Covalent Cysteine Targeting of Bruton’s Tyrosine Kinase (BTK) Family by Withaferin-A Reduces Survival of Glucocorticoid-Resistant Multiple Myeloma MM1 Cells
Abstract
:Simple Summary
Abstract
1. Introduction
2. Results
2.1. GC Therapy Resistance in Multiple Myeloma Is Associated with Hyperactivation of Tyrosine Kinases
2.2. The Tyrosine Kinase Inhibitor Profile of Withaferin A and Ibrutinib Show a High Degree of Similarity
2.3. WA Inhibits BCR-BTK Kinase Activity by Transcriptional Downregulation and Covalent Cysteine-Dependent Targeting of BTK
2.4. Covalent C481 Targeting of BTK by WA in Hinge-6 Domain of the Protein Kinase Cysteinome Classification Reduces Survival of Glucocorticoid Resistant Multiple Myeloma MM1 Cells
3. Discussion
4. Materials and Methods
4.1. Cell Culture and Cell Viability Assays
4.2. Cell Lysis and Peptide Array-Based TK Activity Profiling
4.3. cDNA Conversion Quantitative Real-Time PCR
4.4. RNA Extraction and RNA Sequencing
4.5. Antibodies and Reagents
4.6. Cell Viability after WA Washout
4.7. Protein Extraction and Western Immunoblot Analysis
4.8. WA-Biotin-Based Affinity Purification
4.9. Immunofluorescence Confocal Microscopy
4.10. Covalent Docking of WA with BTK
4.11. Cell Transfections and BRET Measurements
4.12. Nucleofection of siBTK and C481S BTK
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
- Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, E359–E386. [Google Scholar] [CrossRef]
- Hussein, M.A. Multiple myeloma: Most common end-organ damage and management. J. Natl. Compr. Cancer Netw. 2007, 5, 170–178. [Google Scholar] [CrossRef]
- Robak, P.; Drozdz, I.; Szemraj, J.; Robak, T. Drug resistance in multiple myeloma. Cancer Treat. Rev. 2018, 70, 199–208. [Google Scholar] [CrossRef] [PubMed]
- Rajkumar, S.V. Myeloma today: Disease definitions and treatment advances. Am. J. Hematol. 2016, 91, 90–100. [Google Scholar] [CrossRef] [Green Version]
- Yang, W.C.; Lin, S.F. Mechanisms of Drug Resistance in Relapse and Refractory Multiple Myeloma. Biomed. Res. Int. 2015, 2015, 341430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ito, S. Proteasome Inhibitors for the Treatment of Multiple Myeloma. Cancers 2020, 12, 265. [Google Scholar] [CrossRef] [Green Version]
- Abe, Y.; Ishida, T. Immunomodulatory drugs in the treatment of multiple myeloma. Jpn. J. Clin. Oncol. 2019, 49, 695–702. [Google Scholar] [CrossRef]
- Burwick, N.; Sharma, S. Glucocorticoids in multiple myeloma: Past, present, and future. Ann. Hematol. 2019, 98, 19–28. [Google Scholar] [CrossRef]
- Thanendrarajan, S.; Davies, F.E.; Morgan, G.J.; Schinke, C.; Mathur, P.; Heuck, C.J.; Zangari, M.; Epstein, J.; Yaccoby, S.; Weinhold, N.; et al. Monoclonal antibody therapy in multiple myeloma: Where do we stand and where are we going? Immunotherapy 2016, 8, 367–384. [Google Scholar] [CrossRef]
- Imai, Y.; Hirano, M.; Kobayashi, M.; Futami, M.; Tojo, A. HDAC Inhibitors Exert Anti-Myeloma Effects through Multiple Modes of Action. Cancers 2019, 11, 475. [Google Scholar] [CrossRef] [Green Version]
- Kumar, S.K.; Therneau, T.M.; Gertz, M.A.; Lacy, M.Q.; Dispenzieri, A.; Rajkumar, S.V.; Fonseca, R.; Witzig, T.E.; Lust, J.A.; Larson, D.R.; et al. Clinical course of patients with relapsed multiple myeloma. Mayo Clin. Proc. 2004, 79, 867–874. [Google Scholar] [CrossRef] [PubMed]
- Issa, M.E.; Takhsha, F.S.; Chirumamilla, C.S.; Perez-Novo, C.; Vanden Berghe, W.; Cuendet, M. Epigenetic strategies to reverse drug resistance in heterogeneous multiple myeloma. Clin. Epigenet. 2017, 9, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shain, K.H.; Tao, J. The B-cell receptor orchestrates environment-mediated lymphoma survival and drug resistance in B-cell malignancies. Oncogene 2014, 33, 4107–4113. [Google Scholar] [CrossRef] [Green Version]
- Woyach, J.A.; Johnson, A.J.; Byrd, J.C. The B-cell receptor signaling pathway as a therapeutic target in CLL. Blood 2012, 120, 1175–1184. [Google Scholar] [CrossRef] [Green Version]
- Takata, M.; Sabe, H.; Hata, A.; Inazu, T.; Homma, Y.; Nukada, T.; Yamamura, H.; Kurosaki, T. Tyrosine kinases Lyn and Syk regulate B cell receptor-coupled Ca2+ mobilization through distinct pathways. EMBO J. 1994, 13, 1341–1349. [Google Scholar] [CrossRef]
- Tai, Y.T.; Chang, B.Y.; Kong, S.Y.; Fulciniti, M.; Yang, G.; Calle, Y.; Hu, Y.; Lin, J.; Zhao, J.J.; Cagnetta, A.; et al. Bruton tyrosine kinase inhibition is a novel therapeutic strategy targeting tumor in the bone marrow microenvironment in multiple myeloma. Blood 2012, 120, 1877–1887. [Google Scholar] [CrossRef]
