1
|
Li J, Ye C, Li H, Li J. Targeting the IKZF1/BCL-2 axis as a novel therapeutic strategy for treating acute T-cell lymphoblastic leukemia. Cancer Biol Ther 2025; 26:2457777. [PMID: 39862423 PMCID: PMC11776473 DOI: 10.1080/15384047.2025.2457777] [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: 09/25/2024] [Revised: 01/06/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025] Open
Abstract
OBJECTIVES Acute T-cell lymphoblastic leukemia (T-ALL) is a severe hematologic malignancy with limited treatment options and poor long-term survival. This study explores the role of IKZF1 in regulating BCL-2 expression in T-ALL. METHODS CUT&Tag and CUT&Run assays were employed to assess IKZF1 binding to the BCL-2 promoter. IKZF1 overexpression and knockdown experiments were performed in T-ALL cell lines. The effects of CX-4945 and venetoclax, alone and in combination, were evaluated in vitro and in vivo T-ALL models. RESULTS CUT&Tag sequencing identified IKZF1 binding to the BCL-2 promoter, establishing it as a transcriptional repressor. Functional assays demonstrated that IKZF1 overexpression reduced BCL-2 mRNA levels and increased repressive histone marks at the BCL-2 promoter, while IKZF1 knockdown led to elevated BCL-2 expression. CX-4945, a CK2 inhibitor, could reduced BCL-2 levels in T-ALL cells. Notably, knockdown of IKZF1 partially rescued the CX-4945-induced repression of BCL-2. These results underscore the CK2-IKZF1 signaling axis as a key regulator of BCL-2 expression. In vitro, CX-4945 enhanced the cytotoxicity of venetoclax, with the combination showing significant synergistic effects and increased apoptosis in T-ALL cell lines. In vivo studies with cell line-derived xenograft (CDX) and patient-derived xenograft (PDX) models demonstrated that CX-4945 and venetoclax combined therapy provided superior therapeutic efficacy, reducing tumor burden and prolonging survival compared to single-agent treatments. CONCLUSIONS IKZF1 represses BCL-2 in T-ALL, and targeting the CK2-IKZF1 axis with CX-4945 and venetoclax offers a promising therapeutic strategy, showing enhanced efficacy and potential as a novel treatment approach for T-ALL.
Collapse
Affiliation(s)
- Juan Li
- Department of Hematology, Taixing People’s Hospital Affiliated to Yangzhou University, Taixing, China
- Institute of Hematology, Affiliated hospital of Yangzhou University, Taixing, China
| | - Chunmei Ye
- Department of Hematology, Taixing People’s Hospital Affiliated to Yangzhou University, Taixing, China
- Institute of Hematology, Affiliated hospital of Yangzhou University, Taixing, China
| | - Hui Li
- Department of Hematology, Taixing People’s Hospital Affiliated to Yangzhou University, Taixing, China
| | - Jun Li
- Department of Hematology, Taixing People’s Hospital Affiliated to Yangzhou University, Taixing, China
- Institute of Hematology, Affiliated hospital of Yangzhou University, Taixing, China
| |
Collapse
|
2
|
Scuruchi M, Speranza D, Bruschetta G, Vaccaro F, Galeano M, Pallio G, Vaccaro M, Borgia F, Li Pomi F, Collino M, Irrera N. Protein Kinase CK2 Inhibition Represents a Pharmacological Chance for the Treatment of Skin Diseases. Int J Mol Sci 2025; 26:5404. [PMID: 40508214 PMCID: PMC12155107 DOI: 10.3390/ijms26115404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2025] [Revised: 05/30/2025] [Accepted: 06/03/2025] [Indexed: 06/16/2025] Open
Abstract
Protein kinase CK2 has emerged as a pivotal regulator of cellular processes involved in skin homeostasis, including cell proliferation, differentiation and inflammatory response regulation. In fact, CK2 activity dysregulation is implicated in the pathogenesis of different skin diseases, such as psoriasis, cancer and inflammatory dermatoses. CK2 overactivation fosters keratinocyte proliferation and pro-inflammatory cytokine production through the STAT3 and Akt pathways in psoriasis, thus contributing to epidermal hyperplasia and inflammation. In the realm of oncology, CK2 overexpression correlates with tumor progression, facilitating cell survival and metastasis in melanoma and non-melanoma skin cancers. Pharmacological inhibition of CK2 has demonstrated therapeutic potential, with CX-4945 (Silmitasertib) as the most studied adenosine triphosphate-competitive inhibitor (ATP-competitive inhibitor). Preclinical models reveal that CK2 inhibitors effectively mitigate pathological features of psoriasis, regulate keratinocyte differentiation, and suppress tumor growth in skin cancers. These inhibitors also potentiate the efficacy of conventional chemotherapeutics and exhibit anti-inflammatory effects in dermatological conditions. Future research will aim to enhance the specificity and delivery of CK2-targeting therapies, including topical formulations, to minimize systemic side effects. Combination therapies integrating CK2 inhibitors with other agents might offer synergistic benefits in managing skin diseases. This review underscores CK2's critical role in skin and its therapeutic potential as a pharmacological target, advocating for innovative approaches to harness CK2 inhibition in dermatology.
Collapse
Affiliation(s)
- Michele Scuruchi
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy; (M.S.); (D.S.); (M.V.); (F.B.); (F.L.P.); (N.I.)
| | - Desirèe Speranza
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy; (M.S.); (D.S.); (M.V.); (F.B.); (F.L.P.); (N.I.)
| | - Giuseppe Bruschetta
- Department of Veterinary Sciences, University of Messina, Viale G. Palatucci, 98168 Messina, Italy;
| | - Federico Vaccaro
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, Via C. Valeria, 98125 Messina, Italy; (F.V.); (M.G.)
| | - Mariarosaria Galeano
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, Via C. Valeria, 98125 Messina, Italy; (F.V.); (M.G.)
| | - Giovanni Pallio
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, Via C. Valeria, 98125 Messina, Italy; (F.V.); (M.G.)
| | - Mario Vaccaro
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy; (M.S.); (D.S.); (M.V.); (F.B.); (F.L.P.); (N.I.)
| | - Francesco Borgia
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy; (M.S.); (D.S.); (M.V.); (F.B.); (F.L.P.); (N.I.)
| | - Federica Li Pomi
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy; (M.S.); (D.S.); (M.V.); (F.B.); (F.L.P.); (N.I.)
| | - Massimo Collino
- Department of Neurosciences “Rita Levi Montalcini”, University of Torino, Via Verdi, 10124 Torino, Italy;
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy; (M.S.); (D.S.); (M.V.); (F.B.); (F.L.P.); (N.I.)
| |
Collapse
|
3
|
Zhang H, Kong L, Li J, Liu Z, Zhao Y, Lv X, Wu L, Chai L, You H, Jin J, Cao X, Zheng Z, Liu Y, Yan Z, Jin X. SPOP mutations increase PARP inhibitor sensitivity via CK2/PIAS1/SPOP axis in prostate cancer. JCI Insight 2025; 10:e186871. [PMID: 40260915 PMCID: PMC12016936 DOI: 10.1172/jci.insight.186871] [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: 09/10/2024] [Accepted: 03/04/2025] [Indexed: 04/24/2025] Open
Abstract
It is well documented that impaired DNA damage repair (DDR) induces genomic instability that can efficiently increase the sensitivity of prostate cancer (PCa) cells to PARP inhibitors; however, the underlying mechanism remains elusive. Here, we found profound genomic instability in PCa cells with SPOP gene mutations and confirmed the sensitivity of SPOP-mutated PCa cells to olaparib-induced apoptosis. Mechanistically, we identified olaparib-induced CK2-mediated phosphorylation of PIAS1-S468, which in turn mediated SUMOylation of SPOP, thus promoting its E3 ligase activity in the DDR. Moreover, an abnormal CK2/PIAS1/SPOP axis due to SPOP mutations or defects in CK2-mediated phosphorylation of PIAS1, as well as SPOP inhibitor treatment, led to impaired DDR, thus increasing olaparib-induced apoptosis of PCa cells and enhancing olaparib sensitivity in animal models and patient-derived organoids. This suggested that disruption of the CK2/PIAS1/SPOP signaling axis could serve as an indicator for targeted therapy of PCa using a PARP inhibitor.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Lili Kong
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Jinhui Li
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Zhihan Liu
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yiting Zhao
- Department of Ultrasound Medicine, The Affiliated People’s Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xiuyi Lv
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Liangpei Wu
- Department of Ultrasound Medicine, The Affiliated People’s Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Lin Chai
- Department of Ultrasound Medicine, The Affiliated People’s Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Hongjie You
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jiabei Jin
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Xinyi Cao
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Zhong Zheng
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yadong Liu
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Zejun Yan
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xiaofeng Jin
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
4
|
Cruz-Gamero JM, Ballardin D, Lecis B, Zhang CL, Cobret L, Gast A, Morisset-Lopez S, Piskorowski R, Langui D, Jose J, Chevreux G, Rebholz H. Missense mutation in the activation segment of the kinase CK2 models Okur-Chung neurodevelopmental disorder and alters the hippocampal glutamatergic synapse. Mol Psychiatry 2025; 30:1497-1509. [PMID: 39367055 DOI: 10.1038/s41380-024-02762-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 09/14/2024] [Accepted: 09/23/2024] [Indexed: 10/06/2024]
Abstract
Exome sequencing has enabled the identification of causative genes of monogenic forms of autism, amongst them, in 2016, CSNK2A1, the gene encoding the catalytic subunit of the kinase CK2, linking this kinase to Okur-Chung Neurodevelopmental Syndrome (OCNDS), a newly described neurodevelopmental condition with many symptoms resembling those of autism spectrum disorder. Thus far, no preclinical model of this condition exists. Here we describe a knock-in mouse model that harbors the K198R mutation in the activation segment of the α subunit of CK2. This region is a mutational hotspot, representing one-third of patients. These mice exhibit behavioral phenotypes that mirror patient symptoms. Homozygous knock-in mice die mid-gestation while heterozygous knock-in mice are born at half of the expected mendelian ratio and are smaller in weight and size than wildtype littermates. Heterozygous knock-in mice showed alterations in cognition and memory-assessing paradigms, enhanced stereotypies, altered circadian activity patterns, and nesting behavior. Phosphoproteome analysis from brain tissue revealed alterations in the phosphorylation status of major pre- and postsynaptic proteins of heterozygous knock-in mice. In congruence, we detect reduced synaptic maturation in hippocampal neurons and attenuated long-term potentiation in the hippocampus of knock-in mice. Taken together, heterozygous knock-in mice (CK2αK198R/+) exhibit significant face validity, presenting ASD-relevant phenotypes, synaptic deficits, and alterations in synaptic plasticity, all of which strongly validate this line as a mouse model of OCNDS.
Collapse
Affiliation(s)
- Jose M Cruz-Gamero
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Laboratory of Signaling mechanisms in neurological disorders, 75014, Paris, France
| | - Demetra Ballardin
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Laboratory of Signaling mechanisms in neurological disorders, 75014, Paris, France
| | - Barbara Lecis
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Laboratory of Signaling mechanisms in neurological disorders, 75014, Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
| | - Chun-Lei Zhang
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Laboratory of Signaling mechanisms in neurological disorders, 75014, Paris, France
| | - Laetitia Cobret
- Center for Molecular Biophysics-CNRS UPR 4301, Rue Charles Sadron, Orléans, France
| | - Alexander Gast
- Institute of Pharmaceutical and Medicinal Chemistry, Pharmacampus, University of Münster, Münster, Germany
| | | | - Rebecca Piskorowski
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Laboratory of Signaling mechanisms in neurological disorders, 75014, Paris, France
| | - Dominique Langui
- Inserm, Institut du Cerveau, Plateforme ICM-Quant, Paris, France
| | - Joachim Jose
- Institute of Pharmaceutical and Medicinal Chemistry, Pharmacampus, University of Münster, Münster, Germany
| | | | - Heike Rebholz
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Laboratory of Signaling mechanisms in neurological disorders, 75014, Paris, France.
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France.
- Center of Neurodegeneration, Faculty of Medicine, Danube Private University, Krems, Austria.
| |
Collapse
|
5
|
Xu DM, Chen LX, Han H, Mo M. Single-cell and spatial transcriptomics reveal pre-metastatic subsets and therapeutic targets in penile carcinoma. iScience 2025; 28:111765. [PMID: 39925432 PMCID: PMC11804784 DOI: 10.1016/j.isci.2025.111765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/28/2024] [Accepted: 01/06/2025] [Indexed: 02/11/2025] Open
Abstract
Tumor heterogeneity, driven by branching evolution and genomic mutations, complicates cancer treatment. Understanding malignant cell evolution across various tumors aids in identifying pre-metastatic subpopulations for optimized therapies. Using bulk RNA sequencing (6 primary penile carcinomas, 6 metastatic lymph nodes, GSE196978), single-cell RNA sequencing (4 advanced penile carcinomas), spatial transcriptomics (Squamous cell carcinoma [SCC]: GSE144239-GSM4565823 and SCC: GSE144239-GSM4565826), and cell assays with Silmitasertib, we mapped heterogeneity and pinpointed therapeutic targets. In penile carcinoma, we discovered an MMP3+SPP1+ pre-metastatic subset and casein kinase 2 alpha 1 (CK2α) overexpression. The nuclear factor κB (NF-κB) pathway may drive metastasis. Pan-cancer analysis showed that MMP3 and SPP1 link to epithelial mesenchymal transition (EMT) and drug resistance, while CK2α activates oncogenes. Silmitasertib, a CK2α inhibitor, exhibited anti-tumor effects in penile carcinoma cells. Validated across 98 single-cell and 6 spatial datasets, our study advances the understanding of tumorigenesis and metastasis, highlighting Silmitasertib as a potential therapeutic agent.
