1
|
Vestuto V, Ciaglia T, Musella S, Di Sarno V, Smaldone G, Di Matteo F, Scala MC, Napolitano V, Miranda MR, Amodio G, Novi S, Pepe G, Basilicata MG, Gazzillo E, Pace S, Gomez-Monterrey IM, Sala M, Bifulco G, Tecce MF, Campiglia P, Ostacolo C, Lauro G, Manfra M, Bertamino A. A Comprehensive In Vitro Characterization of a New Class of Indole-Based Compounds Developed as Selective Haspin Inhibitors. J Med Chem 2024; 67:12711-12734. [PMID: 39038808 PMCID: PMC11320660 DOI: 10.1021/acs.jmedchem.4c00718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/24/2024]
Abstract
Haspin is an emerging, but rather unexplored, divergent kinase involved in tumor growth by regulating the mitotic phase. In this paper, the in-silico design, synthesis, and biological characterization of a new series of substituted indoles acting as potent Haspin inhibitors are reported. The synthesized derivatives have been evaluated by FRET analysis, showing very potent Haspin inhibition. Then, a comprehensive in-cell investigation highlighted compounds 47 and 60 as the most promising inhibitors. These compounds were challenged for their synergic activity with paclitaxel in 2D and 3D cellular models, demonstrating a twofold improvement of the paclitaxel antitumor activity. Compound 60 also showed remarkable selectivity when tested in a panel of 70 diverse kinases. Finally, in-silico studies provided new insight about the chemical requirements useful to develop new Haspin inhibitors. Biological results, together with the drug-likeness profile of 47 and 60, make these derivatives deserving further studies.
Collapse
Affiliation(s)
- Vincenzo Vestuto
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Tania Ciaglia
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Simona Musella
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Veronica Di Sarno
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Gerardina Smaldone
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Francesca Di Matteo
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Maria Carmina Scala
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Valeria Napolitano
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Maria Rosaria Miranda
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Giuseppina Amodio
- Department
of Medicine, Surgery and Dentistry “Scuola Medica Salernitana″, University of Salerno, Salerno , Baronissi 84034, Italy
| | - Sara Novi
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Giacomo Pepe
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Manuela Giovanna Basilicata
- Department
of Advanced Medical and Surgical Science, University of Campania “Luigi Vanvitelli”, P.zza L. Miraglia 2, Naples 80138, Italy
| | - Erica Gazzillo
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Simona Pace
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | | | - Marina Sala
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Giuseppe Bifulco
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Mario Felice Tecce
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Pietro Campiglia
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Carmine Ostacolo
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Gianluigi Lauro
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| | - Michele Manfra
- Department
of Science, University of Basilicata, Via dell’Ateneo Lucano 10 , Potenza 85100, Italy
| | - Alessia Bertamino
- Department
of Pharmacy, University of Salerno, Via G. Paolo II 132 , Salerno , Fisciano 84084, Italy
| |
Collapse
|
2
|
Shawky MM, Abdallah M, Khalifa H, Aboushady Y, Abadi AH, Engel M, Abdel-Halim M. Synthesis and evaluation of novel N1-acylated 5-(4-pyridinyl)indazole derivatives as potent and selective haspin inhibitors. Bioorg Chem 2024; 145:107235. [PMID: 38447464 DOI: 10.1016/j.bioorg.2024.107235] [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: 01/10/2024] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024]
Abstract
Protein kinase dysregulation was strongly linked to cancer pathogenesis. Moreover, histone alterations were found to be among the most important post-translational modifications that could contribute to cancer growth and development. In this context, haspin, an atypical serine/threonine kinase, phosphorylates histone H3 at threonine-3 and is notably overexpressed in various common cancer types. Herein, we report novel 5-(4-pyridinyl)indazole derivatives as potent and selective haspin inhibitors. Amide coupling at N1 of the indazole ring with m-hydroxyphenyl acetic acid yielded compound 21 with an IC50 value of 78 nM against haspin. This compound showed a meaningful selectivity over 15 of the most common off-targets, including Clk 1-3 and Dyrk1A, 1B, and 2. The most potent haspin inhibitors 5 and 21 effectively inhibited the growth of the NCI-60 cancer cell lines, further emphasizing the success of our scaffold as a new selective lead for the development of anti-cancer therapeutic agents.
