Mechanism of CK2.3, a Novel Mimetic Peptide of Bone Morphogenetic Protein Receptor Type IA, Mediated Osteogenesis. Int. J. Mol. Sci. 2019, 10, 2500. [Google Scholar] [CrossRef] [PubMed]
- Weidner, H.; Gao, V.Y.; Dibert, D.; McTague, S.; Eskander, M.; Duncan, R.; Wang, L.; Nohe, A. CK2.3, a Mimetic Peptide of the BMP Type I Receptor, Increases Activity in Osteoblasts over BMP2. Int. J. Mol. Sci. 2019, 20, 5877. [Google Scholar] [CrossRef] [PubMed]
Name | CK2 Selectivity | Efficacy | Reference |
---|---|---|---|
Silmitasertib (CX-4945) | Selective at low concentrations. However, at nanomolar concentrations, it inhibits other kinases, such as DYRK1A. | Currently, it is the most selective CK2 inhibitor in clinical use and has minimal toxicity. Promotes apoptosis while inhibiting the PI3K/Akt signaling pathway and the cell cycle progression. Inhibits angiogenesis. | [108,117,118,119] |
dimethylamino-4,5,6,7-1H-tetrabromobenzimidazole (DMAT) | Selective for CK2 but inhibits DYRK1A | Induces DNA double-strand breaks. Promotes apoptosis. Inhibition of aldosterone, DHEAS, and androstenedione secretion in adenocarcinoma cell line H295R. | [27,120,121] |
Emodin | Moderately selective | Natural anthraquinone derivative extracted from rhubarb. Inhibits CK2. | [122,123,124] |
Quinalizarin | Fairly selective | Promotes apoptosis in HEK-293 and Jurkat cells. | [125,126] |
IC20 | Extremely selective for CK2α | Not cytotoxic in cancer cells | [127,128,129,130] |
SGC-CK2-1 | Selective | Used as a cellular probe to investigate the intracellular function of CK2. Low inhibitory effect on the proliferation of most cancer cells, effective against a small subset of cancer cells. | [131,132] |
Tetrabromocinnamic Acid (TBCA) | Selective | Derived from 4,5,6,7-tetrabromobenzotriazole (TBB). It is more potent than TBB. Does not inhibit DYRK1 like TBB. Promotes apoptosis in Jurkat cancer cells. Suppresses platelet aggregation/secretion and the cell cycle progression in prostate cancer cells. | [133,134] |
4-(6,8-Dibromo-3-hydroxy-4-oxo-4H-chromo-2-yl)-benzoic acid (FLC26) | Mildly selective | Causes a significant increase in apoptosis in PANC-1 cells | [135,136] |
3,8-dibromo-7-hydroxy-4-methylchromen-2-one (DBC) | Poorly selective | Induces apoptosis in Jurkat cells. | [137] |
CAM4066 | Poorly selective | Acts on the αD region and ATP-binding sites of CK2. The moiety bound to the ATP binding site forms a hydrogen bond with Lys68 and two water molecules. The moiety in the αD site interacts with Pro159 and a conserved water molecule. The linker creates a network of hydrogen bonds. Poor permeability. Methyl ester derivative pro-CAM4066 has better permeability. | [73,113,138] |
CAM4712 | Highly selective | Anti-proliferative | [109,113] |
GO289 | High selectivity | Extremely selective, ideal for clinical use. Inhibition of the phosphorylation sites of multiple clock proteins and suppressed the growth/proliferation of cells of a diverse array of cancers. CK2α and CK2α′ are the primary targets of the drug. | [115,119] |
HY1-Pt | Highly selective. Derived from Silmitasertib. Overcomes cisplatin-induced resistance. | Reversed cisplatin-induced drug resistance. Suppresses DNA damage repair in cancer cells. It also inhibited the Wnt/beta-catenin signaling pathway while activating the mitochondrial apoptosis pathway. It displayed no toxicity to healthy hepatocytes and could be used as a therapeutic for NSCLC. | [107,139] |