Collapse
Affiliation(s)
- Da-Ming Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Ling-Xiao Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| | - Hui Han
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Miao Mo
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| |
Collapse
|
6
|
Khalifa H, ElHady AK, Liu T, Elgaher WAM, Filhol-Cochet O, Cochet C, Abadi AH, Hamed MM, Abdel-Halim M, Engel M. Discovery of a novel, selective CK2 inhibitor class with an unusual basic scaffold. Eur J Med Chem 2025; 282:117048. [PMID: 39566243 DOI: 10.1016/j.ejmech.2024.117048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
CK2 is a Ser/Thr-protein kinase playing a crucial role in promoting cell growth and survival, hence it is considered a promising target for anti-cancer drugs. However, many previously reported CK2 inhibitors lack selectivity. In search of novel scaffolds for selective CK2 inhibition, we identified a dihydropyrido-thieno[2,3-d]pyrimidine derivative displaying submicromolar inhibitory activity against CK2α. This scaffold captured our interest because of the basic secondary amine, a rather unusual motif for CK2 inhibitors. Our optimization strategy comprised the incorporation of a 4-piperazinyl moiety as a linker group and introduction of varying substituents on the pendant phenyl ring. All resulting compounds exhibited potent CK2α inhibition, with IC50 values in the nanomolar range. Compound 10b demonstrated the most balanced activity profile with a cell-free IC50 value of 36.7 nM and a notable cellular activity with a GI50 of 7.3 μM and 7.5 μM against 786-O renal cell carcinoma and U937 lymphoma cells, respectively. 10b displayed excellent selectivity when screened against a challenging kinase selectivity profiling panel. Moreover, 10b inhibited CK2 in the cells, albeit less potently than CX-4945, but induced cell death more strongly than CX-4945. Altogether, we have identified a novel CK2 inhibitory scaffold with drug-like physicochemical properties in a favorable basic pKa range.
Collapse
Affiliation(s)
- Hend Khalifa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt
| | - Ahmed K ElHady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt; School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, New Administrative Capital, Cairo, Egypt
| | - Ting Liu
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, 66123, Saarbrücken, Germany
| | - Walid A M Elgaher
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123, Saarbrücken, Germany
| | - Odile Filhol-Cochet
- University Grenoble Alpes, INSERM 1292, CEA, UMR Biosante, 38000, Grenoble, France
| | - Claude Cochet
- University Grenoble Alpes, INSERM 1292, CEA, UMR Biosante, 38000, Grenoble, France
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt
| | - Mostafa M Hamed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123, Saarbrücken, Germany
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt.
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, 66123, Saarbrücken, Germany.
| |
Collapse
|
7
|
George IA, Sambath J, Dhawale RE, Singh M, Trivedi V, Venkataramanan R, Chauhan R, Kumar P. Phosphoproteomics guides low dose drug combination of cisplatin and silmitasertib against concurrent chemoradiation resistant cervical cancer. Mol Omics 2025; 21:87-100. [PMID: 39665434 DOI: 10.1039/d4mo00147h] [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: 12/13/2024]
Abstract
Cisplatin-based concurrent chemoradiotherapy (CCRT) is the standard treatment for cervical patients with locally advanced disease. Despite the improved survival rates and prognosis observed in patients undergoing CCRT, over 30-40% do not achieve complete response and are at risk of locoregional recurrence. Targeting crucial molecules that confer resistance may improve the clinical outcomes of the treatment resistant patient cohort. Herein, we employed a liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based phosphoproteomic approach to identify the altered phosphophorylation events, activated kinases and dysregulated pathways involved in treatment resistance. We quantified 2531 unique phosphopeptides mapping to 1099 proteins of which 74 proteins were differentially phosphorylated between the cohorts. Pathway analysis revealed dysregulation of the DNA repair pathway and the proteins involved in DNA repair in the non-responder cohort. Additionally, we identified kinase signature associated with CCRT resistance. Kinases such as CSNK2A1, PRKDC, PLK-1, NEK2, ATM and CDK1 are predicted to be activated in non-responders. In particular, we showed that CSNK2A1 is involved in oncogenesis of cervical cancer and pharmacological inhibition led to reduced cell proliferation, migration and colony formation. Moreover, the combination of the CSNK2A1 inhibitor, silmitasertib with cisplatin demonstrated synergism (combination index < 1) and yielded a beneficial reduction in dosage. The dose reduced combination potentially reduced the proliferative, migratory and colony formation ability in vitro. Our findings highlight the potential of phosphoproteomics to identify clinically significant targets and pathways implicated in CCRT resistance. Our study also indicates that combination therapy could serve as an effective treatment strategy to improve the efficacy of patients undergoing CCRT.
Collapse
Affiliation(s)
- Irene A George
- Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India
- Institute of Bioinformatics, Bangalore, 560066, Karnataka, India.
| | - Janani Sambath
- Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India
- Institute of Bioinformatics, Bangalore, 560066, Karnataka, India.
| | - R E Dhawale
- Vedantaa Hospital & Research Centre, Palghar, 401606, Maharashtra, India
| | - Manisha Singh
- Mahavir Cancer Sansthan and Research Centre (MCSRC), Patna, 801505, Bihar, India.
| | - Vinita Trivedi
- Mahavir Cancer Sansthan and Research Centre (MCSRC), Patna, 801505, Bihar, India.
| | - R Venkataramanan
- Karkinos Foundation, Mumbai, 400086, Maharashtra, India
- Karkinos Healthcare Pvt Ltd, Navi Mumbai, 400705, Maharashtra, India
| | - Richa Chauhan
- Mahavir Cancer Sansthan and Research Centre (MCSRC), Patna, 801505, Bihar, India.
| | - Prashant Kumar
- Manipal Academy of Higher Education (MAHE), Manipal, 576104, Karnataka, India
- Institute of Bioinformatics, Bangalore, 560066, Karnataka, India.
- Karkinos Foundation, Mumbai, 400086, Maharashtra, India
- Karkinos Healthcare Pvt Ltd, Navi Mumbai, 400705, Maharashtra, India
- Datar Cancer Genetics, Nashik, 422010, Maharashtra, India
- Centre of Excellence for Cancer - Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, 208016, Uttar Pradesh, India
| |
Collapse
|
8
|
Haga Y, Ray R, Ray RB. Silmitasertib in Combination With Cabozantinib Impairs Liver Cancer Cell Cycle Progression, Induces Apoptosis, and Delays Tumor Growth in a Preclinical Model. Mol Carcinog 2025; 64:72-82. [PMID: 39377735 DOI: 10.1002/mc.23827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/09/2024]
Abstract
The rising incidence of hepatocellular carcinoma (HCC) is a global problem. Several approved treatments, including immune therapy and multi-tyrosine kinase inhibitors, are used for treatment, although the results are not optimum. There is an unmet need to develop highly effective chemotherapies for HCC. Targeting multiple pathways to attack cancer cells is beneficial. Cabozantinib is an orally available bioactive multikinase inhibitor and has a modest effect on HCC treatment. Silmitasertib is an orally bioavailable, potent CK2 inhibitor with a direct role in DNA damage repair and is in clinical trials for other cancers. In this study, we planned to repurpose these existing drugs on HCC treatment. We observed a stronger antiproliferative effect of these two combined drugs on HCC cells generated from different etiologies as compared to the single treatment. Global RNA-seq analyses revealed a decrease in the expression of G2/M cell cycle transition genes in HCC cells following combination treatment, suggesting G2 phase cell arrest. We observed G2/M cell cycle phase arrest in HCC cells upon combination treatment compared to the single-treated or vehicle-treated control cells. The downregulation of CCNA2 and CDC25C following combination therapy further supported the observation. Subsequent analyses demonstrated that combination treatment inhibited 70 kDa ribosomal protein S6 kinase (p70S6K) phosphorylation, and increased Bim expression. Apoptosis of HCC cells were accompanied by increased poly (ADP-ribose) polymerase cleavage and caspase-9 activation. Next, we observed that a combination therapy significantly delayed the progression of HCC xenograft growth as compared to vehicle control. Together, our results suggested combining cabozantinib and silmitasertib would be a promising treatment option for HCC.
Collapse
Affiliation(s)
- Yuki Haga
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Ranjit Ray
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri, USA
- Department of Molecular Microbiology & Immunology, Saint Louis University, St. Louis, Missouri, USA
| | - Ratna B Ray
- Department of Molecular Microbiology & Immunology, Saint Louis University, St. Louis, Missouri, USA
- Department of Pathology, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|
9
|
Kröger L, Borgert S, Lauwers M, Steinkrüger M, Jose J, Pietsch M, Wünsch B. Structure-Activity Relationship Studies of Tetracyclic Pyrrolocarbazoles Inhibiting Heterotetrameric Protein Kinase CK2. Molecules 2024; 30:63. [PMID: 39795120 PMCID: PMC11722180 DOI: 10.3390/molecules30010063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
The serine/threonine kinase CK2 (formerly known as casein kinase II) plays a crucial role in various CNS disorders and is highly expressed in various types of cancer. Therefore, inhibiting this key kinase could be promising for the treatment of these diseases. The CK2 holoenzyme is formed by the recruitment of two catalytically active CK2α and/or CK2α' subunits by a regulatory CK2β dimer. Starting with the lead furocarbazole W16 (4) inhibiting the CK2α/CK2β interaction, analogous pyrrolocarbazoles were prepared and tested for their protein-protein interaction inhibition (PPII). The key step of the synthesis was a multicomponent Levy reaction of 2-(indolyl)acetate 6, benzaldehydes 7, and N-substituted maleimides 8. Targeted modifications were performed by the saponification of the tetracyclic ester 9a, followed by the coupling of the resulting acid 10 with diverse amines. The replacement of the O-atom of the lead furocarbazole 4 by an N-atom in pyrrolocarbazoles retained or even increased the inhibition of the CK2α/CK2β interaction. The large benzyloxazolidinyl moiety of 4 could be replaced by smaller N-substituents without the loss of the PPII. The introduction of larger substituents at the 2-position and/or at p-position of the phenyl moiety at the 10-position to increase the surface for the inhibition of the PPI did not enhance the inhibition of the CK2α/CK2β association. The strong inhibition of the CK2α/CK2β association by the histidine derivative (+)-20a (Ki = 6.1 µM) translated into a high inhibition of the kinase activity of the CK2 holoenzyme (CK2α2β2, IC50 = 2.5 µM). Thus, 20a represents a novel lead compound inhibiting CK2 via the inhibition of the association of the CK2α and Ck2β subunits.
Collapse
Affiliation(s)
- Lukas Kröger
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, D-48149 Münster, Germany; (L.K.); (S.B.); (J.J.)
| | - Sebastian Borgert
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, D-48149 Münster, Germany; (L.K.); (S.B.); (J.J.)
| | - Miriam Lauwers
- Institutes I & II of Pharmacology, Center of Pharmacology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Gleueler Straße 24, D-50931 Cologne, Germany; (M.L.); (M.S.); (M.P.)
| | - Michaela Steinkrüger
- Institutes I & II of Pharmacology, Center of Pharmacology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Gleueler Straße 24, D-50931 Cologne, Germany; (M.L.); (M.S.); (M.P.)
| | - Joachim Jose
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, D-48149 Münster, Germany; (L.K.); (S.B.); (J.J.)
| | - Markus Pietsch
- Institutes I & II of Pharmacology, Center of Pharmacology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Gleueler Straße 24, D-50931 Cologne, Germany; (M.L.); (M.S.); (M.P.)
- Faculty of Applied Natural Sciences, TH Köln-University of Applied Sciences, Campus Leverkusen, Campusplatz 1, D-51379 Leverkusen, Germany
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, D-48149 Münster, Germany; (L.K.); (S.B.); (J.J.)
| |
Collapse
|
10
|
Rumler H, Schmithals C, Werner C, Bollacke A, Aichele D, Götz C, Niefind K, Wünsch B, Jose J. Discovery of 7,9-Dibromo-dihydrodibenzofuran as a Potent Casein Kinase 2 (CK2) Inhibitor: Synthesis, Biological Evaluation, and Structural Studies on E-/ Z-Isomers. ACS Pharmacol Transl Sci 2024; 7:3846-3866. [PMID: 39698287 PMCID: PMC11651316 DOI: 10.1021/acsptsci.4c00426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024]
Abstract
The human protein kinase CK2 is a promising target for cancer treatment. Only two CK2 inhibitors have reached clinical trials until today. Among others, a dibenzofuran scaffold has emerged as highly prospective for the development of new CK2 inhibitors. Thirty-three newly synthesized dibenzofuran-based compounds were tested on their inhibitory potential in vitro. 7,9-Dichloro-8-hydroxy-4-[(phenylamino)methylene]-1,2-dihydro-dibenzo[b,d]furan-3(4H)-one (12b) and 7,9-dibromo-8-hydroxy-4-[(phenylamino)methylene]-1,2-dihydro-dibenzo[b,d]furan-3(4H)-one (12c) showed the lowest IC50 values with 5.8 nM for both. The dibenzofuran-based CK2 inhibitors crossed the cell membrane of LNCaP human prostate carcinoma cells and reduced intracellular CK2 activity. Among 70 kinases from different representative subgroups of the human kinome, CK2 was most strongly inhibited by compound 12c. Co-crystallization of 12c together with CK2α indicated a π-halogen bond of the bromine at position C9 with the gatekeeper amino acid Phe113. CK2α could bind both the E- and Z-isomers of 12c. Our results provide new insights into the structure-activity relationships of dibenzofuran derivatives.