Collapse
Affiliation(s)
- Mona M Shawky
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Mennatallah Abdallah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Hend Khalifa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Youssef Aboushady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Ashraf H Abadi
- 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, D-66123 Saarbrücken, Germany.
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt.
| |
Collapse
|
3
|
Zha J, He J, Wu C, Zhang M, Liu X, Zhang J. Designing drugs and chemical probes with the dualsteric approach. Chem Soc Rev 2023; 52:8651-8677. [PMID: 37990599 DOI: 10.1039/d3cs00650f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Traditionally, drugs are monovalent, targeting only one site on the protein surface. This includes orthosteric and allosteric drugs, which bind the protein at orthosteric and allosteric sites, respectively. Orthosteric drugs are good in potency, whereas allosteric drugs have better selectivity and are solutions to classically undruggable targets. However, it would be difficult to simultaneously reach high potency and selectivity when targeting only one site. Also, both kinds of monovalent drugs suffer from mutation-caused drug resistance. To overcome these obstacles, dualsteric modulators have been proposed in the past twenty years. Compared to orthosteric or allosteric drugs, dualsteric modulators are bivalent (or bitopic) with two pharmacophores. Each of the two pharmacophores bind the protein at the orthosteric and an allosteric site, which could bring the modulator with special properties beyond monovalent drugs. In this study, we comprehensively review the current development of dualsteric modulators. Our main effort reason and illustrate the aims to apply the dualsteric approach, including a "double win" of potency and selectivity, overcoming mutation-caused drug resistance, developments of function-biased modulators, and design of partial agonists. Moreover, the strengths of the dualsteric technique also led to its application outside pharmacy, including the design of highly sensitive fluorescent tracers and usage as molecular rulers. Besides, we also introduced drug targets, designing strategies, and validation methods of dualsteric modulators. Finally, we detail the conclusions and perspectives.
Collapse
Affiliation(s)
- Jinyin Zha
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China.
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jixiao He
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengwei Wu
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingyang Zhang
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyi Liu
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China.
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Zhang
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China.
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Lavogina D, Nasirova N, Sõrmus T, Tähtjärv T, Enkvist E, Viht K, Haljasorg T, Herodes K, Jaal J, Uri A. Conjugates of adenosine mimetics and arginine-rich peptides serve as inhibitors and fluorescent probes but not as long-lifetime photoluminescent probes for protein arginine methyltransferases. J Pept Sci 2023; 29:e3456. [PMID: 36208424 DOI: 10.1002/psc.3456] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
Abstract
The conjugates of an adenosine mimetic and oligo-l-arginine or oligo-d-arginine (ARCs) were initially designed in our research group as inhibitors and photoluminescent probes targeting basophilic protein kinases. Here, we explored a panel of ARCs and their fluorescent derivatives in biochemical assays with members of the protein arginine methyltransferase (PRMT) family, focusing specifically on PRMT1. In the binding/displacement assay with detection of fluorescence anisotropy, we found that ARCs and arginine-rich peptides could serve as high-affinity ligands for PRMT1, whereas the equilibrium dissociation constant values depended dramatically on the number of arginine residues within the compounds. The fluorescently labeled probe ARC-1081 was displaced from its complex with PRMT1 by both S-adenosyl-l-methionine (SAM) and S-adenosyl-l-homocysteine (SAH), indicating binding of the adenosine mimetic of ARCs to the SAM/SAH-binding site within PRMT1. The ARCs that had previously shown microsecond-lifetime photoluminescence in complex with protein kinases did not feature such property in complex with PRMT1, demonstrating the selectivity of the time-resolved readout format. When tested against a panel of PRMT family members in single-dose inhibition experiments, a micromolar concentration of ARC-902 was required for the inhibition of PRMT1 and PRMT7. Overall, our results suggest that the compounds containing multiple arginine residues (including the well-known cell-penetrating peptides) are likely to inhibit PRMT and thus interfere with the epigenetic modification status in complex biological systems, which should be taken into consideration during interpretation of the experimental data.