- Yang, Y.; Shi, J.; Gu, Z.; Salama, M.E.; Das, S.; Wendlandt, E.; Xu, H.; Huang, J.; Tao, Y.; Hao, M.; et al. Bruton tyrosine kinase is a therapeutic target in stem-like cells from multiple myeloma. Cancer Res. 2015, 75, 594–604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, W.; Wei, R.; Liu, S.; Qiao, L.; Hou, J.; Gu, C.; Yang, Y. BTK induces CAM-DR through regulation of CXCR4 degradation in multiple myeloma. Am. J. Transl. Res. 2019, 11, 4139–4150. [Google Scholar] [PubMed]
- Murray, M.Y.; Zaitseva, L.; Auger, M.J.; Craig, J.I.; MacEwan, D.J.; Rushworth, S.A.; Bowles, K.M. Ibrutinib inhibits BTK-driven NF-kappaB p65 activity to overcome bortezomib-resistance in multiple myeloma. Cell Cycle 2015, 14, 2367–2375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Young, R.M.; Staudt, L.M. Targeting pathological B cell receptor signalling in lymphoid malignancies. Nat. Rev. Drug Discov. 2013, 12, 229–243. [Google Scholar] [CrossRef] [PubMed]
- Dasmahapatra, G.; Patel, H.; Dent, P.; Fisher, R.I.; Friedberg, J.; Grant, S. The Bruton tyrosine kinase (BTK) inhibitor PCI-32765 synergistically increases proteasome inhibitor activity in diffuse large-B cell lymphoma (DLBCL) and mantle cell lymphoma (MCL) cells sensitive or resistant to bortezomib. Br. J. Haematol. 2013, 161, 43–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Herman, S.E.; Gordon, A.L.; Hertlein, E.; Ramanunni, A.; Zhang, X.; Jaglowski, S.; Flynn, J.; Jones, J.; Blum, K.A.; Buggy, J.J.; et al. Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765. Blood 2011, 117, 6287–6296. [Google Scholar] [CrossRef]
- Herman, S.E.; Sun, X.; McAuley, E.M.; Hsieh, M.M.; Pittaluga, S.; Raffeld, M.; Liu, D.; Keyvanfar, K.; Chapman, C.M.; Chen, J.; et al. Modeling tumor-host interactions of chronic lymphocytic leukemia in xenografted mice to study tumor biology and evaluate targeted therapy. Leukemia 2013, 27, 2311–2321. [Google Scholar] [CrossRef] [Green Version]
- Yang, Y.; Shaffer, A.L., 3rd; Emre, N.C.; Ceribelli, M.; Zhang, M.; Wright, G.; Xiao, W.; Powell, J.; Platig, J.; Kohlhammer, H.; et al. Exploiting synthetic lethality for the therapy of ABC diffuse large B cell lymphoma. Cancer Cell 2012, 21, 723–737. [Google Scholar] [CrossRef] [Green Version]
- Franco, L.M.; Gadkari, M.; Howe, K.N.; Sun, J.; Kardava, L.; Kumar, P.; Kumari, S.; Hu, Z.; Fraser, I.D.C.; Moir, S.; et al. Immune regulation by glucocorticoids can be linked to cell type-dependent transcriptional responses. J. Exp. Med. 2019, 216, 384–406. [Google Scholar] [CrossRef] [Green Version]
- Kruth, K.A.; Fang, M.; Shelton, D.N.; Abu-Halawa, O.; Mahling, R.; Yang, H.; Weissman, J.S.; Loh, M.L.; Muschen, M.; Tasian, S.K.; et al. Suppression of B-cell development genes is key to glucocorticoid efficacy in treatment of acute lymphoblastic leukemia. Blood 2017, 129, 3000–3008. [Google Scholar] [CrossRef]
- Shi, Y.; Wang, G.; Muhowski, E.M.; McCaw, L.; Wang, C.; Bjarnason, G.; Woyach, J.A.; Spaner, D.E. Ibrutinib reprograms the glucocorticoid receptor in chronic lymphocytic leukemia cells. Leukemia 2019, 33, 1650–1662. [Google Scholar] [CrossRef] [PubMed]
- Manzoni, D.; Catallo, R.; Chebel, A.; Baseggio, L.; Michallet, A.S.; Roualdes, O.; Magaud, J.P.; Salles, G.; Ffrench, M. The ibrutinib B-cell proliferation inhibition is potentiated in vitro by dexamethasone: Application to chronic lymphocytic leukemia. Leuk Res. 2016, 47, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Chari, A.; Larson, S.; Holkova, B.; Cornell, R.F.; Gasparetto, C.; Karanes, C.; Matous, J.V.; Niesvizky, R.; Valent, J.; Lunning, M.; et al. Phase 1 trial of ibrutinib and carfilzomib combination therapy for relapsed or relapsed and refractory multiple myeloma. Leuk Lymphoma 2018, 59, 2588–2594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Richardson, P.G.; Bensinger, W.I.; Huff, C.A.; Costello, C.L.; Lendvai, N.; Berdeja, J.G.; Anderson, L.D., Jr.; Siegel, D.S.; Lebovic, D.; Jagannath, S.; et al. Ibrutinib alone or with dexamethasone for relapsed or relapsed and refractory multiple myeloma: Phase 2 trial results. Br. J. Haematol. 2018, 180, 821–830. [Google Scholar] [CrossRef]
- Chi, J.; Park, J.; Saif, M.W. Ibrutinib-Induced Vasculitis in a Patient with Metastatic Colon Cancer Treated in Combination with Cetuximab. Case Rep. Oncol. Med. 2020, 2020, 6154213. [Google Scholar] [CrossRef]