Collapse
Affiliation(s)
- Hendrik Rumler
- University
of Münster, Institute of Pharmaceutical and Medicinal Chemistry,
Pharmacampus, Münster 48149, Germany
| | - Claudia Schmithals
- University
of Münster, Institute of Pharmaceutical and Medicinal Chemistry,
Pharmacampus, Münster 48149, Germany
| | - Christian Werner
- Institute
of Biochemistry, University of Cologne, Cologne 50674, Germany
| | - Andre Bollacke
- University
of Münster, Institute of Pharmaceutical and Medicinal Chemistry,
Pharmacampus, Münster 48149, Germany
| | - Dagmar Aichele
- University
of Münster, Institute of Pharmaceutical and Medicinal Chemistry,
Pharmacampus, Münster 48149, Germany
| | - Claudia Götz
- Medical Biochemistry
and Molecular Biology, Saarland University, Homburg 66421, Germany
| | - Karsten Niefind
- Institute
of Biochemistry, University of Cologne, Cologne 50674, Germany
| | - Bernhard Wünsch
- University
of Münster, Institute of Pharmaceutical and Medicinal Chemistry,
Pharmacampus, Münster 48149, Germany
| | - Joachim Jose
- University
of Münster, Institute of Pharmaceutical and Medicinal Chemistry,
Pharmacampus, Münster 48149, Germany
| |
Collapse
|
11
|
Jin Q, Harris E, Myers JA, Mehmood R, Cotton A, Shirnekhi HK, Baggett DW, Wen JQ, Schild AB, Bhansali RS, Klein J, Narina S, Pieters T, Yoshimi A, Pruett-Miller SM, Kriwacki R, Abdel-Wahab O, Malinge S, Ntziachristos P, Obeng EA, Crispino JD. Disruption of cotranscriptional splicing suggests RBM39 is a therapeutic target in acute lymphoblastic leukemia. Blood 2024; 144:2417-2431. [PMID: 39316649 PMCID: PMC11628860 DOI: 10.1182/blood.2024024281] [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: 02/12/2024] [Revised: 08/19/2024] [Accepted: 08/31/2024] [Indexed: 09/26/2024] Open
Abstract
ABSTRACT There are only a few options for patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL), thus, this is a major area of unmet medical need. In this study, we reveal that the inclusion of a poison exon in RBM39, which could be induced by both CDK9 or CDK9 independent cyclin-dependent kinases, mitogen-activated protein kinases, glycogen synthase kinases, CDC-like kinases (CMGC) kinase inhibition, is recognized by the nonsense-mediated messenger RNA decay pathway for degradation. Targeting this poison exon in RBM39 with CMGC inhibitors led to protein downregulation and the inhibition of ALL growth, particularly in relapsed/refractory B-ALL. Mechanistically, disruption of cotranscriptional splicing by the inhibition of CMGC kinases, including DYRK1A, or inhibition of CDK9, which phosphorylate the C-terminal domain of RNA polymerase II (Pol II), led to alteration in the SF3B1 and Pol II association. Disruption of SF3B1 and the transcriptional elongation complex altered Pol II pausing, which promoted the inclusion of a poison exon in RBM39. Moreover, RBM39 ablation suppressed the growth of human B-ALL, and targeting RBM39 with sulfonamides, which degrade RBM39 protein, showed strong antitumor activity in preclinical models. Our data reveal that relapsed/refractory B-ALL is susceptible to pharmacologic and genetic inhibition of RBM39 and provide 2 potential strategies to target this axis.
Collapse
Affiliation(s)
- Qi Jin
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Ethan Harris
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Jacquelyn A. Myers
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Rashid Mehmood
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Anitria Cotton
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Hazheen K. Shirnekhi
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - David W. Baggett
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jeremy Qiang Wen
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Andrew B. Schild
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Rahul S. Bhansali
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jonathon Klein
- Center for Advanced Genome Engineering and the Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Shilpa Narina
- Center for Advanced Genome Engineering and the Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Tim Pieters
- Department of Biomolecular Medicine, Center for Medical Genetics and Cancer Research Institute, Ghent University, Ghent, Belgium
| | - Akihide Yoshimi
- Division of Cancer RNA Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Shondra M. Pruett-Miller
- Center for Advanced Genome Engineering and the Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Richard Kriwacki
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Omar Abdel-Wahab
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sebastien Malinge
- Translational Genomics in Leukemia, Cancer Centre, The Kids Research Institute Australia, Nedlands, WA, Australia
| | - Panagiotis Ntziachristos
- Department of Biomolecular Medicine, Center for Medical Genetics and Cancer Research Institute, Ghent University, Ghent, Belgium
| | - Esther A. Obeng
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - John D. Crispino
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
12
|
Bova V, Mannino D, Salako AE, Esposito E, Filippone A, Scuderi SA. Casein Kinase 2 Inhibitor, CX-4945, Induces Apoptosis and Restores Blood-Brain Barrier Homeostasis in In Vitro and In Vivo Models of Glioblastoma. Cancers (Basel) 2024; 16:3936. [PMID: 39682125 DOI: 10.3390/cancers16233936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/21/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Background: In oncology, casein kinase 2 (CK2), a serine/threonine kinase, has a dual action, regulating cellular processes and acting as an oncogenic promoter. Methods: This study examined the effect of CX-4945, a selective CK2 inhibitor, in a human U-87 glioblastoma (GBM) cell line, treated with CX-4945 (5, 10, and 15 μM) for 24 h. Similarly, the hCMEC/D3 cell line was used to mimic the blood-brain barrier (BBB), examining the ability of CX-4945 to restore BBB homeostasis, after stimulation with lipopolysaccharide (LPS) and then treated with CX-4945 (5, 10, and 15 μM). Results: We reported that CX-4945 reduced the proliferative activity and modulated the main pathways involved in tumor progression including apoptosis. Furthermore, in confirmation of the in vitro study, performing a xenograft model, we demonstrated that CX-4945 exerted promising antiproliferative effects, also restoring the tight junctions' expression. Conclusions: These new insights into the molecular signaling of CK2 in GBM and BBB demonstrate that CX-4945 could be a promising approach for future GBM therapy, not only in the tumor microenvironment but also at the BBB level.
Collapse
Affiliation(s)
- Valentina Bova
- Department of Chemical, Biological, Pharmaceutical, Environmental Science, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98166 Messina, Italy
| | - Deborah Mannino
- Department of Chemical, Biological, Pharmaceutical, Environmental Science, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98166 Messina, Italy
| | - Ayomide E Salako
- Department of Chemical, Biological, Pharmaceutical, Environmental Science, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98166 Messina, Italy
- Department of Statistics, Computer Science, Applications (DiSIA), University of Florence, 50121 Firenze, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical, Environmental Science, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98166 Messina, Italy
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmaceutical, Environmental Science, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98166 Messina, Italy
| | - Sarah A Scuderi
- Department of Chemical, Biological, Pharmaceutical, Environmental Science, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98166 Messina, Italy
| |
Collapse
|
13
|
Yi Y, Dai L, Lan Y, Tan C, Vázquez-Blomquist DM, Zeng G, Jiang D, Yang K, Perea SE, Perera Y. CIGB-300 internalizes and impairs viability of NSCLC cells lacking actionable targets by inhibiting casein kinase-2 signaling. Sci Rep 2024; 14:26038. [PMID: 39472715 PMCID: PMC11522547 DOI: 10.1038/s41598-024-75990-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/09/2024] [Indexed: 11/02/2024] Open
Abstract
Overall response rates in advanced Non-Small Cell Lung Cancer (NSCLC) remains low. Thus, novel molecular targets, tailored drugs and/or drug combinations are needed. Casein Kinase-2 (CK2) is a constitutively active and frequently over-expressed enzyme which fosters tumor survival, proliferation and metastasis. By using a clinical-grade and Cell Penetrating Peptide-based inhibitor coined as CIGB-300, we explore the anti-neoplastic effects caused by interruption of CK2 signaling in lung cancer cells lacking EGFR, ALK and ROS mutations. CIGB-300 penetrated and impaired viability and proliferation of Lung Adenocarcinoma (LUAD) (A549, NCI-H522) and Lung Squamous Carcinoma (LUSC) (NCI-H226 and SK-MES-1) cells in a dose-response manner. The differential activity could not be explained by overall peptide uptake or its subcellular distribution, as evidenced by flow cytometry and confocal microscopy. Upon internalization, CIGB-300 interacted with CK2 catalytic subunits (ɑ1/ɑ2) and CK2 substrates, thus impairing phosphorylation of enzyme substrates (CDC37s13, NPM1s125) and downstream proteins (RPS6s325/326). CK2 inhibition induced an early Reactive Oxygen Species (ROS) and mitochondrial membrane depolarization, which predates lung cancer cell death. Finally, intravenous injection of CIGB-300 in a cell line-based xenograft corroborated CIGB-300's anti-tumor effects and suggested concurrent in situ reductions of CSNK2ɑ subunit and downstream RPS6s235/236 phosphorylation. Overall, CIGB-300 therapeutic hypothesis and antineoplastic effects demonstrated herein, further support the evaluation of this clinical-grade CK2 inhibitor in advanced NSCLC with limited therapeutic options.
Collapse
Affiliation(s)
- Ying Yi
- China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Yangjiaqiao Street, Lengshuitan District, Yongzhou City, 425000, Hunan Province, People's Republic of China
| | - Lingfeng Dai
- China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Yangjiaqiao Street, Lengshuitan District, Yongzhou City, 425000, Hunan Province, People's Republic of China
| | - Yaqin Lan
- China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Yangjiaqiao Street, Lengshuitan District, Yongzhou City, 425000, Hunan Province, People's Republic of China
| | - Changyuan Tan
- China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Yangjiaqiao Street, Lengshuitan District, Yongzhou City, 425000, Hunan Province, People's Republic of China
| | - Dania M Vázquez-Blomquist
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, Center for Genetic Engineering & Biotechnology (CIGB), Havana, 10600, Cuba
| | - Guirong Zeng
- Hunan Pharmaceutical Medicines Research Center (HPMRC), Kangtian Road No. 123, National Biological Industry Base, Changsha, Hunan Province, People's Republic of China
| | - Dejian Jiang
- Hunan Pharmaceutical Medicines Research Center (HPMRC), Kangtian Road No. 123, National Biological Industry Base, Changsha, Hunan Province, People's Republic of China
| | - Ke Yang
- China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Yangjiaqiao Street, Lengshuitan District, Yongzhou City, 425000, Hunan Province, People's Republic of China.
| | - Silvio E Perea
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, Center for Genetic Engineering & Biotechnology (CIGB), Havana, 10600, Cuba.
| | - Yasser Perera
- China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Development and Construction Investment Co., Ltd. (YDCI), Yangjiaqiao Street, Lengshuitan District, Yongzhou City, 425000, Hunan Province, People's Republic of China.
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, Center for Genetic Engineering & Biotechnology (CIGB), Havana, 10600, Cuba.
| |
Collapse
|
14
|
Ghani MU, Shi J, Du Y, Zhong L, Cui H. A comprehensive review on the dynamics of protein kinase CK2 in cancer development and optimizing therapeutic strategies. Int J Biol Macromol 2024; 280:135814. [PMID: 39306165 DOI: 10.1016/j.ijbiomac.2024.135814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 10/13/2024]
Abstract
Protein kinase 2 (CK2) is an enzyme ubiquitously present and exhibits extensive kinase activity. It has been strongly linked to tumor progression through the abnormal phosphorylation of key proteins. Research has consistently demonstrated that CK2 is deregulated in various cancer types, with enhanced protein expression and nuclear distribution in tumor cells. CK2 plays a crucial role in a complex network that promotes cell infiltration, migration, proliferation, apoptosis, and cancer progression through multiple pathways, including PI3K/AKT, JAK2/STAT3, ATF4/CDKN1, and HSP90/Cdc37. In addition to its role in cancer growth, there is mounting evidence that CK2 may also affect the immunological dynamics of cancer by altering immune cell functions within the tumor microenvironment, thus facilitating tumor immune evasion. Recent research has increasingly focused on CK2, recognizing it as a therapeutic objective for oncological interventions. This review will critically examine the structure and signaling pathways of CK2, highlighting the significance of further research aimed at enhancing our understanding of the CK2 machinery. Finally, we conclude by refining therapeutic options, notably transitioning from non-pharmacological techniques to strategic CK2 inhibitor use. This development shortens the path to the desired outcome, establishing a pioneering standard in cancer therapy.
Collapse
Affiliation(s)
- Muhammad Usman Ghani
- Medical Research Institute, Southwest University, Chongqing 400715, China; State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Junbo Shi
- Medical Research Institute, Southwest University, Chongqing 400715, China; State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Yi Du
- Medical Research Institute, Southwest University, Chongqing 400715, China; State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Liping Zhong
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Hongjuan Cui
- Medical Research Institute, Southwest University, Chongqing 400715, China; State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
15
|
Marada A, Walter C, Suhm T, Shankar S, Nandy A, Brummer T, Dhaouadi I, Vögtle FN, Meisinger C. DYRK1A signalling synchronizes the mitochondrial import pathways for metabolic rewiring. Nat Commun 2024; 15:5265. [PMID: 38902238 PMCID: PMC11189921 DOI: 10.1038/s41467-024-49611-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Mitochondria require an extensive proteome to maintain a variety of metabolic reactions, and changes in cellular demand depend on rapid adaptation of the mitochondrial protein composition. The TOM complex, the organellar entry gate for mitochondrial precursors in the outer membrane, is a target for cytosolic kinases to modulate protein influx. DYRK1A phosphorylation of the carrier import receptor TOM70 at Ser91 enables its efficient docking and thus transfer of precursor proteins to the TOM complex. Here, we probe TOM70 phosphorylation in molecular detail and find that TOM70 is not a CK2 target nor import receptor for MIC19 as previously suggested. Instead, we identify TOM20 as a MIC19 import receptor and show off-target inhibition of the DYRK1A-TOM70 axis with the clinically used CK2 inhibitor CX4945 which activates TOM20-dependent import pathways. Taken together, modulation of DYRK1A signalling adapts the central mitochondrial protein entry gate via synchronization of TOM70- and TOM20-dependent import pathways for metabolic rewiring. Thus, DYRK1A emerges as a cytosolic surveillance kinase to regulate and fine-tune mitochondrial protein biogenesis.
Collapse
Affiliation(s)
- Adinarayana Marada
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Corvin Walter
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Tamara Suhm
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Sahana Shankar
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Arpita Nandy
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Tilman Brummer
- Institute of Molecular Medicine, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany
- German Cancer Consortium DKTK Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ines Dhaouadi
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - F-Nora Vögtle
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, 69120, Heidelberg, Germany.
- Network Aging Research, Heidelberg University, 69120, Heidelberg, Germany.
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
| | - Chris Meisinger
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany.