Collapse
Affiliation(s)
- Darja Lavogina
- Institute of Chemistry, University of Tartu, Tartu, Estonia.,Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Naila Nasirova
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Tanel Sõrmus
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Taavo Tähtjärv
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Erki Enkvist
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Kaido Viht
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Tõiv Haljasorg
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Koit Herodes
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Jana Jaal
- Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.,Department of Radiotherapy and Oncological Therapy, Tartu University Hospital, Tartu, Estonia
| | - Asko Uri
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| |
Collapse
|
5
|
Liu Y, Yang H, Fang Y, Xing Y, Pang X, Li Y, Zhang Y, Liu Y. Function and inhibition of Haspin kinase: targeting multiple cancer therapies by antimitosis. J Pharm Pharmacol 2022; 75:445-465. [PMID: 36334086 DOI: 10.1093/jpp/rgac080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022]
Abstract
Abstract
Objectives
Haploid germ cell-specific nuclear protein kinase (Haspin) is a serine/threonine kinase as an atypical kinase, which is structurally distinct from conventional protein kinases.
Key findings
Functionally, Haspin is involved in important cell cycle progression, particularly in critical mitosis regulating centromeric sister chromatid cohesion during prophase and prometaphase, and subsequently ensuring proper chromosome alignment during metaphase and the normal chromosome segregation during anaphase. However, increasing evidence has demonstrated that Haspin is significantly upregulated in a variety of cancer cells in addition to normal proliferating somatic cells. Its knockdown or small molecule inhibition could prevent cancer cell growth and induce apoptosis by disrupting the regular mitotic progression. Given the specificity of its expressed tissues or cells and the uniqueness of its current known substrate, Haspin can be a promising target against cancer. Consequently, selective synthetic and natural inhibitors of Haspin have been widely developed to determine their inhibitory power for various cancer cells in vivo and in vitro.
Summary
Here our perspective includes a comprehensive review of the roles and structure of Haspin, its relatively potent and selective inhibitors and Haspin’s preliminary studies in a variety of cancers.
Collapse
Affiliation(s)
- Yongjian Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Hongliu Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Yongsheng Fang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Yantao Xing
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Xinxin Pang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Yang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Yuanyuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| | - Yonggang Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine , Beijing , China
| |
Collapse
|
6
|
Li L, Li S, Wang H, Li L, Wang P, Shen D, Dang X. GSG2 promotes tumor growth through regulating cell proliferation in hepatocellular carcinoma. Biochem Biophys Res Commun 2022; 625:109-115. [DOI: 10.1016/j.bbrc.2022.07.093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 07/23/2022] [Indexed: 12/24/2022]
|
7
|
Yu W, Liu W, Feng Y, Zhu C. Knockdown of GSG2 Suppresses the Progression of Colorectal Cancer Cells. Genet Test Mol Biomarkers 2022; 26:26-36. [PMID: 35089075 DOI: 10.1089/gtmb.2020.0298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction: As a serine/threonine kinase, Haspin (GSG2) has been reportedly associated with the development of malignant tumors. However, few studies have reported the role of GSG2 in colorectal cancer (CRC). Materials and Methods: Based on data from the Oncomine databases, GSG2 was found to be highly expressed in CRC patients' tissues. Therefore, the expression of GSG2 in CRC cell lines was subsequently evaluated. GSG2 loss-of-function experiments were conducted by infection with a lentivirus expressing shRNAs against GSG2. Colony-formation and cell viabilities were assessed using clonogenic and 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays, respectively. Migration was assessed using wound-healing and transwell assays. A GSG2 inhibitor experiment was used to investigate the key role of GSG2 in CRC. Immunoprecipitation was used to investigate the interaction between GSG2 and p-H3. In addition, apoptosis was evaluated by quantifying caspase 3/7 activities, and western blot analyses were used to investigate the underlying mechanisms of GSG2 in CRC. Results: GSG2 was found to be highly expressed in CRC tissues and cells. Furthermore, GSG2 knock-down suppressed proliferation, colony formation and invasion, and induced apoptosis in CRC cells. Mechanistically, GSG2 was revealed to regulate Myc, NF-κB, Snail-1, and β-catenin signaling. Conclusion: Collectively, we demonstrate that GSG2 is a potential biomarker of CRC, and that GSG2 interference suppresses the progression of CRC and promotes apoptosis in vitro. These data suggest GSG2 as a putative oncogene, but will require additional in vivo studies to confirm.