- Burger, J.A.; Tedeschi, A.; Barr, P.M.; Robak, T.; Owen, C.; Ghia, P.; Bairey, O.; Hillmen, P.; Bartlett, N.L.; Li, J.; et al. Ibrutinib as Initial Therapy for Patients with Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2015, 373, 2425–2437. [Google Scholar] [CrossRef]
- Guha, A.; Derbala, M.H.; Zhao, Q.; Wiczer, T.E.; Woyach, J.A.; Byrd, J.C.; Awan, F.T.; Addison, D. Ventricular Arrhythmias Following Ibrutinib Initiation for Lymphoid Malignancies. J. Am. Coll. Cardiol. 2018, 72, 697–698. [Google Scholar] [CrossRef] [PubMed]
- Salem, J.E.; Manouchehri, A.; Bretagne, M.; Lebrun-Vignes, B.; Groarke, J.D.; Johnson, D.B.; Yang, T.; Reddy, N.M.; Funck-Brentano, C.; Brown, J.R.; et al. Cardiovascular Toxicities Associated with Ibrutinib. J. Am. Coll. Cardiol. 2019, 74, 1667–1678. [Google Scholar] [CrossRef]
- Barf, T.; Covey, T.; Izumi, R.; van de Kar, B.; Gulrajani, M.; van Lith, B.; van Hoek, M.; de Zwart, E.; Mittag, D.; Demont, D.; et al. Acalabrutinib (ACP-196): A Covalent Bruton Tyrosine Kinase Inhibitor with a Differentiated Selectivity and In Vivo Potency Profile. J. Pharmacol. Exp. Ther. 2017, 363, 240–252. [Google Scholar] [CrossRef]
- George, B.; Chowdhury, S.M.; Hart, A.; Sircar, A.; Singh, S.K.; Nath, U.K.; Mamgain, M.; Singhal, N.K.; Sehgal, L.; Jain, N. Ibrutinib Resistance Mechanisms and Treatment Strategies for B-Cell lymphomas. Cancers 2020, 12, 1328. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.; Liu, C.; Tsui, S.T.; Liu, D. Second-generation inhibitors of Bruton tyrosine kinase. J. Hematol. Oncol. 2016, 9, 80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Woyach, J.; Huang, Y.; Rogers, K.; Bhat, S.A.; Grever, M.R.; Lozanski, A.; Doong, T.-J.; Blachly, J.S.; Lozanski, G.; Jones, D.; et al. Resistance to Acalabrutinib in CLL Is Mediated Primarily By BTK Mutations. Blood 2019, 134, 504. [Google Scholar] [CrossRef]
- Mathews Griner, L.A.; Guha, R.; Shinn, P.; Young, R.M.; Keller, J.M.; Liu, D.; Goldlust, I.S.; Yasgar, A.; McKnight, C.; Boxer, M.B.; et al. High-throughput combinatorial screening identifies drugs that cooperate with ibrutinib to kill activated B-cell-like diffuse large B-cell lymphoma cells. Proc. Natl. Acad. Sci. USA 2014, 111, 2349–2354. [Google Scholar] [CrossRef] [Green Version]
- Lucas, D.M.; Still, P.C.; Perez, L.B.; Grever, M.R.; Kinghorn, A.D. Potential of plant-derived natural products in the treatment of leukemia and lymphoma. Curr. Drug Targets 2010, 11, 812–822. [Google Scholar] [CrossRef]
- Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef]
- Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Supuran, C.T.; International Natural Product Sciences Taskforce. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 1–17. [Google Scholar] [CrossRef] [PubMed]
- Hassannia, B.; Logie, E.; Vandenabeele, P.; Vanden Berghe, T.; Vanden Berghe, W. Withaferin A: From ayurvedic folk medicine to preclinical anti-cancer drug. Biochem. Pharmacol. 2020, 173, 113602. [Google Scholar] [CrossRef] [PubMed]
- Dom, M.; Offner, F.; Vanden Berghe, W.; Van Ostade, X. Proteomic characterization of Withaferin A-targeted protein networks for the treatment of monoclonal myeloma gammopathies. J. Proteom. 2018, 179, 17–29. [Google Scholar] [CrossRef]
- Issa, M.E.; Cuendet, M. Withaferin A induces cell death and differentiation in multiple myeloma cancer stem cells. Medchemcomm 2017, 8, 112–121. [Google Scholar] [CrossRef] [PubMed]
- McKenna, M.K.; Gachuki, B.W.; Alhakeem, S.S.; Oben, K.N.; Rangnekar, V.M.; Gupta, R.C.; Bondada, S. Anti-cancer activity of withaferin A in B-cell lymphoma. Cancer Biol. Ther. 2015, 16, 1088–1098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heyninck, K.; Lahtela-Kakkonen, M.; Van der Veken, P.; Haegeman, G.; Vanden Berghe, W. Withaferin A inhibits NF-kappaB activation by targeting cysteine 179 in IKKbeta. Biochem. Pharmacol. 2014, 91, 501–509. [Google Scholar] [CrossRef] [PubMed]
- Hahm, E.R.; Lee, J.; Singh, S.V. Role of mitogen-activated protein kinases and Mcl-1 in apoptosis induction by withaferin A in human breast cancer cells. Mol. Carcinog. 2014, 53, 907–916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grogan, P.T.; Sleder, K.D.; Samadi, A.K.; Zhang, H.; Timmermann, B.N.; Cohen, M.S. Cytotoxicity of withaferin A in glioblastomas involves induction of an oxidative stress-mediated heat shock response while altering Akt/mTOR and MAPK signaling pathways. Investig. New Drugs 2013, 31, 545–557. [Google Scholar] [CrossRef] [Green Version]