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
16
|
Wang S, Li Y, Lin Y, Li J, Guo L, Wang H, Lin X, Liu Z, Zhang B, Liao Z, Zhang Z. Bioinformatics analysis and experimental verification of the cancer-promoting effect of DHODH in clear cell renal cell carcinoma. Sci Rep 2024; 14:11985. [PMID: 38796629 PMCID: PMC11127953 DOI: 10.1038/s41598-024-62738-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a malignant tumor of the urinary system. To explore the potential mechanisms of DHODH in ccRCC, we analyzed its molecular characteristics using public databases. TCGA pan-cancer dataset was used to analyze DHODH expression in different cancer types and TCGA ccRCC dataset was used to assess differential expression, prognosis correlation, immune infiltration, single-gene, and functional enrichment due to DHODH. The GSCALite and CellMiner databases were employed to explore drugs and perform molecular docking analysis with DHODH. Protein-protein interaction networks and ceRNA regulatory networks of DHODH were constructed using multiple databases. The effect of DHODH on ccRCC was confirmed in vitro. DHODH was highly expressed in ccRCC. Immune infiltration analysis revealed that DHODH may be involved in regulating the infiltration of immunosuppressive cells such as Tregs. Notably, DHODH influenced ccRCC progression by forming regulatory networks with molecules, such as hsa-miR-26b-5p and UMPS and significantly enhanced the malignant characteristics of ccRCC cells. Several drugs, such as lapatinib, silmitasertib, itraconazole, and dasatinib, were sensitive to DHODH expression and exhibited strong molecular binding with it. Thus, DHODH may promote ccRCC progression and is a candidate effective therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Songsong Wang
- Department of Urology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
- School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Yan Li
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Yilong Lin
- School of Medicine, Xiamen University, Xiamen, 361000, China
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Junting Li
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, China
| | - Lang Guo
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Haoyu Wang
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Xinyuan Lin
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, China
| | - Ziming Liu
- Department of Urology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China.
- School of Medicine, Xiamen University, Xiamen, 361000, China.
| | - Bingqi Zhang
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China.
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China.
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China.
| | - Zhengming Liao
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China.
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China.
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China.
| | - Zhongmin Zhang
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China.
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China.
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China.
| |
Collapse
|
17
|
Hoenigsperger H, Koepke L, Acharya D, Hunszinger V, Freisem D, Grenzner A, Wiese S, Kirchhoff F, Gack MU, Sparrer KM. CSNK2 suppresses autophagy by activating FLN-NHL-containing TRIM proteins. Autophagy 2024; 20:994-1014. [PMID: 37938186 PMCID: PMC11135829 DOI: 10.1080/15548627.2023.2281128] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 11/09/2023] Open
Abstract
Macroautophagy/autophagy is a tightly regulated cellular process integral to homeostasis and innate immunity. As such, dysregulation of autophagy is associated with cancer, neurodegenerative disorders, and infectious diseases. While numerous factors that promote autophagy have been characterized, the key mechanisms that prevent excessive autophagy are less well understood. Here, we identify CSNK2/CK2 (casein kinase 2) as a negative regulator of autophagy. Pharmacological inhibition of CSNK2 activity or siRNA-mediated depletion of CSNK2 increased basal autophagic flux in cell lines and primary human lung cells. Vice versa, ectopic expression of CSNK2 reduced autophagic flux. Mechanistically, CSNK2 interacted with the FLN (filamin)-NHL domain-containing tripartite motif (TRIM) family members TRIM2, TRIM3 and TRIM71. Our data show that recruitment of CSNK2 to the C-terminal NHL domain of TRIM3 lead to its robust phosphorylation at serine 661 by CSNK2. A phosphorylation-defective mutant of TRIM3 was unable to reduce autophagosome numbers indicating that phosphorylation by CSNK2 is required for TRIM-mediated autophagy inhibition. All three TRIMs facilitated inactivation of the ULK1-BECN1 autophagy initiation complex by facilitating ULK1 serine 757 phosphorylation. Inhibition of CSNK2 promoted autophagy upon influenza A virus (IAV) and measles virus (MeV) infection. In line with this, targeting of CSNK2 or depletion of TRIM2, TRIM3 or TRIM71 enhanced autophagy-dependent restriction of IAV, MeV and human immunodeficiency virus 1 (HIV-1). Thus, our results identify the CSNK2-TRIM2, -TRIM3, -TRIM71 axis as a key regulatory pathway that limits autophagy. Targeting this axis may allow for therapeutic induction of autophagy against viral infections and in diseases associated with dysregulated autophagy.Abbreviation: ATG5: autophagy related 5; BafA1: bafilomycin A1; BECN1: beclin 1; CCD: coiled-coil domain; CSNK2/CK2: casein kinase 2; CSNK2A1: casein kinase 2 alpha 1; CSNK2A2: casein kinase 2 alpha 2; CSNK2B: casein kinase 2 beta; FLN: filamin; HeLa GL: HeLa cells stably expressing eGFP-LC3B; HIV-1: human immunodeficiency virus 1; IAV: influenza A virus; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3; MeV: measles virus; MTOR: mechanistic target of rapamycin kinase; RING: really interesting new gene; SQSTM1/p62: sequestosome 1; TRIM: tripartite motif; ULK1: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Helene Hoenigsperger
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Baden-Wuerttemberg, Germany
| | - Lennart Koepke
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Baden-Wuerttemberg, Germany
| | - Dhiraj Acharya
- Florida Research and Innovation Center, Cleveland Clinic, Port St Lucie, Florida, USA
| | - Victoria Hunszinger
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Baden-Wuerttemberg, Germany
| | - Dennis Freisem
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Baden-Wuerttemberg, Germany
| | - Alexander Grenzner
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Baden-Wuerttemberg, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, Ulm University, Ulm, Baden-Wuerttemberg, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Baden-Wuerttemberg, Germany
| | - Michaela U. Gack
- Florida Research and Innovation Center, Cleveland Clinic, Port St Lucie, Florida, USA
| | - Konstantin M.J. Sparrer
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Baden-Wuerttemberg, Germany
| |
Collapse
|
18
|
Zhao D, Guo Y, Wei H, Jia X, Zhi Y, He G, Nie W, Huang L, Wang P, Laster KV, Liu Z, Wang J, Lee MH, Dong Z, Liu K. Multi-omics characterization of esophageal squamous cell carcinoma identifies molecular subtypes and therapeutic targets. JCI Insight 2024; 9:e171916. [PMID: 38652547 PMCID: PMC11141925 DOI: 10.1172/jci.insight.171916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the predominant form of esophageal cancer and is characterized by an unfavorable prognosis. To elucidate the distinct molecular alterations in ESCC and investigate therapeutic targets, we performed a comprehensive analysis of transcriptomics, proteomics, and phosphoproteomics data derived from 60 paired treatment-naive ESCC and adjacent nontumor tissue samples. Additionally, we conducted a correlation analysis to describe the regulatory relationship between transcriptomic and proteomic processes, revealing alterations in key metabolic pathways. Unsupervised clustering analysis of the proteomics data stratified patients with ESCC into 3 subtypes with different molecular characteristics and clinical outcomes. Notably, subtype III exhibited the worst prognosis and enrichment in proteins associated with malignant processes, including glycolysis and DNA repair pathways. Furthermore, translocase of inner mitochondrial membrane domain containing 1 (TIMMDC1) was validated as a potential prognostic molecule for ESCC. Moreover, integrated kinase-substrate network analysis using the phosphoproteome nominated candidate kinases as potential targets. In vitro and in vivo experiments further confirmed casein kinase II subunit α (CSNK2A1) as a potential kinase target for ESCC. These underlying data represent a valuable resource for researchers that may provide better insights into the biology and treatment of ESCC.
Collapse
Affiliation(s)
- Dengyun Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, Henan, China
| | - Yaping Guo
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
| | - Huifang Wei
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Xuechao Jia
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Yafei Zhi
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Guiliang He
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Wenna Nie
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Limeng Huang
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Penglei Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | | | - Zhicai Liu
- Linzhou Cancer Hospital, Anyang, Henan, China
| | - Jinwu Wang
- Linzhou Cancer Hospital, Anyang, Henan, China
| | - Mee-Hyun Lee
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- College of Korean Medicine, Dongshin University, Naju, Jeonnam, Republic of Korea
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, Henan, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, Henan, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
19
|
Rios SA, Oyervides S, Uribe D, Reyes AM, Fanniel V, Vazquez J, Keniry M. Emerging Therapies for Glioblastoma. Cancers (Basel) 2024; 16:1485. [PMID: 38672566 PMCID: PMC11048459 DOI: 10.3390/cancers16081485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Glioblastoma is most commonly a primary brain tumor and the utmost malignant one, with a survival rate of approximately 12-18 months. Glioblastoma is highly heterogeneous, demonstrating that different types of cells from the same tumor can manifest distinct gene expression patterns and biological behaviors. Conventional therapies such as temozolomide, radiation, and surgery have limitations. As of now, there is no cure for glioblastoma. Alternative treatment methods to eradicate glioblastoma are discussed in this review, including targeted therapies to PI3K, NFKβ, JAK-STAT, CK2, WNT, NOTCH, Hedgehog, and TGFβ pathways. The highly novel application of oncolytic viruses and nanomaterials in combating glioblastoma are also discussed. Despite scores of clinical trials for glioblastoma, the prognosis remains poor. Progress in breaching the blood-brain barrier with nanomaterials and novel avenues for targeted and combination treatments hold promise for the future development of efficacious glioblastoma therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Megan Keniry
- School of Integrative Biological and Chemical Sciences, College of Sciences, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA; (S.A.R.); (D.U.); (A.M.R.)
| |
Collapse
|
20
|
Zhang B, Liu H, Wu F, Ding Y, Wu J, Lu L, Bajpai AK, Sang M, Wang X. Identification of hub genes and potential molecular mechanisms related to drug sensitivity in acute myeloid leukemia based on machine learning. Front Pharmacol 2024; 15:1359832. [PMID: 38650628 PMCID: PMC11033397 DOI: 10.3389/fphar.2024.1359832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/21/2024] [Indexed: 04/25/2024] Open
Abstract
Background: Acute myeloid leukemia (AML) is the most common form of leukemia among adults and is characterized by uncontrolled proliferation and clonal expansion of hematopoietic cells. There has been a significant improvement in the treatment of younger patients, however, prognosis in the elderly AML patients remains poor. Methods: We used computational methods and machine learning (ML) techniques to identify and explore the differential high-risk genes (DHRGs) in AML. The DHRGs were explored through multiple in silico approaches including genomic and functional analysis, survival analysis, immune infiltration, miRNA co-expression and stemness features analyses to reveal their prognostic importance in AML. Furthermore, using different ML algorithms, prognostic models were constructed and validated using the DHRGs. At the end molecular docking studies were performed to identify potential drug candidates targeting the selected DHRGs. Results: We identified a total of 80 DHRGs by comparing the differentially expressed genes derived between AML patients and normal controls and high-risk AML genes identified by Cox regression. Genetic and epigenetic alteration analyses of the DHRGs revealed a significant association of their copy number variations and methylation status with overall survival (OS) of AML patients. Out of the 137 models constructed using different ML algorithms, the combination of Ridge and plsRcox maintained the highest mean C-index and was used to build the final model. When AML patients were classified into low- and high-risk groups based on DHRGs, the low-risk group had significantly longer OS in the AML training and validation cohorts. Furthermore, immune infiltration, miRNA coexpression, stemness feature and hallmark pathway analyses revealed significant differences in the prognosis of the low- and high-risk AML groups. Drug sensitivity and molecular docking studies revealed top 5 drugs, including carboplatin and austocystin-D that may significantly affect the DHRGs in AML. Conclusion: The findings from the current study identified a set of high-risk genes that may be used as prognostic and therapeutic markers for AML patients. In addition, significant use of the ML algorithms in constructing and validating the prognostic models in AML was demonstrated. Although our study used extensive bioinformatics and machine learning methods to identify the hub genes in AML, their experimental validations using knock-out/-in methods would strengthen our findings.
Collapse
Affiliation(s)
- Boyu Zhang
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Haiyan Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Fengxia Wu
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Yuhong Ding
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Jiarun Wu
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Lu Lu
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Akhilesh K. Bajpai
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Mengmeng Sang
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Xinfeng Wang
- Department of Hematology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
21
|
Hussain S, Guo Y, Huo Y, Shi J, Hou Y. Regulation of cancer progression by CK2: an emerging therapeutic target. Med Oncol 2024; 41:94. [PMID: 38526625 DOI: 10.1007/s12032-024-02316-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 01/28/2024] [Indexed: 03/27/2024]
Abstract
Casein kinase II (CK2) is an enzyme with pleiotropic kinase activity that catalyzes the phosphorylation of lots of substrates, including STAT3, p53, JAK2, PTEN, RELA, and AKT, leading to the regulation of diabetes, cardiovascular diseases, angiogenesis, and tumor progression. CK2 is observed to have high expression in multiple types of cancer, which is associated with poor prognosis. CK2 holds significant importance in the intricate network of pathways involved in promoting cell proliferation, invasion, migration, apoptosis, and tumor growth by multiple pathways such as JAK2/STAT3, PI3K/AKT, ATF4/p21, and HSP90/Cdc37. In addition to the regulation of cancer progression, increasing evidence suggests that CK2 could regulate tumor immune responses by affecting immune cell activity in the tumor microenvironment resulting in the promotion of tumor immune escape. Therefore, inhibition of CK2 is initially proposed as a pivotal candidate for cancer treatment. In this review, we discussed the role of CK2 in cancer progression and tumor therapy.
Collapse
Affiliation(s)
- Shakeel Hussain
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yilei Guo
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yu Huo
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Juanjuan Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yongzhong Hou
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| |
Collapse
|
22
|
Fassl A, Sicinski P. PC4: A new regulator of cyclin D1 transcript levels. J Cell Biol 2024; 223:e202401056. [PMID: 38393314 PMCID: PMC10890923 DOI: 10.1083/jcb.202401056] [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] [Indexed: 02/25/2024] Open
Abstract
The expression of cyclin proteins is tightly regulated during the cell cycle, to allow precise activation of cyclin-dependent kinases. In this issue, Pan et al. (https://doi.org/10.1083/jcb.202308066) identify an RNA-binding protein, PC4, as a regulator of cyclin D1 mRNA stability in hepatocellular carcinoma cells. This study provides a new mechanism regulating the levels of a key cell cycle protein, cyclin D1, in human cells.