Collapse
Affiliation(s)
- Wenyan Yu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Liu
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuxin Feng
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chunrong Zhu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
8
|
Crystal Structure-Guided Design of Bisubstrate Inhibitors and Photoluminescent Probes for Protein Kinases of the PIM Family. Molecules 2021; 26:molecules26144353. [PMID: 34299628 PMCID: PMC8307404 DOI: 10.3390/molecules26144353] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 11/23/2022] Open
Abstract
We performed an X-ray crystallographic study of complexes of protein kinase PIM-1 with three inhibitors comprising an adenosine mimetic moiety, a linker, and a peptide-mimetic (d-Arg)6 fragment. Guided by the structural models, simplified chemical structures with a reduced number of polar groups and chiral centers were designed. The developed inhibitors retained low-nanomolar potency and possessed remarkable selectivity toward the PIM kinases. The new inhibitors were derivatized with biotin or fluorescent dye Cy5 and then applied for the detection of PIM kinases in biochemical solutions and in complex biological samples. The sandwich assay utilizing a PIM-2-selective detection antibody featured a low limit of quantification (44 pg of active recombinant PIM-2). Fluorescent probes were efficiently taken up by U2OS cells and showed a high extent of co-localization with PIM-1 fused with a fluorescent protein. Overall, the developed inhibitors and derivatives represent versatile chemical tools for studying PIM function in cellular systems in normal and disease physiology.
Collapse
|
9
|
Lee S, Kim J, Jo J, Chang JW, Sim J, Yun H. Recent advances in development of hetero-bivalent kinase inhibitors. Eur J Med Chem 2021; 216:113318. [PMID: 33730624 DOI: 10.1016/j.ejmech.2021.113318] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/16/2021] [Accepted: 02/16/2021] [Indexed: 12/13/2022]
Abstract
Identifying a pharmacological agent that targets only one of more than 500 kinases present in humans is an important challenge. One potential solution to this problem is the development of bivalent kinase inhibitors, which consist of two connected fragments, each bind to a dissimilar binding site of the bisubstrate enzyme. The main advantage of bivalent (type V) kinase inhibitors is generating more interactions with target enzymes that can enhance the molecules' selectivity and affinity compared to single-site inhibitors. Earlier type V inhibitors were not suitable for the cellular environment and were mostly used in in vitro studies. However, recently developed bivalent compounds have high kinase affinity, high biological and chemical stability in vivo. This review summarized the hetero-bivalent kinase inhibitors described in the literature from 2014 to the present. We attempted to classify the molecules by serine/threonine and tyrosine kinase inhibitors, and then each target kinase and its hetero-bivalent inhibitor was assessed in depth. In addition, we discussed the analysis of advantages, limitations, and perspectives of bivalent kinase inhibitors compared with the monovalent kinase inhibitors.