- Mandal, C.; Dutta, A.; Mallick, A.; Chandra, S.; Misra, L.; Sangwan, R.S.; Mandal, C. Withaferin A induces apoptosis by activating p38 mitogen-activated protein kinase signaling cascade in leukemic cells of lymphoid and myeloid origin through mitochondrial death cascade. Apoptosis 2008, 13, 1450–1464. [Google Scholar] [CrossRef]
- Oh, J.H.; Lee, T.J.; Kim, S.H.; Choi, Y.H.; Lee, S.H.; Lee, J.M.; Kim, Y.H.; Park, J.W.; Kwon, T.K. Induction of apoptosis by withaferin A in human leukemia U937 cells through down-regulation of Akt phosphorylation. Apoptosis 2008, 13, 1494–1504. [Google Scholar] [CrossRef] [PubMed]
- Palagani, A.; Op de Beeck, K.; Naulaerts, S.; Diddens, J.; Sekhar Chirumamilla, C.; Van Camp, G.; Laukens, K.; Heyninck, K.; Gerlo, S.; Mestdagh, P.; et al. Ectopic microRNA-150-5p transcription sensitizes glucocorticoid therapy response in MM1S multiple myeloma cells but fails to overcome hormone therapy resistance in MM1R cells. PLoS ONE 2014, 9, e113842. [Google Scholar] [CrossRef]
- Greenstein, S.; Krett, N.L.; Kurosawa, Y.; Ma, C.; Chauhan, D.; Hideshima, T.; Anderson, K.C.; Rosen, S.T. Characterization of the MM.1 human multiple myeloma (MM) cell lines: A model system to elucidate the characteristics, behavior, and signaling of steroid-sensitive and -resistant MM cells. Exp. Hematol. 2003, 31, 271–282. [Google Scholar] [CrossRef]
- Hilhorst, R.; Houkes, L.; van den Berg, A.; Ruijtenbeek, R. Peptide microarrays for detailed, high-throughput substrate identification, kinetic characterization, and inhibition studies on protein kinase A. Anal. Biochem. 2009, 387, 150–161. [Google Scholar] [CrossRef] [PubMed]
- Chirumamilla, C.S.; Fazil, M.; Perez-Novo, C.; Rangarajan, S.; de Wijn, R.; Ramireddy, P.; Verma, N.K.; Vanden Berghe, W. Profiling Activity of Cellular Kinases in Migrating T-Cells. Methods Mol. Biol. 2019, 1930, 99–113. [Google Scholar] [CrossRef] [PubMed]
- Love, M.I.; Huber, W.; Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef] [Green Version]
- Chauhan, D.; Auclair, D.; Robinson, E.K.; Hideshima, T.; Li, G.; Podar, K.; Gupta, D.; Richardson, P.; Schlossman, R.L.; Krett, N.; et al. Identification of genes regulated by dexamethasone in multiple myeloma cells using oligonucleotide arrays. Oncogene 2002, 21, 1346–1358. [Google Scholar] [CrossRef] [Green Version]
- Berglof, A.; Hamasy, A.; Meinke, S.; Palma, M.; Krstic, A.; Mansson, R.; Kimby, E.; Osterborg, A.; Smith, C.I. Targets for Ibrutinib Beyond B Cell Malignancies. Scand. J. Immunol. 2015, 82, 208–217. [Google Scholar] [CrossRef]
- Cheng, S.; Ma, J.; Guo, A.; Lu, P.; Leonard, J.P.; Coleman, M.; Liu, M.; Buggy, J.J.; Furman, R.R.; Wang, Y.L. BTK inhibition targets in vivo CLL proliferation through its effects on B-cell receptor signaling activity. Leukemia 2014, 28, 649–657. [Google Scholar] [CrossRef]
- Patel, V.; Balakrishnan, K.; Bibikova, E.; Ayres, M.; Keating, M.J.; Wierda, W.G.; Gandhi, V. Comparison of Acalabrutinib, A Selective Bruton Tyrosine Kinase Inhibitor, with Ibrutinib in Chronic Lymphocytic Leukemia Cells. Clin. Cancer Res. 2017, 23, 3734–3743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chu, Y.; Lee, S.; Shah, T.; Yin, C.; Barth, M.; Miles, R.R.; Ayello, J.; Morris, E.; Harrison, L.; Van de Ven, C.; et al. Ibrutinib significantly inhibited Bruton’s tyrosine kinase (BTK) phosphorylation,in-vitro proliferation and enhanced overall survival in a preclinical Burkitt lymphoma (BL) model. Oncoimmunology 2019, 8, e1512455. [Google Scholar] [CrossRef] [PubMed]
- Pan, Z.; Scheerens, H.; Li, S.-J.; Schultz, B.E.; Sprengeler, P.A.; Burrill, L.C.; Mendonca, R.V.; Sweeney, M.D.; Scott, K.C.K.; Grothaus, P.G.; et al. Discovery of Selective Irreversible Inhibitors for Bruton’s Tyrosine Kinase. ChemMedChem 2007, 2, 58–61. [Google Scholar] [CrossRef]
- Vanden Berghe, W.; Sabbe, L.; Kaileh, M.; Haegeman, G.; Heyninck, K. Molecular insight in the multifunctional activities of Withaferin A. Biochem. Pharmacol. 2012, 84, 1282–1291. [Google Scholar] [CrossRef]
- Grover, A.; Shandilya, A.; Agrawal, V.; Pratik, P.; Bhasme, D.; Bisaria, V.S.; Sundar, D. Hsp90/Cdc37 chaperone/co-chaperone complex, a novel junction anticancer target elucidated by the mode of action of herbal drug Withaferin A. BMC Bioinform. 2011, 12 (Suppl. S1), S30. [Google Scholar] [CrossRef] [Green Version]