Collapse
Affiliation(s)
- Anne Fassl
- Department of Urology, Goethe-University Frankfurt, University Hospital, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Frankfurt am Main, Germany
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Choi Y, Yu SR, Lee Y, Na AY, Lee S, Heitman J, Seo R, Lee HS, Lee JS, Bahn YS. Casein kinase 2 complex: a central regulator of multiple pathobiological signaling pathways in Cryptococcus neoformans. mBio 2024; 15:e0327523. [PMID: 38193728 PMCID: PMC10865844 DOI: 10.1128/mbio.03275-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 01/10/2024] Open
Abstract
The casein kinase 2 (CK2) complex has garnered extensive attention over the past decades as a potential therapeutic target for diverse human diseases, including cancer, diabetes, and obesity, due to its pivotal roles in eukaryotic growth, differentiation, and metabolic homeostasis. While CK2 is also considered a promising antifungal target, its role in fungal pathogens remains unexplored. In this study, we investigated the functions and regulatory mechanisms of the CK2 complex in Cryptococcus neoformans, a major cause of fungal meningitis. The cryptococcal CK2 complex consists of a single catalytic subunit, Cka1, and two regulatory subunits, Ckb1 and Ckb2. Our findings show that Cka1 plays a primary role as a protein kinase, while Ckb1 and Ckb2 have major and minor regulatory functions, respectively, in growth, cell cycle control, morphogenesis, stress response, antifungal drug resistance, and virulence factor production. Interestingly, triple mutants lacking all three subunits (cka1Δ ckb1Δ ckb2Δ) exhibited more severe phenotypic defects than the cka1Δ mutant alone, suggesting that Ckb1/2 may have Cka1-independent functions. In a murine model of systemic cryptococcosis, cka1Δ and cka1Δ ckb1Δ ckb2Δ mutants showed severely reduced virulence. Transcriptomic, proteomic, and phosphoproteomic analyses further revealed that the CK2 complex controls a wide array of effector proteins involved in transcriptional regulation, cell cycle control, nutrient metabolisms, and stress responses. Most notably, CK2 disruption led to dysregulation of key signaling cascades central to C. neoformans pathogenicity, including the Hog1, Mpk1 MAPKs, cAMP/PKA, and calcium/calcineurin signaling pathways. In summary, our study provides novel insights into the multifaceted roles of the fungal CK2 complex and presents a compelling case for targeting it in the development of new antifungal drugs.IMPORTANCEThe casein kinase 2 (CK2) complex, crucial for eukaryotic growth, differentiation, and metabolic regulation, presents a promising therapeutic target for various human diseases, including cancer, diabetes, and obesity. Its potential as an antifungal target is further highlighted in this study, which explores CK2's functions in C. neoformans, a key fungal meningitis pathogen. The CK2 complex in C. neoformans, comprising the Cka1 catalytic subunit and Ckb1/2 regulatory subunits, is integral to processes like growth, cell cycle, morphogenesis, stress response, drug resistance, and virulence. Our findings of CK2's role in regulating critical signaling pathways, including Hog1, Mpk1 MAPKs, cAMP/PKA, and calcium/calcineurin, underscore its importance in C. neoformans pathogenicity. This study provides valuable insights into the fungal CK2 complex, reinforcing its potential as a target for novel antifungal drug development and pointing out a promising direction for creating new antifungal agents.
Collapse
Affiliation(s)
- Yeseul Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Seong-Ryong Yu
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Yujin Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Ann-Yae Na
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Sangkyu Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Ran Seo
- AmtixBio Co., Ltd., Hanam-si, Gyeonggi-do, South Korea
| | - Han-Seung Lee
- AmtixBio Co., Ltd., Hanam-si, Gyeonggi-do, South Korea
| | | | - Yong-Sun Bahn
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| |
Collapse
|
24
|
Wang P, Sun S, Lam S, Lockwood WW. New insights into the biology and development of lung cancer in never smokers-implications for early detection and treatment. J Transl Med 2023; 21:585. [PMID: 37653450 PMCID: PMC10472682 DOI: 10.1186/s12967-023-04430-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/10/2023] [Indexed: 09/02/2023] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide. Despite never smokers comprising between 10 and 25% of all cases, lung cancer in never smokers (LCNS) is relatively under characterized from an etiological and biological perspective. The application of multi-omics techniques on large patient cohorts has significantly advanced the current understanding of LCNS tumor biology. By synthesizing the findings of multi-omics studies on LCNS from a clinical perspective, we can directly translate knowledge regarding tumor biology into implications for patient care. Primarily focused on never smokers with lung adenocarcinoma, this review details the predominance of driver mutations, particularly in East Asian patients, as well as the frequency and importance of germline variants in LCNS. The mutational patterns present in LCNS tumors are thoroughly explored, highlighting the high abundance of the APOBEC signature. Moreover, this review recognizes the spectrum of immune profiles present in LCNS tumors and posits how it can be translated to treatment selection. The recurring and novel insights from multi-omics studies on LCNS tumor biology have a wide range of clinical implications. Risk factors such as exposure to outdoor air pollution, second hand smoke, and potentially diet have a genomic imprint in LCNS at varying degrees, and although they do not encompass all LCNS cases, they can be leveraged to stratify risk. Germline variants similarly contribute to a notable proportion of LCNS, which warrants detailed documentation of family history of lung cancer among never smokers and demonstrates value in developing testing for pathogenic variants in never smokers for early detection in the future. Molecular driver subtypes and specific co-mutations and mutational signatures have prognostic value in LCNS and can guide treatment selection. LCNS tumors with no known driver alterations tend to be stem-like and genes contributing to this state may serve as potential therapeutic targets. Overall, the comprehensive findings of multi-omics studies exert a wide influence on clinical management and future research directions in the realm of LCNS.
Collapse
Affiliation(s)
- Peiyao Wang
- Department of Integrative Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - Sophie Sun
- Department of Medical Oncology, British Columbia Cancer Agency Vancouver, Vancouver, BC, Canada
| | - Stephen Lam
- Department of Integrative Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - William W Lockwood
- Department of Integrative Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada.
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
25
|
Patel S, Vyas VK, Sharma M, Ghate M. Structure-guided discovery of adenosine triphosphate-competitive casein kinase 2 inhibitors. Future Med Chem 2023; 15:987-1014. [PMID: 37307219 DOI: 10.4155/fmc-2023-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023] Open
Abstract
Casein kinase 2 (CK2) is a ubiquitous, highly pleiotropic serine-threonine kinase. CK2 has been identified as a potential drug target for the treatment of cancer and related disorders. Several adenosine triphosphate-competitive CK2 inhibitors have been identified and have progressed at different levels of clinical trials. This review presents details of CK2 protein, structural insights into adenosine triphosphate binding pocket, current clinical trial candidates and their analogues. Further, it includes the emerging structure-based drug design approaches, chemistry, structure-activity relationship and biological screening of potent and selective CK2 inhibitors. The authors tabulated the details of CK2 co-crystal structures because these co-crystal structures facilitated the structure-guided discovery of CK2 inhibitors. The narrow hinge pocket compared with related kinases provides useful insights into the discovery of CK2 inhibitors.
Collapse
Affiliation(s)
- Shivani Patel
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Manmohan Sharma
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Manjunath Ghate
- School of Pharmacy, National Forensic Science University, Gandhinagar, Gujarat, 382007, India
| |
Collapse
|
26
|
van Ree JH, Jeganathan KB, Fierro Velasco RO, Zhang C, Can I, Hamada M, Li H, Baker DJ, van Deursen JM. Hyperphosphorylated PTEN exerts oncogenic properties. Nat Commun 2023; 14:2983. [PMID: 37225693 PMCID: PMC10209192 DOI: 10.1038/s41467-023-38740-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 05/12/2023] [Indexed: 05/26/2023] Open
Abstract
PTEN is a multifaceted tumor suppressor that is highly sensitive to alterations in expression or function. The PTEN C-tail domain, which is rich in phosphorylation sites, has been implicated in PTEN stability, localization, catalytic activity, and protein interactions, but its role in tumorigenesis remains unclear. To address this, we utilized several mouse strains with nonlethal C-tail mutations. Mice homozygous for a deletion that includes S370, S380, T382 and T383 contain low PTEN levels and hyperactive AKT but are not tumor prone. Analysis of mice containing nonphosphorylatable or phosphomimetic versions of S380, a residue hyperphosphorylated in human gastric cancers, reveal that PTEN stability and ability to inhibit PI3K-AKT depends on dynamic phosphorylation-dephosphorylation of this residue. While phosphomimetic S380 drives neoplastic growth in prostate by promoting nuclear accumulation of β-catenin, nonphosphorylatable S380 is not tumorigenic. These data suggest that C-tail hyperphosphorylation creates oncogenic PTEN and is a potential target for anti-cancer therapy.
Collapse
Affiliation(s)
- Janine H van Ree
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Karthik B Jeganathan
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Ismail Can
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Masakazu Hamada
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Jan M van Deursen
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
27
|
Davis-Gilbert Z, Krämer A, Dunford JE, Howell S, Senbabaoglu F, Wells CI, Bashore FM, Havener TM, Smith JL, Hossain MA, Oppermann U, Drewry DH, Axtman AD. Discovery of a Potent and Selective Naphthyridine-Based Chemical Probe for Casein Kinase 2. ACS Med Chem Lett 2023; 14:432-441. [PMID: 37077385 PMCID: PMC10108397 DOI: 10.1021/acsmedchemlett.2c00530] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/07/2023] [Indexed: 03/15/2023] Open
Abstract
Naphthyridine-based inhibitors were synthesized to yield a potent and cell-active inhibitor of casein kinase 2 (CK2). Compound 2 selectively inhibits CK2α and CK2α' when profiled broadly, thereby making it an exquisitely selective chemical probe for CK2. A negative control that is structurally related but lacks a key hinge-binding nitrogen (7) was designed on the basis of structural studies. Compound 7 does not bind CK2α or CK2α' in cells and demonstrates excellent kinome-wide selectivity. Differential anticancer activity was observed when compound 2 was profiled alongside a structurally distinct CK2 chemical probe: SGC-CK2-1. This naphthyridine-based chemical probe (2) represents one of the best available small molecule tools with which to interrogate biology mediated by CK2.
Collapse
Affiliation(s)
- Zachary
W. Davis-Gilbert
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Andreas Krämer
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue-Strabe 9, Frankfurt 60438, Germany
- Structural
Genomics Consortium, Buchmann Institute for Life Sciences, Goethe University Frankfurt, Max-von-Laue-Strabe 15, Frankfurt 60438, Germany
- Frankfurt
Cancer Institute, Paul-Ehrlich-Straße
42-44, Frankfurt 60596, Germany
| | - James E. Dunford
- Botnar
Research Centre, Nuffield Department of Orthopaedics, Rheumatology
and Musculoskeletal Sciences, University
of Oxford, Oxford OX3 7LD, United Kingdom
| | - Stefanie Howell
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Filiz Senbabaoglu
- Botnar
Research Centre, Nuffield Department of Orthopaedics, Rheumatology
and Musculoskeletal Sciences, University
of Oxford, Oxford OX3 7LD, United Kingdom
| | - Carrow I. Wells
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Frances M. Bashore
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Tammy M. Havener
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jeffery L. Smith
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mohammad A. Hossain
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Udo Oppermann
- Botnar
Research Centre, Nuffield Department of Orthopaedics, Rheumatology
and Musculoskeletal Sciences, University
of Oxford, Oxford OX3 7LD, United Kingdom
- Oxford
Translational
Myeloma Centre, University of Oxford, Oxford OX3 7LD, United Kingdom
| | - David H. Drewry
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- UNC
Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Alison D. Axtman
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
28
|
Song M, Pang L, Zhang M, Qu Y, Laster KV, Dong Z. Cdc2-like kinases: structure, biological function, and therapeutic targets for diseases. Signal Transduct Target Ther 2023; 8:148. [PMID: 37029108 PMCID: PMC10082069 DOI: 10.1038/s41392-023-01409-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 04/09/2023] Open
Abstract
The CLKs (Cdc2-like kinases) belong to the dual-specificity protein kinase family and play crucial roles in regulating transcript splicing via the phosphorylation of SR proteins (SRSF1-12), catalyzing spliceosome molecular machinery, and modulating the activities or expression of non-splicing proteins. The dysregulation of these processes is linked with various diseases, including neurodegenerative diseases, Duchenne muscular dystrophy, inflammatory diseases, viral replication, and cancer. Thus, CLKs have been considered as potential therapeutic targets, and significant efforts have been exerted to discover potent CLKs inhibitors. In particular, clinical trials aiming to assess the activities of the small molecules Lorecivivint on knee Osteoarthritis patients, and Cirtuvivint and Silmitasertib in different advanced tumors have been investigated for therapeutic usage. In this review, we comprehensively documented the structure and biological functions of CLKs in various human diseases and summarized the significance of related inhibitors in therapeutics. Our discussion highlights the most recent CLKs research, paving the way for the clinical treatment of various human diseases.
Collapse
Affiliation(s)
- Mengqiu Song
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
- China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan, 450008, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
| | - Luping Pang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Research Center of Basic Medicine, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Mengmeng Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yingzi Qu
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Kyle Vaughn Laster
- China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan, 450008, China
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan, 450008, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China.
- Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
29
|
Lindberg MF, Deau E, Arfwedson J, George N, George P, Alfonso P, Corrionero A, Meijer L. Comparative Efficacy and Selectivity of Pharmacological Inhibitors of DYRK and CLK Protein Kinases. J Med Chem 2023; 66:4106-4130. [PMID: 36876904 DOI: 10.1021/acs.jmedchem.2c02068] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Dual-specificity, tyrosine phosphorylation-regulated kinases (DYRKs) and cdc2-like kinases (CLKs) play a large variety of cellular functions and are involved in several diseases (cognitive disorders, diabetes, cancers, etc.). There is, thus, growing interest in pharmacological inhibitors as chemical probes and potential drug candidates. This study presents an unbiased evaluation of the kinase inhibitory activity of a library of 56 reported DYRK/CLK inhibitors on the basis of comparative, side-by-side, catalytic activity assays on a panel of 12 recombinant human kinases, enzyme kinetics (residence time and Kd), in-cell inhibition of Thr-212-Tau phosphorylation, and cytotoxicity. The 26 most active inhibitors were modeled in the crystal structure of DYRK1A. The results show a rather large diversity of potencies and selectivities among the reported inhibitors and emphasize the difficulties to avoid "off-targets" in this area of the kinome. The use of a panel of DYRKs/CLKs inhibitors is suggested to analyze the functions of these kinases in cellular processes.