Collapse
Affiliation(s)
- Seungbeom Lee
- College of Pharmacy, CHA University, Pocheon-si, Gyeonggi-do, 11160, Republic of Korea
| | - Jisu Kim
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Jeyun Jo
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Jae Won Chang
- Department of Pharmacology & Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Department of Hematology & Medical Oncology, School of Medicine, Emory University, Atlanta, GA, USA; Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jaehoon Sim
- College of Pharmacy, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Hwayoung Yun
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
10
|
Zhu D, Gu X, Lin Z, Yu D, Wang J, Li L. HASPIN is involved in the progression of gallbladder carcinoma. Exp Cell Res 2020; 390:111863. [PMID: 31987787 DOI: 10.1016/j.yexcr.2020.111863] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/19/2020] [Accepted: 01/22/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Gallbladder carcinoma (GBC) is a common malignant tumor of the biliary system, but the current treatment of GBC is unsatisfactory. Therefore, new treatment targets and strategies are urgently needed. METHODS The expression of HASPIN in GBC was detected by immunohistochemical staining. HASPIN knockdown cell model was constructed by lentivirus infection, and the infection efficiency of lentivirus and knockdown efficiency of shHASPIN were verified by fluorescence immunoassay, qRT-PCR and Western blot. The effects of HASPIN knockdown on cell proliferation, clone-formation ability and apoptosis were determined by MTT, clone formation assay, flow cytometry and Human Apoptosis Antibody Array in vitro. Besides, the effect of HASPIN knockdown on the growth of GBC solid tumors was demonstrated in vivo. RESULTS The expression of HASPIN in GBC was up-regulated and positively correlated with the pathological grade of GBC. ShHASPIN significantly down-regulated the mRNA and protein levels of HASPIN, suggesting that HASPIN knockdown cell model was successfully constructed in vitro. After HASPIN knockdown, the proliferation and clone-formation ability of GBC cells were observably inhibited, the apoptotic levels were markedly increased, and the expression of Caspase 3, IGFBP-5, p21 and sTNF-R1 related to apoptotic pathway was up-regulated. Furthermore, HASPIN knockdown inhibited the growth of GBC in vivo. CONCLUSION HASPIN was up-regulated in GBC and played an important role in promoting the progress of GBC.
Collapse
MESH Headings
- Aged
- Animals
- Apoptosis
- Carcinoma/genetics
- Carcinoma/metabolism
- Carcinoma/pathology
- Carcinoma/therapy
- Caspase 3/genetics
- Caspase 3/metabolism
- Cell Line, Tumor
- Cell Proliferation
- Clone Cells
- Cyclin-Dependent Kinase Inhibitor p21/genetics
- Cyclin-Dependent Kinase Inhibitor p21/metabolism
- Female
- Gallbladder Neoplasms/genetics
- Gallbladder Neoplasms/metabolism
- Gallbladder Neoplasms/pathology
- Gallbladder Neoplasms/therapy
- Gene Expression Regulation, Neoplastic
- Humans
- Insulin-Like Growth Factor Binding Protein 5/genetics
- Insulin-Like Growth Factor Binding Protein 5/metabolism
- Intracellular Signaling Peptides and Proteins/antagonists & inhibitors
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Mice
- Mice, Nude
- Middle Aged
- Protein Array Analysis
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Signal Transduction
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Dawei Zhu
- Department of Gynaecology and Obstetrics, Daping Hospital, Army Medical University, China
| | - Xing Gu
- Department of Gynaecology and Obstetrics, Daping Hospital, Army Medical University, China
| | - Zhenyu Lin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Dandan Yu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jing Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| | - Li Li
- Department of Gynaecology and Obstetrics, Daping Hospital, Army Medical University, China.