- Bradshaw, J.M.; McFarland, J.M.; Paavilainen, V.O.; Bisconte, A.; Tam, D.; Phan, V.T.; Romanov, S.; Finkle, D.; Shu, J.; Patel, V.; et al. Prolonged and tunable residence time using reversible covalent kinase inhibitors. Nat. Chem. Biol. 2015, 11, 525–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, C.U.; Grossmann, T.N. Reversible covalent inhibition of a protein target. Angew. Chem. Int. Ed. Engl. 2012, 51, 8699–8700. [Google Scholar] [CrossRef] [PubMed]
- Serafimova, I.M.; Pufall, M.A.; Krishnan, S.; Duda, K.; Cohen, M.S.; Maglathlin, R.L.; McFarland, J.M.; Miller, R.M.; Frodin, M.; Taunton, J. Reversible targeting of noncatalytic cysteines with chemically tuned electrophiles. Nat. Chem. Biol. 2012, 8, 471–476. [Google Scholar] [CrossRef]
- Leproult, E.; Barluenga, S.; Moras, D.; Wurtz, J.M.; Winssinger, N. Cysteine mapping in conformationally distinct kinase nucleotide binding sites: Application to the design of selective covalent inhibitors. J. Med. Chem. 2011, 54, 1347–1355. [Google Scholar] [CrossRef]
- Chaikuad, A.; Koch, P.; Laufer, S.A.; Knapp, S. The Cysteinome of Protein Kinases as a Target in Drug Development. Angew. Chem. Int. Ed. 2018, 57, 4372–4385. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Gray, N.S. Rational design of inhibitors that bind to inactive kinase conformations. Nat. Chem. Biol. 2006, 2, 358–364. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Yang, P.L.; Gray, N.S. Targeting cancer with small molecule kinase inhibitors. Nat. Rev. Cancer 2009, 9, 28–39. [Google Scholar] [CrossRef] [PubMed]
- Liu, Q.; Sabnis, Y.; Zhao, Z.; Zhang, T.; Buhrlage, S.J.; Jones, L.H.; Gray, N.S. Developing irreversible inhibitors of the protein kinase cysteinome. Chem. Biol. 2013, 20, 146–159. [Google Scholar] [CrossRef] [Green Version]
- Laskowski, R.A.; Swindells, M.B. LigPlot+: Multiple ligand-protein interaction diagrams for drug discovery. J. Chem. Inf. Model 2011, 51, 2778–2786. [Google Scholar] [CrossRef]
- Gu, C.; Peng, H.; Lu, Y.; Yang, H.; Tian, Z.; Yin, G.; Zhang, W.; Lu, S.; Zhang, Y.; Yang, Y. BTK suppresses myeloma cellular senescence through activating AKT/P27/Rb signaling. Oncotarget 2017, 8, 56858–56867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, Z.; Bourne, P.E. Progress with covalent small-molecule kinase inhibitors. Drug Discov. Today 2018, 23, 727–735. [Google Scholar] [CrossRef]
- Rao, S.; Gurbani, D.; Du, G.; Everley, R.A.; Browne, C.M.; Chaikuad, A.; Tan, L.; Schroder, M.; Gondi, S.; Ficarro, S.B.; et al. Leveraging Compound Promiscuity to Identify Targetable Cysteines within the Kinome. Cell Chem. Biol. 2019, 26, 818–829.e9. [Google Scholar] [CrossRef] [PubMed]
- Prasanna, K.S.P.; Bharathi, P.S. Withaferin A suppresses the expression of vascular endothelial growth factor in Ehrlich ascites tumor cells via Sp1 transcription factor. Curr. Trends Biotechnol. Pharm. 2010, 1, 138–148. [Google Scholar]
- Bottoni, A.; Rizzotto, L.; Lai, T.H.; Liu, C.; Smith, L.L.; Mantel, R.; Reiff, S.; El-Gamal, D.; Larkin, K.; Johnson, A.J.; et al. Targeting BTK through microRNA in chronic lymphocytic leukemia. Blood 2016, 128, 3101–3112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Terzi, H.E.M. In vitro comparison of cytotoxic effects of bortezomib resistant U266 myeloma cell line (U266/VELR) on combination of ibrutinib with carfilzomib and lenalidomid drugs. Cumhur. Med. J. 2019, 41, 698–702. [Google Scholar] [CrossRef]
- Kraus, J.; Kraus, M.; Liu, N.; Besse, L.; Bader, J.; Geurink, P.P.; de Bruin, G.; Kisselev, A.F.; Overkleeft, H.; Driessen, C. The novel beta2-selective proteasome inhibitor LU-102 decreases phosphorylation of I kappa B and induces highly synergistic cytotoxicity in combination with ibrutinib in multiple myeloma cells. Cancer Chemother. Pharmacol. 2015, 76, 383–396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, J.; Gong, W.; Liu, S.; Li, Q.; Guo, M.; Wang, J.; Wang, S.; Chen, N.; Wang, Y.; Liu, Q.; et al. Ibrutinib targets microRNA-21 in multiple myeloma cells by inhibiting NF-kappaB and STAT3. Tumour. Biol. 2018, 40, 1010428317731369. [Google Scholar] [CrossRef] [Green Version]
- Rushworth, S.A.; Bowles, K.M.; Barrera, L.N.; Murray, M.Y.; Zaitseva, L.; MacEwan, D.J. BTK inhibitor ibrutinib is cytotoxic to myeloma and potently enhances bortezomib and lenalidomide activities through NF-kappaB. Cell Signal 2013, 25, 106–112. [Google Scholar] [CrossRef] [PubMed]
- Lindvall, J.M.; Blomberg, K.E.; Berglof, A.; Yang, Q.; Smith, C.I.; Islam, T.C. Gene expression profile of B cells from Xid mice and Btk knockout mice. Eur. J. Immunol. 2004, 34, 1981–1991. [Google Scholar] [CrossRef]