Collapse
Affiliation(s)
| | - Emmanuel Deau
- Perha Pharmaceuticals, Perharidy Peninsula, 29680 Roscoff, France
| | - Jonas Arfwedson
- Perha Pharmaceuticals, Perharidy Peninsula, 29680 Roscoff, France
| | - Nicolas George
- Oncodesign, 25-27 avenue du Québec, 91140 Villebon-sur-Yvette, France
| | - Pascal George
- Perha Pharmaceuticals, Perharidy Peninsula, 29680 Roscoff, France
| | - Patricia Alfonso
- Enzymlogic, Qube Technology Park, C/Santiago Grisolía, 2, 28760 Madrid, Spain
| | - Ana Corrionero
- Enzymlogic, Qube Technology Park, C/Santiago Grisolía, 2, 28760 Madrid, Spain
| | - Laurent Meijer
- Perha Pharmaceuticals, Perharidy Peninsula, 29680 Roscoff, France
| |
Collapse
|
30
|
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. [PMID: 36745746 DOI: 10.1021/acs.jmedchem.2c01523] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CK2 (casein kinase 2) is a serine/threonine protein kinase that is ubiquitous in eukaryotic cells and plays important roles in a variety of cellular functions, including cell growth, apoptosis, circadian rhythms, DNA damage repair, transcription, and translation. CK2 is involved in cancer pathogenesis and the occurrence of many diseases. Therefore, targeting CK2 is a promising therapeutic strategy. Although many CK2-specific small-molecule inhibitors have been developed, only CX-4945 has progressed to clinical trials. In recent years, novel CK2 inhibitors have gradually become a research hotspot, which is expected to overcome the limitations of traditional inhibitors. Herein, we summarize the structure, biological functions, and disease relevance of CK2 and emphatically analyze the structure-activity relationship (SAR) and binding modes of small-molecule CK2 inhibitors. We also discuss the latest progress of novel strategies, providing insights into new drugs targeting CK2 for clinical practice.
Collapse
Affiliation(s)
- Yijia Chen
- Joint Research Institution of Altitude Health, Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.,College of Life Sciences, Sichuan University, Chengdu, Sichuan 610064, China
| | - Yuxi Wang
- Joint Research Institution of Altitude Health, Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.,Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.,Tianfu Jincheng Laboratory, Chengdu, Sichuan 610041, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Zhilan Zhou
- Joint Research Institution of Altitude Health, Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shu Cao
- West China School of Stomatology Sichuan University, Chengdu, Sichuan 610064, China
| | - Jifa Zhang
- Joint Research Institution of Altitude Health, Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.,Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.,Tianfu Jincheng Laboratory, Chengdu, Sichuan 610041, China
| |
Collapse
|
31
|
Mitrovský O, Myslivcová D, Macháčková-Lopotová T, Obr A, Čermáková K, Ransdorfová Š, Březinová J, Klamová H, Žáčková M. Inhibition of casein kinase 2 induces cell death in tyrosine kinase inhibitor resistant chronic myelogenous leukemia cells. PLoS One 2023; 18:e0284876. [PMID: 37141212 PMCID: PMC10159124 DOI: 10.1371/journal.pone.0284876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 04/11/2023] [Indexed: 05/05/2023] Open
Abstract
Chronic myelogenous leukemia (CML) is a myeloproliferative disease characterized by the BCR-ABL oncogene. Despite the high performance of treatment with tyrosine kinase inhibitors (TKI), about 30% of patients develop resistance to the therapy. To improve the outcomes, identification of new targets of treatment is needed. Here, we explored the Casein Kinase 2 (CK2) as a potential target for CML therapy. Previously, we detected increased phosphorylation of HSP90β Serine 226 in patients non-responding to TKIs imatinib and dasatinib. This site is known to be phosphorylated by CK2, which was also linked to CML resistance to imatinib. In the present work, we established six novel imatinib- and dasatinib-resistant CML cell lines, all of which had increased CK2 activation. A CK2 inhibitor, CX-4945, induced cell death of CML cells in both parental and resistant cell lines. In some cases, CK2 inhibition also potentiated the effects of TKI on the cell metabolic activity. No effects of CK2 inhibition were observed in normal mononuclear blood cells from healthy donors and BCR-ABL negative HL60 cell line. Our data indicate that CK2 kinase supports CML cell viability even in cells with different mechanisms of resistance to TKI, and thus represents a potential target for treatment.
Collapse
Affiliation(s)
- Ondřej Mitrovský
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague 2, Czech Republic
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Denisa Myslivcová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague 2, Czech Republic
| | | | - Adam Obr
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague 2, Czech Republic
| | - Kamila Čermáková
- Laboratory of PCR Diagnostics of Leukemias, Institute of Hematology and Blood Transfusion, Prague 2, Czech Republic
| | - Šárka Ransdorfová
- Department of Cytogenetics, Institute of Hematology and Blood Transfusion, Prague 2, Czech Republic
| | - Jana Březinová
- Department of Cytogenetics, Institute of Hematology and Blood Transfusion, Prague 2, Czech Republic
| | - Hana Klamová
- Clinical Division, Institute of Hematology and Blood Transfusion, Prague 2, Czech Republic
| | - Markéta Žáčková
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague 2, Czech Republic
| |
Collapse
|
32
|
Fan J, Tong G, Chen X, Li S, Yu Y, Zhu S, Zhu K, Hu Z, Dong Y, Chen R, Zhu J, Gong W, Hu Z, Zhou B, Chen Y, Jin L, Cong W. CK2 blockade alleviates liver fibrosis by suppressing activation of hepatic stellate cells via the Hedgehog pathway. Br J Pharmacol 2023; 180:44-61. [PMID: 36070072 DOI: 10.1111/bph.15945] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Liver fibrosis is a serious cause of morbidity and mortality worldwide characterized by accumulation of extracellular matrix produced by hepatic stellate cells (HSCs). The protein kinase CK2 is a pro-survival kinase overexpressed in human tumours. However, the biological role of CK2 in liver fibrosis is largely unknown. We aimed to investigate the mechanism by which CK2 promotes liver fibrosis. EXPERIMENTAL APPROACH In vitro, LX-2 cells were stimulated with transforming growth factor-β (TGF-β). HSCs were also isolated for research. In vivo, the adeno-associated virus AAV-sh-csnk2a1 was used to knockdown CK2α specifically in HSCs, and CX-4945 was used to pharmacologically inhibit the enzymatic activity of CK2 in murine models of fibrosis induced by carbon tetrachloride (CCl4 ) and a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet. Histological and biochemical analyses were performed to study the involvement of CK2 in regulation of fibrogenic and fibrolytic factors as well as activation properties of HSCs. KEY RESULTS HSC-specific genetic invalidation of CK2α or pharmacological inhibition of CK2 protected mice treated with CCl4 or fed a DDC diet against liver fibrosis and HSC accumulation. Mechanistically, CK2α, which bound to Smoothened (SMO), was a positive regulator of the Hedgehog signal transduction pathway. CK2 prevented ubiquitination and proteasomal degradation of SMO, which was abolished by knockdown of CK2α or pharmacological inhibition of CK2. CONCLUSIONS AND IMPLICATIONS CK2 activation is critical to sustain the activated and fibrogenic phenotype of HSCs via SMO stabilization. Therefore, inactivation of CK2 by CX-4945 may be of therapeutic interest for liver fibrotic diseases.
Collapse
Affiliation(s)
- Junfu Fan
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaozan Tong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xixi Chen
- Department of Pharmacy, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Santie Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Yu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shunan Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kunxuan Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zijing Hu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yonggan Dong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Rui Chen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junjie Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenjie Gong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhicheng Hu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bin Zhou
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiming Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
33
|
Kwon J, Zhang J, Mok B, Han C. CK2-Mediated Phosphorylation Upregulates the Stability of USP13 and Promotes Ovarian Cancer Cell Proliferation. Cancers (Basel) 2022; 15:cancers15010200. [PMID: 36612196 PMCID: PMC9818633 DOI: 10.3390/cancers15010200] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Ubiquitin-specific Peptidase 13 (USP13) is a deubiquitinating enzyme that regulates the stability or function of its substrate. USP13 is highly amplified in human ovarian cancer, and elevated expression of USP13 promotes tumorigenesis and metastasis of ovarian cancer. However, there is little known about USP13 post-translational modifications and their role in ovarian cancer. Here, we found that USP13 is phosphorylated at Thr122 in ovarian cancer cells. Phosphorylated Thr122 (pT122) on endogenous USP13 was observed in most human ovarian cancer cells, and the abundance of this phosphorylation was correlated to the total level of USP13. We further demonstrated that Casein kinase 2 (CK2) directly interacts with and phosphorylates USP13 at Thr122, which promotes the stability of USP13 protein. Finally, we showed that Threonine 122 is important for cell proliferation of ovarian cancer cells. Our findings may reveal a novel regulatory mechanism for USP13, which may lead to novel therapeutic targeting of USP13 in ovarian cancer.
Collapse
Affiliation(s)
- Juntae Kwon
- Department of Oncology, Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Jinmin Zhang
- Department of Oncology, Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Boram Mok
- Department of Oncology, Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Cecil Han
- Department of Oncology, Georgetown University School of Medicine, Washington, DC 20007, USA
- Lombardi Comprehensive Cancer Center, Georgetown University School of Medicine, Washington, DC 20007, USA
- Correspondence:
| |
Collapse
|
34
|
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:biomedicines11010043. [PMID: 36672551 PMCID: PMC9856093 DOI: 10.3390/biomedicines11010043] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Large cell lung carcinoma (LCLC) is one form of NSCLC that spreads more aggressively than some other forms, and it represents an unmet medical need. Here, we investigated for the first time the effect of the anti-CK2 CIGB-300 peptide in NCI-H460 cells as an LCLC model. NCI-H460 cells were highly sensitive toward CIGB-300 cytotoxicity, reaching a peak of apoptosis at 6 h. Moreover, CIGB-300 slightly impaired the cell cycle of NCI-H460 cells. The CIGB-300 interactomics profile revealed in more than 300 proteins that many of them participated in biological processes relevant in cancer. Interrogation of the CK2 subunits targeting by CIGB-300 indicated the higher binding of the peptide to the CK2α' catalytic subunit by in vivo pull-down assays plus immunoblotting analysis and confocal microscopy. The down-regulation of both phosphorylation and protein levels of the ribonuclear protein S6 (RPS6) was observed 48 h post treatment. Altogether, we have found that NCI-H460 cells are the most CIGB-300-sensitive solid tumor cell line described so far, and also, the findings we provide here uncover novel features linked to CK2 targeting by the CIGB-300 anticancer peptide.
Collapse
|
35
|
Glinkina K, Nemati F, Teunisse AFAS, Gelmi MC, Etienne V, Kuipers MJ, Alsafadi S, Jager MJ, Decaudin D, Jochemsen AG. Preclinical Evaluation of Trabectedin in Combination With Targeted Inhibitors for Treatment of Metastatic Uveal Melanoma. Invest Ophthalmol Vis Sci 2022; 63:14. [PMID: 36515935 PMCID: PMC9756579 DOI: 10.1167/iovs.63.13.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose Uveal melanoma (UM) is considered a rare disease; yet, it is the most common intraocular malignancy in adults. Although the primary tumor may be efficiently managed, more than 50% of patients with UM develop distant metastases. The mortality at the first year after diagnosis of metastatic UM has been estimated at 81%, and the poor prognosis has not improved in the past years due to the lack of effective therapies. Methods In order to search for novel therapeutic possibilities for metastatic UM, we performed a small-scale screen of targeted drug combinations. We verified the targets of the tested compounds by western blotting and PCR and clarified the mechanism of action of the selected combinations by caspase 3 and 7 activity assay and flow cytometry. The best two combinations were tested in a mouse patient-derived xenograft (PDX) UM model as putative therapeutics for metastatic UM. Results Combinations of the multitarget drug trabectedin with either the CK2/CLK double-inhibitor CX-4945 (silmitasertib) or the c-MET/TAM (TYRO3, Axl, MERTK) receptor inhibitors foretinib and cabozantinib demonstrated synergistic effects and induced apoptosis (relative caspase 3 and 7 activity increased up to 20.5-fold in UM cell lines). In the case of the combination of foretinib and cabozantinib, inhibition of the TAM receptors, but not c-Met, was essential to inhibit the growth of UM cells. Monotreatment with trabectedin inhibited tumor growth by 42%, 49%, and 35% in the MM26, MM309, and MM339 PDX mouse models, respectively. Conclusions Trabectedin alone or in combination with cabozantinib inhibited tumor growth in PDX UM mouse models. Blocking of MERTK, rather than TYRO3, activity inhibited UM cell growth and synergized with trabectedin.
Collapse
Affiliation(s)
- Kseniya Glinkina
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Fariba Nemati
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University, Paris, France
| | - Amina F. A. S. Teunisse
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria Chiara Gelmi
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Vesnie Etienne
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University, Paris, France
| | - Muriel J. Kuipers
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Samar Alsafadi
- Uveal Melanoma Translational Group, Department of Translational Research, Institut Curie, PSL University, Paris, France
| | - Martine J. Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Didier Decaudin
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University, Paris, France,Department of Medical Oncology, Institut Curie, PSL University, Paris, France
| | - Aart G. Jochemsen
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
36
|
Alsagaby SA, Iqbal D, Ahmad I, Patel H, Mir SA, Madkhali YA, Oyouni AAA, Hawsawi YM, Alhumaydhi FA, Alshehri B, Alturaiki W, Alanazi B, Mir MA, Al Abdulmonem W. In silico investigations identified Butyl Xanalterate to competently target CK2α (CSNK2A1) for therapy of chronic lymphocytic leukemia. Sci Rep 2022; 12:17648. [PMID: 36271116 PMCID: PMC9587039 DOI: 10.1038/s41598-022-21546-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/28/2022] [Indexed: 01/18/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is an incurable malignancy of B-cells. In this study, bioinformatics analyses were conducted to identify possible pathogenic roles of CK2α, which is a protein encoded by CSNK2A1, in the progression and aggressiveness of CLL. Furthermore, various computational tools were used to search for a competent inhibitor of CK2α from fungal metabolites that could be proposed for CLL therapy. In CLL patients, high-expression of CSNK2A1 was associated with early need for therapy (n = 130, p < 0.0001) and short overall survival (OS; n = 107, p = 0.005). Consistently, bioinformatics analyses showed CSNK2A1 to associate with/play roles in CLL proliferation and survival-dependent pathways. Furthermore, PPI network analysis identified interaction partners of CK2α (PPI enrichment p value = 1 × 10-16) that associated with early need for therapy (n = 130, p < 0.003) and have been known to heavily impact on the progression of CLL. These findings constructed a rational for targeting CK2α for CLL therapy. Consequently, computational analyses reported 35 fungal metabolites out of 5820 (filtered from 19,967 metabolites) to have lower binding energy (ΔG: - 10.9 to - 11.7 kcal/mol) and better binding affinity (Kd: 9.77 × 107 M-1 to 3.77 × 108 M-1) compared with the native ligand (ΔG: - 10.8, Kd: 8.3 × 107 M--1). Furthermore, molecular dynamics simulation study established that Butyl Xanalterate-CK2α complex continuously remained stable throughout the simulation time (100 ns). Moreover, Butyl Xanalterate interacted with most of the catalytic residues, where complex was stabilized by more than 65% hydrogen bond interactions, and a significant hydrophobic interaction with residue Phe113. Here, high-expression of CSNK2A1 was implicated in the progression and poor prognosis of CLL, making it a potential therapeutic target in the disease. Butyl Xanalterate showed stable and strong interactions with CK2α, thus we propose it as a competitive inhibitor of CK2α for CLL therapy.