| |
Collapse
|
11
|
Pietsch M, Viht K, Schnitzler A, Ekambaram R, Steinkrüger M, Enkvist E, Nienberg C, Nickelsen A, Lauwers M, Jose J, Uri A, Niefind K. Unexpected CK2β-antagonistic functionality of bisubstrate inhibitors targeting protein kinase CK2. Bioorg Chem 2020; 96:103608. [PMID: 32058103 DOI: 10.1016/j.bioorg.2020.103608] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/11/2019] [Accepted: 01/20/2020] [Indexed: 01/17/2023]
Abstract
Protein kinase CK2, a heterotetrameric holoenzyme composed of two catalytic chains (CK2α) attached to a homodimer of regulatory subunits (CK2β), is a target for drug development for cancer therapy. Here, we describe the tetraiodobenzimidazole derivative ARC-3140, a bisubstrate inhibitor addressing the ATP site and the substrate-binding site of CK2 with extraordinary affinity (Ki = 84 pM). In a crystal structure of ARC-3140 in complex with CK2α, three copies of the inhibitor are visible, one of them at the CK2β interface of CK2α. Subsequent interaction studies based on microscale thermophoresis and fluorescence anisotropy changes revealed a significant impact of ARC-3140 and of its tetrabromo equivalent ARC-1502 on the CK2α/CK2β interaction. A structural inspection revealed that ARC-3140, unlike CK2β antagonists described so far, interferes with both sub-interfaces of the bipartite CK2α/CK2β interaction. Thus, ARC-3140 is a lead for the further development of highly effective compounds perturbating the quaternary structure of the CK2α2β2 holoenzyme.
Collapse
Affiliation(s)
- Markus Pietsch
- Institut II für Pharmakologie, Zentrum für Pharmakologie, Medizinische Fakultät, Universität zu Köln, Gleueler Str. 24, D-50931 Köln, Germany
| | - Kaido Viht
- Institute of Chemistry, University of Tartu, 14A Ravila St., 50411 Tartu, Estonia
| | - Alexander Schnitzler
- Institut für Biochemie, Department für Chemie, Universität zu Köln, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Ramesh Ekambaram
- Institute of Chemistry, University of Tartu, 14A Ravila St., 50411 Tartu, Estonia
| | - Michaela Steinkrüger
- Institut II für Pharmakologie, Zentrum für Pharmakologie, Medizinische Fakultät, Universität zu Köln, Gleueler Str. 24, D-50931 Köln, Germany
| | - Erki Enkvist
- Institute of Chemistry, University of Tartu, 14A Ravila St., 50411 Tartu, Estonia
| | - Christian Nienberg
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, PharmaCampus, Corrensstr. 48, D-48149 Münster, Germany
| | - Anna Nickelsen
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, PharmaCampus, Corrensstr. 48, D-48149 Münster, Germany
| | - Miriam Lauwers
- Institut II für Pharmakologie, Zentrum für Pharmakologie, Medizinische Fakultät, Universität zu Köln, Gleueler Str. 24, D-50931 Köln, Germany
| | - Joachim Jose
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, PharmaCampus, Corrensstr. 48, D-48149 Münster, Germany
| | - Asko Uri
- Institute of Chemistry, University of Tartu, 14A Ravila St., 50411 Tartu, Estonia.
| | - Karsten Niefind
- Institut für Biochemie, Department für Chemie, Universität zu Köln, Zülpicher Str. 47, D-50674 Köln, Germany.
| |
Collapse
|
12
|
Amoussou NG, Bigot A, Roussakis C, Robert JMH. Haspin: a promising target for the design of inhibitors as potent anticancer drugs. Drug Discov Today 2017; 23:409-415. [PMID: 29031622 DOI: 10.1016/j.drudis.2017.10.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/03/2017] [Accepted: 10/05/2017] [Indexed: 12/30/2022]
Abstract
Protein kinases constitute a large group of enzymes in eukaryotes and have an important role in many cellular processes. Several of these proteins are active kinases, such as haploid germ cell-specific nuclear protein kinase (Haspin), an atypical eukaryotic protein kinase that lacks sequence similarity with other eukaryotic protein kinases. Haspin is a serine/threonine kinase that associates with chromosome and phosphorylates threonine 3 of histone 3 during mitosis. Haspin overexpression or deletion results in defective mitosis. It has been shown that Haspin inhibitors have potent anti-tumoral effects. Given that the only Haspin substrate is threonine 3 of histone 3, inhibition of Haspin might have fewer adverse effects compared with other anticancer agents. Here, we highlight the chemical structures and actions of currently known Haspin inhibitors.