- Roman-Garcia, S.; Merino-Cortes, S.V.; Gardeta, S.R.; de Bruijn, M.J.W.; Hendriks, R.W.; Carrasco, Y.R. Distinct Roles for Bruton’s Tyrosine Kinase in B Cell Immune Synapse Formation. Front. Immunol. 2018, 9, 2027. [Google Scholar] [CrossRef] [PubMed]
- Chirumamilla, C.S.; Perez-Novo, C.; Van Ostade, X.; Vanden Berghe, W. Molecular insights into cancer therapeutic effects of the dietary medicinal phytochemical withaferin A. Proc. Nutr. Soc. 2017, 76, 96–105. [Google Scholar] [CrossRef]
- Grossman, E.A.; Ward, C.C.; Spradlin, J.N.; Bateman, L.A.; Huffman, T.R.; Miyamoto, D.K.; Kleinman, J.I.; Nomura, D.K. Covalent Ligand Discovery against Druggable Hotspots Targeted by Anti-cancer Natural Products. Cell Chem. Biol. 2017, 24, 1368–1376.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shinohara, H.; Yasuda, T.; Aiba, Y.; Sanjo, H.; Hamadate, M.; Watarai, H.; Sakurai, H.; Kurosaki, T. PKC beta regulates BCR-mediated IKK activation by facilitating the interaction between TAK1 and CARMA1. J. Exp. Med. 2005, 202, 1423–1431. [Google Scholar] [CrossRef]
- Mohammad, D.K.; Nore, B.F.; Smith, C.I.E. Terminating B cell receptor signaling. Oncotarget 2017, 8, 109857–109858. [Google Scholar] [CrossRef] [PubMed]
- Franks, S.E.; Cambier, J.C. Putting on the Brakes: Regulatory Kinases and Phosphatases Maintaining B Cell Anergy. Front. Immunol. 2018, 9, 665. [Google Scholar] [CrossRef] [PubMed]
- Dom, M.; Vanden Berghe, W.; Van Ostade, X. Broad-spectrum antitumor properties of Withaferin A: A proteomic perspective. RSC Med. Chem. 2020, 11, 30–50. [Google Scholar] [CrossRef] [PubMed]
- Widodo, N.; Priyandoko, D.; Shah, N.; Wadhwa, R.; Kaul, S.C. Selective killing of cancer cells by Ashwagandha leaf extract and its component Withanone involves ROS signaling. PLoS ONE 2010, 5, e13536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pires, N.; Gota, V.; Gulia, A.; Hingorani, L.; Agarwal, M.; Puri, A. Safety and pharmacokinetics of Withaferin-A in advanced stage high grade osteosarcoma: A phase I trial. J. Ayurveda Integr. Med. 2020, 11, 68–72. [Google Scholar] [CrossRef]
- Patel, S.B.; Rao, N.J.; Hingorani, L.L. Safety assessment of Withania somnifera extract standardized for Withaferin A: Acute and sub-acute toxicity study. J. Ayurveda Integr. Med. 2016, 7, 30–37. [Google Scholar] [CrossRef] [Green Version]
- Thaiparambil, J.T.; Bender, L.; Ganesh, T.; Kline, E.; Patel, P.; Liu, Y.; Tighiouart, M.; Vertino, P.M.; Harvey, R.D.; Garcia, A.; et al. Withaferin A inhibits breast cancer invasion and metastasis at sub-cytotoxic doses by inducing vimentin disassembly and serine 56 phosphorylation. Int. J. Cancer 2011, 129, 2744–2755. [Google Scholar] [CrossRef]
- Smith, P.K.; Krohn, R.I.; Hermanson, G.T.; Mallia, A.K.; Gartner, F.H.; Provenzano, M.D.; Fujimoto, E.K.; Goeke, N.M.; Olson, B.J.; Klenk, D.C. Measurement of protein using bicinchoninic acid. Anal. Biochem. 1985, 150, 76–85. [Google Scholar] [CrossRef]
- Hilhorst, R.; Houkes, L.; Mommersteeg, M.; Musch, J.; van den Berg, A.; Ruijtenbeek, R. Peptide microarrays for profiling of serine/threonine kinase activity of recombinant kinases and lysates of cells and tissue samples. Methods Mol. Biol. 2013, 977, 259–271. [Google Scholar] [CrossRef] [PubMed]
- Folkvord, S.; Flatmark, K.; Dueland, S.; de Wijn, R.; Groholt, K.K.; Hole, K.H.; Nesland, J.M.; Ruijtenbeek, R.; Boender, P.J.; Johansen, M.; et al. Prediction of response to preoperative chemoradiotherapy in rectal cancer by multiplex kinase activity profiling. Int. J. Radiat. Oncol. Biol. Phys. 2010, 78, 555–562. [Google Scholar] [CrossRef] [PubMed]
- Maat, W.; el Filali, M.; Dirks-Mulder, A.; Luyten, G.P.; Gruis, N.A.; Desjardins, L.; Boender, P.; Jager, M.J.; van der Velden, P.A. Episodic Src activation in uveal melanoma revealed by kinase activity profiling. Br. J. Cancer 2009, 101, 312–319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Safaei, J.; Manuch, J.; Gupta, A.; Stacho, L.; Pelech, S. Prediction of 492 human protein kinase substrate specificities. Proteome Sci. 2011, 9 (Suppl. S1), S6. [Google Scholar] [CrossRef] [Green Version]
- Versele, M.; Talloen, W.; Rockx, C.; Geerts, T.; Janssen, B.; Lavrijssen, T.; King, P.; Gohlmann, H.W.; Page, M.; Perera, T. Response prediction to a multitargeted kinase inhibitor in cancer cell lines and xenograft tumors using high-content tyrosine peptide arrays with a kinetic readout. Mol. Cancer Ther. 2009, 8, 1846–1855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kinexus|PhosphoNET. Available online: http://www.phosphonet.ca/ (accessed on 21 May 2019).