Collapse
Affiliation(s)
- Suliman A. Alsagaby
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Danish Iqbal
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Iqrar Ahmad
- grid.412233.50000 0001 0641 8393Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra 425405 India
| | - Harun Patel
- grid.412233.50000 0001 0641 8393Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra 425405 India
| | - Shabir Ahmad Mir
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Yahya Awaji Madkhali
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Atif Abdulwahab A. Oyouni
- grid.440760.10000 0004 0419 5685Department of Biology, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia ,grid.440760.10000 0004 0419 5685Genome and Biotechnology Unit, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Yousef M. Hawsawi
- grid.415310.20000 0001 2191 4301Research Center, King Faisal Specialist Hospital and Research Center, P.O. Box 40047, Jeddah, 21499 Kingdom of Saudi Arabia ,grid.411335.10000 0004 1758 7207College of Medicine, Al-Faisal University, P.O. Box 50927, Riyadh, 11533 Kingdom of Saudi Arabia
| | - Fahad A. Alhumaydhi
- grid.412602.30000 0000 9421 8094Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Bader Alshehri
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Wael Alturaiki
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Bader Alanazi
- grid.415277.20000 0004 0593 1832Biomedical Research Administration, Research Center, King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia ,Prince Mohammed bin Abdulaziz Medical City, AlJouf, Kingdom of Saudi Arabia
| | - Manzoor Ahmad Mir
- grid.412997.00000 0001 2294 5433Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Waleed Al Abdulmonem
- grid.412602.30000 0000 9421 8094Department of Pathology, College of Medicine, Qassim University, Qassim, Kingdom of Saudi Arabia
| |
Collapse
|
37
|
Trembley JH, Kren BT, Afzal M, Scaria GA, Klein MA, Ahmed K. Protein kinase CK2 – diverse roles in cancer cell biology and therapeutic promise. Mol Cell Biochem 2022; 478:899-926. [PMID: 36114992 PMCID: PMC9483426 DOI: 10.1007/s11010-022-04558-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022]
Abstract
The association of protein kinase CK2 (formerly casein kinase II or 2) with cell growth and proliferation in cells was apparent at early stages of its investigation. A cancer-specific role for CK2 remained unclear until it was determined that CK2 was also a potent suppressor of cell death (apoptosis); the latter characteristic differentiated its function in normal versus malignant cells because dysregulation of both cell growth and cell death is a universal feature of cancer cells. Over time, it became evident that CK2 exerts its influence on a diverse range of cell functions in normal as well as in transformed cells. As such, CK2 and its substrates are localized in various compartments of the cell. The dysregulation of CK2 is documented in a wide range of malignancies; notably, by increased CK2 protein and activity levels with relatively moderate change in its RNA abundance. High levels of CK2 are associated with poor prognosis in multiple cancer types, and CK2 is a target for active research and testing for cancer therapy. Aspects of CK2 cellular roles and targeting in cancer are discussed in the present review, with focus on nuclear and mitochondrial functions and prostate, breast and head and neck malignancies.
Collapse
Affiliation(s)
- Janeen H Trembley
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA.
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Betsy T Kren
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA
| | - Muhammad Afzal
- Department of Biochemistry, Riphah International University, Islamabad, Pakistan
| | - George A Scaria
- Hematology/Oncology Section, Primary Care Service Line, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA
| | - Mark A Klein
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
- Hematology/Oncology Section, Primary Care Service Line, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Khalil Ahmed
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA.
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
- Department of Urology, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
38
|
Firnau MB, Brieger A. CK2 and the Hallmarks of Cancer. Biomedicines 2022; 10:1987. [PMID: 36009534 PMCID: PMC9405757 DOI: 10.3390/biomedicines10081987] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer is a leading cause of death worldwide. Casein kinase 2 (CK2) is commonly dysregulated in cancer, impacting diverse molecular pathways. CK2 is a highly conserved serine/threonine kinase, constitutively active and ubiquitously expressed in eukaryotes. With over 500 known substrates and being estimated to be responsible for up to 10% of the human phosphoproteome, it is of significant importance. A broad spectrum of diverse types of cancer cells has been already shown to rely on disturbed CK2 levels for their survival. The hallmarks of cancer provide a rationale for understanding cancer's common traits. They constitute the maintenance of proliferative signaling, evasion of growth suppressors, resisting cell death, enabling of replicative immortality, induction of angiogenesis, the activation of invasion and metastasis, as well as avoidance of immune destruction and dysregulation of cellular energetics. In this work, we have compiled evidence from the literature suggesting that CK2 modulates all hallmarks of cancer, thereby promoting oncogenesis and operating as a cancer driver by creating a cellular environment favorable to neoplasia.
Collapse
Affiliation(s)
| | - Angela Brieger
- Department of Internal Medicine I, Biomedical Research Laboratory, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
39
|
Kim JM, Yang YS, Xie J, Lee O, Kim J, Hong J, Boldyreff B, Filhol O, Chun H, Greenblatt MB, Gao G, Shim JH. Regulation of sclerostin by the SIRT1 stabilization pathway in osteocytes. Cell Death Differ 2022; 29:1625-1638. [PMID: 35169297 PMCID: PMC9345882 DOI: 10.1038/s41418-022-00952-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 11/09/2022] Open
Abstract
Osteocytes play a critical role in bone remodeling through the secretion of paracrine factors regulating the differentiation and activity of osteoblasts and osteoclasts. Sclerostin is a key osteocyte-derived factor that suppresses bone formation and promotes bone resorption, therefore regulators of sclerostin secretion are a likely source of new therapeutic strategies for treatment of skeletal disorders. Here, we demonstrate that protein kinase CK2 (casein kinase 2) controls sclerostin expression in osteocytes via the deubiquitinase ubiquitin-specific peptidase 4 (USP4)-mediated stabilization of Sirtuin1 (SIRT1). Deletion of CK2 regulatory subunit, Csnk2b, in osteocytes (Csnk2bDmp1) results in low bone mass due to elevated levels of sclerostin. This phenotype in Csnk2bDmp1 mice was partly reversed when sclerostin expression was downregulated by a single intravenous injection with bone-targeting adeno-associated virus 9 (AAV9) carrying an artificial-microRNA that targets Sost. Mechanistically, CK2-induced phosphorylation of USP4 is important for stabilization of SIRT1 by suppressing ubiquitin-dependent proteasomal degradation. Upregulated expression of SIRT1 inhibits sclerostin transcription in osteocytes. Collectively, the CK2-USP4-SIRT1 pathway is crucial for the regulation of sclerostin expression in osteocytes to maintain bone homeostasis.
Collapse
Affiliation(s)
- Jung-Min Kim
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Yeon-Suk Yang
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Jun Xie
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- Viral Vector Core, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Oksun Lee
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - JiHea Kim
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Jaehyoung Hong
- Department of Mathematical Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | | | - Odile Filhol
- Interdisciplinary Research Institute of Grenoble, IRIG-Biosanté, University Grenoble Alpes, CEA, UMR 1292, F-38000, Grenoble, France
| | - Hyonho Chun
- Department of Mathematical Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
- Hospital for Special Surgery, New York, NY, 10021, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- Viral Vector Core, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Jae-Hyuck Shim
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
40
|
Nipun VB, Amin KA. Recent Advances in Protein Kinase CK2, a Potential Therapeutic Target in Cancer. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022; 48:919-931. [DOI: 10.1134/s1068162022050144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- V. B. Nipun
- Cancer Research Center, Shantou University Medical Collage, Shantou, Guangdong, 515041, PR China
- Department of Chemistry, Faculty of Science, University of Imam Abdulrahman Bin Faisal university, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - K. A. Amin
- Department of Chemistry, Faculty of Science, University of Imam Abdulrahman Bin Faisal university, P.O. Box 1982, Dammam, 31441, Saudi Arabia
- Basic and Applied Scientific Research Center, Imam Abdulrahman Bin Faisal university, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| |
Collapse
|
41
|
Zhou L, Wang H, Liu H, Huang Z, Wang Z, Zhou X, Mu X. The synergistic therapeutic effect of imatinib and protein kinase CK2 Inhibition correlates with PI3K-AKT activation in gastrointestinal stromal tumors. Clin Res Hepatol Gastroenterol 2022; 46:101886. [PMID: 35183792 DOI: 10.1016/j.clinre.2022.101886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 12/28/2021] [Accepted: 02/01/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract. Casein kinase 2 (CK2) has been reported to be involved in several cellular processes in multiple cancers. However, the role of CK2 in GIST remains unclear. AIM We aimed to investigate the combinatorial treatment of imatinib (IM) and CK2 inhibition on the progression of GISTs. METHODS GIST biopsies and adjacent normal tissues were collected from patients. GIST882 and GIST48 cell lines were subjected to investigate the effect of IM and CK2 inhibition in GIST cells. CCK-8 assay, Caspase-3 activity assay, western blotting, and flow cytometry analysis were employed in the present investigation. RESULTS Our results showed that CK2 was highly expressed in GIST biopsies, and inhibition of CK2 resulted in decrease in cell viability and increase in apoptosis of GIST cells. Moreover, the combination treatment with CX-4945 (CX) and IM resulted in a more significant decrease in cell viability and increase in cell apoptosis compared with mono-treatment. Mechanistically, the combination treatment influenced the activation of the PI3K/AKT pathway. The activation of the PI3K/AKT pathway reversed the synergistic impacts of the combined treatment on cell viability and apoptosis. CONCLUSION Our results demonstrated that inhibition of CK2 combined with IM exhibited a synergistic anti-cancer effect on GIST cells through inactivation of the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Linsen Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, Jiangsu 215006, China; Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School and The First people's Hospital of Yancheng, No. 166 Yulongxi Road, Yancheng, Jiangsu 224001, China
| | - Hao Wang
- Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School and The First people's Hospital of Yancheng, No. 166 Yulongxi Road, Yancheng, Jiangsu 224001, China
| | - Haofeng Liu
- Department of General Surgery, Tumor Hospital Affiliated to Nantong University and Nantong Tumor Hospital, Nantong, Jiangsu 226361, China
| | - Zhijun Huang
- Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School and The First people's Hospital of Yancheng, No. 166 Yulongxi Road, Yancheng, Jiangsu 224001, China
| | - Zhiqiang Wang
- Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School and The First people's Hospital of Yancheng, No. 166 Yulongxi Road, Yancheng, Jiangsu 224001, China
| | - Xiaojun Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, Jiangsu 215006, China.
| | - Xiangming Mu
- Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School and The First people's Hospital of Yancheng, No. 166 Yulongxi Road, Yancheng, Jiangsu 224001, China.
| |
Collapse
|
42
|
Mishra S, Kinoshita C, Axtman AD, Young JE. Evaluation of a Selective Chemical Probe Validates That CK2 Mediates Neuroinflammation in a Human Induced Pluripotent Stem Cell-Derived Microglial Model. Front Mol Neurosci 2022; 15:824956. [PMID: 35774866 PMCID: PMC9239073 DOI: 10.3389/fnmol.2022.824956] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/20/2022] [Indexed: 01/11/2023] Open
Abstract
Novel treatments for neurodegenerative disorders are in high demand. It is imperative that new protein targets be identified to address this need. Characterization and validation of nascent targets can be accomplished very effectively using highly specific and potent chemical probes. Human induced pluripotent stem cells (hiPSCs) provide a relevant platform for testing new compounds in disease relevant cell types. However, many recent studies utilizing this platform have focused on neuronal cells. In this study, we used hiPSC-derived microglia-like cells (MGLs) to perform side-by-side testing of a selective chemical probe, SGC-CK2-1, compared with an advanced clinical candidate, CX-4945, both targeting casein kinase 2 (CK2), one of the first kinases shown to be dysregulated in Alzheimer's disease (AD). CK2 can mediate neuroinflammation in AD, however, its role in microglia, the innate immune cells of the central nervous system (CNS), has not been defined. We analyzed available RNA-seq data to determine the microglial expression of kinases inhibited by SGC-CK2-1 and CX-4945 with a reported role in mediating inflammation in glial cells. As proof-of-concept for using hiPSC-MGLs as a potential screening platform, we used both wild-type (WT) MGLs and MGLs harboring a mutation in presenilin-1 (PSEN1), which is causative for early-onset, familial AD (FAD). We stimulated these MGLs with pro-inflammatory lipopolysaccharides (LPS) derived from E. coli and observed strong inhibition of the expression and secretion of proinflammatory cytokines by simultaneous treatment with SGC-CK2-1. A direct comparison shows that SGC-CK2-1 was more effective at suppression of proinflammatory cytokines than CX-4945. Together, these results validate a selective chemical probe, SGC-CK2-1, in human microglia as a tool to reduce neuroinflammation.