Collapse
Affiliation(s)
- Nathalie Gisèle Amoussou
- Université de Nantes, Nantes Atlantique Universités, Cibles et Médicaments du Cancer et de l'Immunité IICiMed-AE1155, Institut de Recherche en Santé 2, 22, rue Bénoni-Goulin, F-44000 Nantes, France; Université d'Abomey-Calavi, Faculté des Sciences de la Santé, Laboratoire de Chimie Pharmaceutique Organique, 01 BP 188 Cotonou, Benin
| | - André Bigot
- Université d'Abomey-Calavi, Faculté des Sciences de la Santé, Unité d'Enseignement et de Recherche en Immunologie, 01 BP 188 Cotonou, Benin
| | - Christos Roussakis
- Université de Nantes, Nantes Atlantique Universités, Cibles et Médicaments du Cancer et de l'Immunité IICiMed-AE1155, Institut de Recherche en Santé 2, 22, rue Bénoni-Goulin, F-44000 Nantes, France
| | - Jean-Michel H Robert
- Université de Nantes, Nantes Atlantique Universités, Cibles et Médicaments du Cancer et de l'Immunité IICiMed-AE1155, Institut de Recherche en Santé 2, 22, rue Bénoni-Goulin, F-44000 Nantes, France.
| |
Collapse
|
13
|
Han L, Wang P, Sun Y, Liu S, Dai J. Anti-Melanoma Activities of Haspin Inhibitor CHR-6494 Deployed as a Single Agent or in a Synergistic Combination with MEK Inhibitor. J Cancer 2017; 8:2933-2943. [PMID: 28928884 PMCID: PMC5604444 DOI: 10.7150/jca.20319] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/28/2017] [Indexed: 12/28/2022] Open
Abstract
Background: Melanoma is a heterogeneous malignancy that presents an immense challenge in therapeutic development. Recent approaches targeting the oncogenic MAP kinase pathways have shown tremendous improvement in the overall survival of patients with advanced melanoma. However, there is still an urgent need for identification of new strategies to overcome drug resistances and to improve therapeutic efficacy. Haspin (Haploid Germ Cell-Specific Nuclear Protein Kinase) belongs to a selected group of mitotic kinases and is required for normal mitosis progression. In contrast to inhibitors of other mitotic kinases, anti-tumor potential of haspin inhibitors has not been well explored. Herein, we aim to examine effects of CHR-6494, a small molecule inhibitor of haspin, in melanoma cells. Methods: Anti-tumor activities of the haspin inhibitor CHR-6494 were tested in a number of melanoma cell lines either as a single agent or in combination with the MEK inhibitor Trametinib (GSK1120212). Experiments are based on: 1) Cell viability determined by the crystal violet staining assay; 2) apoptotic responses measured by the caspase 3/7 activity assay and western blot analysis for the level of cleaved PARP (Poly ADP-Ribose Polymerase); 3) cell cycle analysis conducted using flow cytometry; and 4) cell migratory ability assessed by the scratch assay and the transwell migration assay. Results: We have found that CHR-6494 alone elicits a dose dependent inhibitory effect on the viability of several melanoma cell lines. This growth inhibition is accompanied by an increase in apoptotic responses. More importantly, CHR-6494 appears to synergize with the MEK inhibitor Trametinib in suppressing cell growth and enhancing apoptosis in both wild type and BRAFV600E mutant melanoma cell lines. Administering of these two small molecules as a combination is also capable of suppressing cell migration to a greater extent than the individual agent. Conclusion: These results suggest that haspin can be considered as a viable anti-melanoma target, and that concomitant inhibition of haspin and MEK activities with small molecules could represent a novel therapeutic strategy with improved efficacy for treatment of melanoma.