- IPA, Q. Ingenuity Pathway Analysis. Available online: http://www.ingenuity.com/products/ipa (accessed on 15 September 2020).
- Andrews, S. FastQC: A Quality Control Tool for High Throughput Sequence Data; Babraham Bioinformatics, Babraham Institute: Cambridge, UK, 2010. [Google Scholar]
- Dobin, A.; Davis, C.A.; Schlesinger, F.; Drenkow, J.; Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras, T.R. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 2013, 29, 15–21. [Google Scholar] [CrossRef]
- Thomas, P.D.; Campbell, M.J.; Kejariwal, A.; Mi, H.; Karlak, B.; Daverman, R.; Diemer, K.; Muruganujan, A.; Narechania, A. PANTHER: A library of protein families and subfamilies indexed by function. Genome Res. 2003, 13, 2129–2141. [Google Scholar] [CrossRef] [Green Version]
- Szarc vel Szic, K.; Op de Beeck, K.; Ratman, D.; Wouters, A.; Beck, I.M.; Declerck, K.; Heyninck, K.; Fransen, E.; Bracke, M.; De Bosscher, K.; et al. Pharmacological levels of Withaferin A (Withania somnifera) trigger clinically relevant anticancer effects specific to triple negative breast cancer cells. PLoS ONE 2014, 9, e87850. [Google Scholar] [CrossRef] [Green Version]
- Schindelin, J.; Arganda-Carreras, I.; Frise, E.; Kaynig, V.; Longair, M.; Pietzsch, T.; Preibisch, S.; Rueden, C.; Saalfeld, S.; Schmid, B.; et al. Fiji: An open-source platform for biological-image analysis. Nat. Methods 2012, 9, 676–682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heyninck, K.; Sabbe, L.; Chirumamilla, C.S.; Szarc Vel Szic, K.; Vander Veken, P.; Lemmens, K.J.A.; Lahtela-Kakkonen, M.; Naulaerts, S.; Op de Beeck, K.; Laukens, K.; et al. Withaferin A induces heme oxygenase (HO-1) expression in endothelial cells via activation of the Keap1/Nrf2 pathway. Biochem. Pharmacol. 2016, 109, 48–61. [Google Scholar] [CrossRef]
- Guex, N.; Peitsch, M.C. SWISS-MODEL and the Swiss-PdbViewer: An environment for comparative protein modeling. Electrophoresis 1997, 18, 2714–2723. [Google Scholar] [CrossRef] [PubMed]
- Pettersen, E.F.; Goddard, T.D.; Huang, C.C.; Couch, G.S.; Greenblatt, D.M.; Meng, E.C.; Ferrin, T.E. UCSF Chimera—A visualization system for exploratory research and analysis. J. Comput. Chem. 2004, 25, 1605–1612. [Google Scholar] [CrossRef] [Green Version]
- PyMOL. The PyMOL Molecular Graphics System, Version 2.0; PyMOL: Schrödinger, NY, USA, 2017. [Google Scholar]
- Dayalan Naidu, S.; Dinkova-Kostova, A.T. KEAP1, a cysteine-based sensor and a drug target for the prevention and treatment of chronic disease. Open Biol. 2020, 10, 200105. [Google Scholar] [CrossRef] [PubMed]
- Kobayashi, M.; Li, L.; Iwamoto, N.; Nakajima-Takagi, Y.; Kaneko, H.; Nakayama, Y.; Eguchi, M.; Wada, Y.; Kumagai, Y.; Yamamoto, M. The antioxidant defense system Keap1-Nrf2 comprises a multiple sensing mechanism for responding to a wide range of chemical compounds. Mol. Cell Biol. 2009, 29, 493–502. [Google Scholar] [CrossRef] [Green Version]
Gene Ontology Term | Symbol | Gene ID | Name | Log2FC | p-Adj. |
---|---|---|---|---|---|
ECM and cell–cell adhesion | RELN * | 5649 | Reelin | 2.9 | 7.4 × 10−160 |
PLXNB2 | 23654 | Plexin B2 | 1.8 | 7.1 × 10−96 | |
PODXL2 * | 50512 | Podocalyxin like 2 | 2.6 | 6.8 × 10−83 | |
ESAM | 90952 | Endothelial cell adhesion molecule | 2.0 | 1.7 × 10−80 | |
PRKX * | 5613 | Protein kinase X-linked | 1.1 | 8.1 × 10−79 | |
ACP5 | 54 | Acid phosphatase 5, tartrate-resistant | 4.8 | 2.0 × 10−77 | |
GPCR signaling | GNG7 | 2788 | G protein subunit gamma 7 | 1.5 | 5.7 × 10−99 |
UTS2R | 2837 | Urotensin 2 receptor | 1.9 | 5.4 × 10−77 | |
BCR signaling | BTK * | 695 | Bruton’s tyrosine kinase | 2.8 | 1.5 × 10−216 |