Name | Co-Target(s) | Clinical Efficacy | Reference |
---|---|---|---|
Compound 58 | BRD4 | Able to overcome drug resistance in cancer treatment. | [114,140] |
Compound 60 | BRD4 | Highly selective. Reduces tumor growth and lessens cancer symptoms in vivo and in-vitro, with no apparent side effects. It is considered a potential therapeutic in triple-negative breast cancer. | [114,140] |
Naphtho[2,1-b:7,6-b′]difuran-2,8-dicarboxylic acid hydrate (CPA), CPB, AMR | PIM | Lack of cell permeability; hence, it cannot be used clinically. | [145] |
8-hydroxy-4-methyl-9-nitrobenzol(g)chrome-2-one (NBC) | PIM | Induces apoptosis. | [137,146] |
1-β-D-2′-deoxyribofuranosyl-4,5,6,7-tetrabromo-1H-benzimidazole (TDB) | PIM | Extremely high selectivity indicates it has clinical potential. | [147] |
Compound 66 | PIM | Cytotoxic against cancer cells but not healthy cells. Inhibits the proliferation of various cancer cell lines. Reduces the viability of cancer cells more effectively than Silmitasertib. It is membrane-permeable. | [146] |
6-(4-Hydroxy-3-methoxybenzylidene)-5-imino-2-(trifluoromethyl)-5H-(1,3,4)thiadiazolo[3,2-a]pyrimidin-7(6H)-one (SRPIN803; CK2 inhibitor XIII). | SRPK1 | Inhibits aberrant angiogenesis. Significantly inhibits cell viability in Jurkat cell lines. Prevents the formation of intraocular neovascularization in vivo. | [148] |
Compound 108600 | TNIK, DYRK1 | The inhibitory effect on CK2α’ is ten times stronger than on CK2α. Inhibits tumor growth in breast cancer cells and overcomes chemical resistance. In vitro and in vivo studies suggest it is an optimal inhibitor in clinical settings. | [149] |
Name | Status for Clinical Application | Reference |
---|---|---|
CIGB-300 | Cell-permeable. Inhibits angiogenesis and metastasis. Used in early clinical trials in combination with chemoradiotherapy as a therapeutic against cervical cancer. Administered by injection into the tumor. Targets the phosphoacceptor domain. Releases histamine from the cells, possibly due to higher intracellular calcium levels in the cell. | [42] |
4,5,6,7-tetrabromobenzotriazole (TBBt) | Moderately effective as an anti-cancer drug. Induces apoptosis in tumor cells. Inhibits CK2α subunit. They are used in Sepsis-Induced Acute Kidney Injury. | [153,154] |
4,5,6,7-tetrabromobenzimidazole (TBBz) | Able to target specific molecular forms of CK2. It is more effective in inducing apoptosis and necrosis in tumor cells than TBBt. Inhibits CK2α subunit activity. It was tested in glioblastoma cell lines. | [153,155] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Pandit, V.; DeGeorge, K.; Nohe, A. Scoping Pleiotropy of CK2 in Musculoskeletal Disorders for a Novel Targeting Approach. Kinases Phosphatases 2024, 2, 43-66. https://doi.org/10.3390/kinasesphosphatases2010004
Pandit V, DeGeorge K, Nohe A. Scoping Pleiotropy of CK2 in Musculoskeletal Disorders for a Novel Targeting Approach. Kinases and Phosphatases. 2024; 2(1):43-66. https://doi.org/10.3390/kinasesphosphatases2010004
Chicago/Turabian StylePandit, Venu, Kailey DeGeorge, and Anja Nohe. 2024. "Scoping Pleiotropy of CK2 in Musculoskeletal Disorders for a Novel Targeting Approach" Kinases and Phosphatases 2, no. 1: 43-66. https://doi.org/10.3390/kinasesphosphatases2010004