Collapse
Affiliation(s)
- Lili Han
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072 P. R. China
| | - Peiling Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072 P. R. China
| | - Yang Sun
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072 P. R. China
| | - Sijing Liu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072 P. R. China
| | - Jun Dai
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072 P. R. China.,Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin, Madison, WI 53705 USA.,Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, MA 02129 USA
| |
Collapse
|
14
|
Binding assay for characterization of protein kinase inhibitors possessing sub-picomolar to sub-millimolar affinity. Anal Biochem 2017; 531:67-77. [DOI: 10.1016/j.ab.2017.05.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/16/2017] [Indexed: 02/06/2023]
|
15
|
Kestav K, Viht K, Konovalov A, Enkvist E, Uri A, Lavogina D. Slowly on, Slowly off: Bisubstrate-Analogue Conjugates of 5-Iodotubercidin and Histone H3 Peptide Targeting Protein Kinase Haspin. Chembiochem 2017; 18:790-798. [DOI: 10.1002/cbic.201600697] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Indexed: 01/08/2023]
Affiliation(s)
- Katrin Kestav
- Institute of Chemistry; University of Tartu; Ravila 14A 50411 Tartu Estonia
| | - Kaido Viht
- Institute of Chemistry; University of Tartu; Ravila 14A 50411 Tartu Estonia
| | - Anton Konovalov
- Institute of Chemistry; University of Tartu; Ravila 14A 50411 Tartu Estonia
| | - Erki Enkvist
- Institute of Chemistry; University of Tartu; Ravila 14A 50411 Tartu Estonia
| | - Asko Uri
- Institute of Chemistry; University of Tartu; Ravila 14A 50411 Tartu Estonia
| | - Darja Lavogina
- Institute of Chemistry; University of Tartu; Ravila 14A 50411 Tartu Estonia
| |
Collapse
|
16
|
Lavogina D, Kestav K, Chaikuad A, Heroven C, Knapp S, Uri A. Co-crystal structures of the protein kinase haspin with bisubstrate inhibitors. Acta Crystallogr F Struct Biol Commun 2016; 72:339-45. [PMID: 27139824 PMCID: PMC4854560 DOI: 10.1107/s2053230x16004611] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/16/2016] [Indexed: 12/18/2022] Open
Abstract
Haspin is a mitotic protein kinase that is responsible for the phosphorylation of Thr3 of histone H3, thereby creating a recognition motif for docking of the chromosomal passenger complex that is crucial for the progression of cell division. Here, two high-resolution models of haspin with previously reported inhibitors consisting of an ATP analogue and a histone H3(1-7) peptide analogue are presented. The structures of the complexes confirm the bisubstrate character of the inhibitors by revealing the signature binding modes of the moieties targeting the ATP-binding site and the protein substrate-binding site of the kinase. This is the first structural model of a bisubstrate inhibitor targeting haspin. The presented structural data represent a model for the future development of more specific haspin inhibitors.
Collapse
Affiliation(s)
- Darja Lavogina
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411 Tartu, Estonia
| | - Katrin Kestav
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411 Tartu, Estonia
| | - Apirat Chaikuad
- Nuffield Department of Clinical Medicine, Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, England
| | - Christina Heroven
- Nuffield Department of Clinical Medicine, Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, England
| | - Stefan Knapp
- Nuffield Department of Clinical Medicine, Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, England
- Institute for Pharmaceutical Chemistry and Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-Universität, D-60438 Frankfurt am Main, Germany
| | - Asko Uri
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411 Tartu, Estonia
| |
Collapse
|