TNFRSF8 | 943 | TNF receptor superfamily member 8 | 3.7 | 6.9 × 10−93 | |
CD52 | 1043 | CD52 molecule | 3.5 | 5.6 × 10−83 | |
mRNA/protein stability | CTAG2 | 30848 | Cancer/testis antigen 2 | 8.5 | 4.0 × 10−168 |
LINC01518 | 101929397 | Long intergenic non-protein Coding RNA 1518 | 7.9 | 1.4 × 10−147 | |
CMTR1 | 23070 | Cap methyltransferase 1 | −1.1 | 9.6 × 10−115 | |
TMEM25 | 84866 | Transmembrane protein 25 | 4.8 | 6.0 × 10−79 | |
Cell cycle regulation | CDKN2A | 1029 | Cyclin-dependent kinase inhibitor 2A | 9.4 | 1.7 × 10−210 |
Cytoskeleton | TUBB4A | 10382 | Tubulin beta 4A class IVa | 3.6 | 6.1 × 10−165 |
Inflammation | NLRP11 | 204801 | NLR family pyrin domain containing 11 | 4.9 | 1.2 × 10−220 |
Transmembrane transport | SLC38A5 | 92745 | Solute carrier family 38 member 5 | 1.8 | 1.7 × 10−92 |
ABCG2* | 9429 | ATP-binding cassette subfamily G member 2 | 5.6 | 2.0 × 10−84 |
Site | Subsite | Representative Kinases |
---|---|---|
Gatekeeper region | GK | MOK |
GK + 1 | SgK494 | |
GK − 1 | MAP2K4, MKK3, MAP2K6, KHS1, KHS2, GCK | |
DFG region | DFG + 1 | MAP3K8, MOS, MAP3K4, PINK1 |
DFG + 2 | PKCz, PKCi, AKT1, AKT2, AKT3, PKCg, SGK1F, SGK2 | |
DFG − 1 | PBK, TGFbR2, CDKL3, CDKL2, PRP4, MNK2, MNK1 | |
Glycine rich loop region | Glycineloop | WNK4, WNK1, WNK2, WNK3, HER3 |
Glycineloop 1 | ZAK | |
Glycineloop 2 | SgK496, MEKK1, PLK2, PLK3, PLK1, RSK1 | |
Glycineloop 3 | SgK493 | |
Glycineloop5 | FGFR1, FGFR2, FGFR3, FGFR4 | |
Hinge binding region | Hinge 1 | FGFR4, TTK, MAPKAPK2, MAPKAPK3 |
Hinge 2 | IKKa, IKKb, LKB1, NEK4, Wee1, SLK, FLT4, KDR | |
Hinge 3 | Ron, FGR, SgK494, Kit, CSFR, FLT3 | |
Hinge 4 | SgK110, BubR1, LKB1, TBK1 | |
Hinge 5 | PINK1, EphB3 | |
Hinge 6 | MAP2K7, TEC, TXK, ITK, BTK, BMX, BLK, HER2, EGFR, HER4, JAK3 | |
Hinge 7 | JNK1, JNK2, JNK3 | |
Roof region | Roof sheet | HER3 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Logie, E.; Chirumamilla, C.S.; Perez-Novo, C.; Shaw, P.; Declerck, K.; Palagani, A.; Rangarajan, S.; Cuypers, B.; De Neuter, N.; Mobashar Hussain Urf Turabe, F.; et al. Covalent Cysteine Targeting of Bruton’s Tyrosine Kinase (BTK) Family by Withaferin-A Reduces Survival of Glucocorticoid-Resistant Multiple Myeloma MM1 Cells. Cancers 2021, 13, 1618. https://doi.org/10.3390/cancers13071618
Logie E, Chirumamilla CS, Perez-Novo C, Shaw P, Declerck K, Palagani A, Rangarajan S, Cuypers B, De Neuter N, Mobashar Hussain Urf Turabe F, et al. Covalent Cysteine Targeting of Bruton’s Tyrosine Kinase (BTK) Family by Withaferin-A Reduces Survival of Glucocorticoid-Resistant Multiple Myeloma MM1 Cells. Cancers. 2021; 13(7):1618. https://doi.org/10.3390/cancers13071618
Chicago/Turabian StyleLogie, Emilie, Chandra S. Chirumamilla, Claudina Perez-Novo, Priyanka Shaw, Ken Declerck, Ajay Palagani, Savithri Rangarajan, Bart Cuypers, Nicolas De Neuter, Fazil Mobashar Hussain Urf Turabe, and et al. 2021. "Covalent Cysteine Targeting of Bruton’s Tyrosine Kinase (BTK) Family by Withaferin-A Reduces Survival of Glucocorticoid-Resistant Multiple Myeloma MM1 Cells" Cancers 13, no. 7: 1618. https://doi.org/10.3390/cancers13071618
APA StyleLogie, E., Chirumamilla, C. S., Perez-Novo, C., Shaw, P., Declerck, K., Palagani, A., Rangarajan, S., Cuypers, B., De Neuter, N., Mobashar Hussain Urf Turabe, F., Kumar Verma, N., Bogaerts, A., Laukens, K., Offner, F., Van Vlierberghe, P., Van Ostade, X., & Berghe, W. V. (2021). Covalent Cysteine Targeting of Bruton’s Tyrosine Kinase (BTK) Family by Withaferin-A Reduces Survival of Glucocorticoid-Resistant Multiple Myeloma MM1 Cells. Cancers, 13(7), 1618. https://doi.org/10.3390/cancers13071618