Collapse
Affiliation(s)
- Swati Mishra
- Department of Laboratory Medicine and Pathology, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Chizuru Kinoshita
- Department of Laboratory Medicine and Pathology, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Alison D. Axtman
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jessica E. Young
- Department of Laboratory Medicine and Pathology, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
43
|
New kinase and HDAC hybrid inhibitors: recent advances and perspectives. Future Med Chem 2022; 14:745-766. [PMID: 35543381 DOI: 10.4155/fmc-2021-0276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cancer is the second most common cause of death worldwide. It can easily acquire resistance to treatments, demanding new therapeutic strategies, such as simultaneous inhibition of kinase and HDAC enzymes with hybrid inhibitors. Different approaches to this have varied according to their targets, with a few common trends, such as the usage of heterocycle scaffolds for kinase interaction, especially pyrimidine and quinazolines, and hydroxamic acids and benzamides for HDAC inhibition. Besides the hybrid compounds developed focusing on the inhibition tyrosine kinase and receptor tyrosine kinase, many advances have occurred in the development of serine-threonine kinase/HDAC and lipid kinase/HDAC novel compounds. Here, the latest strategies employed in this research area will be reviewed, alongside trends in inhibitor design, and observed gaps will be punctuated.
Collapse
|
44
|
Schwarz R, Richter A, Ito ERD, Murua Escobar H, Junghanß C, Hinz B. Validation of an LC-MS/MS Method for the Quantification of the CK2 Inhibitor Silmitasertib (CX-4945) in Human Plasma. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27082394. [PMID: 35458589 PMCID: PMC9028559 DOI: 10.3390/molecules27082394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/15/2022] [Accepted: 03/22/2022] [Indexed: 11/16/2022]
Abstract
Silmitasertib (CX-4945) is currently being investigated in clinical trials against various types of cancer. The U.S. Food and Drug Administration (FDA) has already granted orphan drug designation to the compound for the treatment of advanced cholangiocarcinoma, medulloblastoma, and biliary tract cancer. Silmitasertib inhibits the serine/threonine protein kinase CK2, which exerts a proliferation-promoting and anti-apoptotic effect on cancer cells. In view of current and future applications, the measurement of silmitasertib levels in plasma is expected to play an important role in the evaluation of therapeutic and toxic concentrations in cancer patients. In the present work, we therefore present an LC-MS/MS method for the quantification of silmitasertib in human plasma. Using a simple liquid-liquid extraction with ethyl acetate and a mixture of n-hexane and ethyl acetate, this method can be performed in any laboratory with mass spectrometry. The validation was carried out according to the FDA guideline.
Collapse
Affiliation(s)
- Rico Schwarz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18057 Rostock, Germany; (R.S.); (E.R.D.I.)
| | - Anna Richter
- Clinic for Hematology, Oncology and Palliative Care, Rostock University Medical Center, 18057 Rostock, Germany; (A.R.); (H.M.E.); (C.J.)
| | - Elisabeth R. D. Ito
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18057 Rostock, Germany; (R.S.); (E.R.D.I.)
| | - Hugo Murua Escobar
- Clinic for Hematology, Oncology and Palliative Care, Rostock University Medical Center, 18057 Rostock, Germany; (A.R.); (H.M.E.); (C.J.)
| | - Christian Junghanß
- Clinic for Hematology, Oncology and Palliative Care, Rostock University Medical Center, 18057 Rostock, Germany; (A.R.); (H.M.E.); (C.J.)
| | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18057 Rostock, Germany; (R.S.); (E.R.D.I.)
- Correspondence:
| |
Collapse
|
45
|
Bijani S, Iqbal D, Mirza S, Jain V, Jahan S, Alsaweed M, Madkhali Y, Alsagaby SA, Banawas S, Algarni A, Alrumaihi F, Rawal RM, Alturaiki W, Shah A. Green Synthesis and Anticancer Potential of 1,4-Dihydropyridines-Based Triazole Derivatives: In Silico and In Vitro Study. Life (Basel) 2022; 12:519. [PMID: 35455010 PMCID: PMC9029820 DOI: 10.3390/life12040519] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 11/16/2022] Open
Abstract
A library of 1,4-dihydropyridine-based 1,2,3-triazol derivatives has been designed, synthesized, and evaluated their cytotoxic potential on colorectal adenocarcinoma (Caco-2) cell lines. All compounds were characterized and identified based on their 1H and 13C NMR (Nuclear Magnetic Resonance) spectroscopic data. Furthermore, molecular docking of best anticancer hits with target proteins (protein kinase CK2α, tankyrase1, and tankyrase2) has been performed. Our results implicated that most of these compounds have significant antiproliferative activity with IC50 values between 0.63 ± 0.05 and 5.68 ± 0.14 µM. Moreover, the mechanism of action of most active compounds 13ab' and 13ad' suggested that they induce cell death through apoptosis in the late apoptotic phase as well as dead phase, and they could promote cell cycle arrest at the G2/M phase. Furthermore, the molecular docking study illustrated that 13ad' possesses better binding interaction with the catalytic residues of target proteins involved in cell proliferation and antiapoptotic pathways. Based on our in vitro and in silico study, 13ad' was found to be a highly effective anti-cancerous compound. The present data indicate that dihydropyridine-linked 1,2,3-triazole conjugates can be generated as potent anticancer agents.
Collapse
Affiliation(s)
- Sabera Bijani
- Department of Chemistry, Marwadi University, Rajkot 360005, Gujarat, India; (S.B.); (V.J.)
- Center of Excellence, National Facility for Drug Discovery Complex, Department of Chemistry, Saurashtra University, Rajkot 360005, Gujarat, India
| | - Danish Iqbal
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia; (S.J.); (M.A.); (Y.M.); (S.A.A.); (S.B.); (W.A.)
- Health and Basic Sciences Research Center, Majmaah University, Al Majmaah 15341, Saudi Arabia
| | - Sheefa Mirza
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa;
| | - Vicky Jain
- Department of Chemistry, Marwadi University, Rajkot 360005, Gujarat, India; (S.B.); (V.J.)
- Center of Excellence, National Facility for Drug Discovery Complex, Department of Chemistry, Saurashtra University, Rajkot 360005, Gujarat, India
| | - Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia; (S.J.); (M.A.); (Y.M.); (S.A.A.); (S.B.); (W.A.)
| | - Mohammed Alsaweed
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia; (S.J.); (M.A.); (Y.M.); (S.A.A.); (S.B.); (W.A.)
| | - Yahya Madkhali
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia; (S.J.); (M.A.); (Y.M.); (S.A.A.); (S.B.); (W.A.)
| | - Suliman A. Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia; (S.J.); (M.A.); (Y.M.); (S.A.A.); (S.B.); (W.A.)
| | - Saeed Banawas
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia; (S.J.); (M.A.); (Y.M.); (S.A.A.); (S.B.); (W.A.)
- Health and Basic Sciences Research Center, Majmaah University, Al Majmaah 15341, Saudi Arabia
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Abdulrahman Algarni
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Northern Border University, Arar 91431, Saudi Arabia;
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51425, Saudi Arabia;
| | - Rakesh M. Rawal
- Department of Life Sciences, Gujarat University, Ahmedabad 380009, Gujarat, India;
| | - Wael Alturaiki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia; (S.J.); (M.A.); (Y.M.); (S.A.A.); (S.B.); (W.A.)
| | - Anamik Shah
- Center of Excellence, National Facility for Drug Discovery Complex, Department of Chemistry, Saurashtra University, Rajkot 360005, Gujarat, India
- B/H Forensic Laboratory, Saurashtra University Karmachari Cooperative Society, Rajkot 360005, Gujarat, India
| |
Collapse
|
46
|
Zhou L, Zhang Z, Nice E, Huang C, Zhang W, Tang Y. Circadian rhythms and cancers: the intrinsic links and therapeutic potentials. J Hematol Oncol 2022; 15:21. [PMID: 35246220 PMCID: PMC8896306 DOI: 10.1186/s13045-022-01238-y] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
The circadian rhythm is an evolutionarily conserved time-keeping system that comprises a wide variety of processes including sleep-wake cycles, eating-fasting cycles, and activity-rest cycles, coordinating the behavior and physiology of all organs for whole-body homeostasis. Acute disruption of circadian rhythm may lead to transient discomfort, whereas long-term irregular circadian rhythm will result in the dysfunction of the organism, therefore increasing the risks of numerous diseases especially cancers. Indeed, both epidemiological and experimental evidence has demonstrated the intrinsic link between dysregulated circadian rhythm and cancer. Accordingly, a rapidly increasing understanding of the molecular mechanisms of circadian rhythms is opening new options for cancer therapy, possibly by modulating the circadian clock. In this review, we first describe the general regulators of circadian rhythms and their functions on cancer. In addition, we provide insights into the mechanisms underlying how several types of disruption of the circadian rhythm (including sleep-wake, eating-fasting, and activity-rest) can drive cancer progression, which may expand our understanding of cancer development from the clock perspective. Moreover, we also summarize the potential applications of modulating circadian rhythms for cancer treatment, which may provide an optional therapeutic strategy for cancer patients.
Collapse
Affiliation(s)
- Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Edouard Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Acupuncture and Chronobiology Laboratory of Sichuan Province, Chengdu, 610075, China.
| |
Collapse
|
47
|
Wińska P, Widło Ł, Senkara E, Koronkiewicz M, Cieśla JM, Krzyśko A, Skierka K, Cieśla J. Inhibition of Protein Kinase CK2 Affects Thymidylate Synthesis Cycle Enzyme Level and Distribution in Human Cancer Cells. Front Mol Biosci 2022; 9:847829. [PMID: 35281258 PMCID: PMC8914513 DOI: 10.3389/fmolb.2022.847829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Thymidylate synthase (TS), dihydrofolate reductase (DHFR), and serine hydroxymethyltransferase (SHMT) constitute the thymidylate synthesis cycle providing thymidylate for DNA synthesis and repair. Our previous studies indicated that TS and DHFR are the substrates of protein kinase CK2. This work has been aimed at the elucidation of the effect of CK2 activity on cell cycle progression, thymidylate synthesis enzyme expression and localization, and the role of CK2-mediated TS phosphorylation in in vitro di- and trimolecular complex formation. The results were obtained by means of western blot, confocal microscopy, flow cytometry, quantitative polymerase chain reaction (QPCR), quartz crystal microbalance with dissipation monitoring (QCM-D), and microthermophoresis (MST). Our research indicates that CK2 inhibition does not change the levels of the transcripts; however, it affects the protein levels of DHFR and TS in both tested cell lines, i.e., A549 and CCRF-CEM, and the level of SHMT1 in CCRF-CEM cells. Moreover, we show that CK2-mediated phosphorylation of TS enables the protein (pTS) interaction with SHMT1 and leads to the stability of the tri-complex containing SHMT1, DHFR, and pTS. Our results suggest an important regulatory role of CK2-mediated phosphorylation for inter- and intracellular protein level of enzymes involved in the thymidylate biosynthesis cycle.
Collapse
Affiliation(s)
- Patrycja Wińska
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
- *Correspondence: Patrycja Wińska, ; Joanna Cieśla,
| | - Łukasz Widło
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Elżbieta Senkara
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | | | - Jarosław M. Cieśla
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Alicja Krzyśko
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Katarzyna Skierka
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Joanna Cieśla
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
- *Correspondence: Patrycja Wińska, ; Joanna Cieśla,
| |
Collapse
|
48
|
Pucko EB, Ostrowski RP. Inhibiting CK2 among Promising Therapeutic Strategies for Gliomas and Several Other Neoplasms. Pharmaceutics 2022; 14:331. [PMID: 35214064 PMCID: PMC8877581 DOI: 10.3390/pharmaceutics14020331] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
In gliomas, casein kinase 2 (CK2) plays a dominant role in cell survival and tumour invasiveness and is upregulated in many brain tumours. Among CK2 inhibitors, benzimidazole and isothiourea derivatives hold a dominant position. While targeting glioma tumour cells, they show limited toxicity towards normal cells. Research in recent years has shown that these compounds can be suitable as components of combined therapies with hyperbaric oxygenation. Such a combination increases the susceptibility of glioma tumour cells to cell death via apoptosis. Moreover, researchers planning on using any other antiglioma investigational pharmaceutics may want to consider using these agents in combination with CK2 inhibitors. However, different compounds are not equally effective when in such combination. More research is needed to elucidate the mechanism of treatment and optimize the treatment regimen. In addition, the role of CK2 in gliomagenesis and maintenance seems to have been challenged recently, as some compounds structurally similar to CK2 inhibitors do not inhibit CK2 while still being effective at reducing glioma viability and invasion. Furthermore, some newly developed inhibitors specific for CK2 do not appear to have strong anticancer properties. Further experimental and clinical studies of these inhibitors and combined therapies are warranted.
Collapse
Affiliation(s)
| | - Robert P. Ostrowski
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| |
Collapse
|
49
|
SGC-CK2-1 Is an Efficient Inducer of Insulin Production and Secretion in Pancreatic β-Cells. Pharmaceutics 2021; 14:pharmaceutics14010019. [PMID: 35056914 PMCID: PMC8778508 DOI: 10.3390/pharmaceutics14010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022] Open
Abstract
The pyrazolopyrimidine based compound SGC-CK2-1 is a potent and highly specific CK2 inhibitor and a new tool to study the biological functions of protein kinase CK2 irrespective from off-target effects. We used this compound in comparison with the well-established CK2 inhibitor CX-4945 to analyze the importance of CK2 for insulin production and secretion from pancreatic β-cells. Both inhibitors affected the proliferation and viability of MIN6 cells only marginally and downregulated the endogenous CK2 activity to a similar level. Furthermore, both inhibitors increased the message for insulin and boosted the secretion of insulin from storage vesicles. Thus, regarding the high specificity of SGC-CK2-1, we can clearly attribute the observed effects to biological functions of protein kinase CK2.
Collapse
|
50
|
Birus R, El-Awaad E, Ballentin L, Alchab F, Aichele D, Ettouati L, Götz C, Le Borgne M, Jose J. 4,5,7-Trisubstituted indeno[1,2-b]indole inhibits CK2 activity in tumor cells equivalent to CX-4945 and shows strong anti-migratory effects. FEBS Open Bio 2021; 12:394-411. [PMID: 34873879 PMCID: PMC8804612 DOI: 10.1002/2211-5463.13346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/22/2021] [Accepted: 12/06/2021] [Indexed: 11/07/2022] Open
Abstract
Highly pleiotropic and constitutively active protein kinase CK2 is a key target in cancer therapy, but only one small-molecule inhibitor has reached clinical trials-CX-4945. In this study, we present the indeno[1,2-b]indole derivative 5-isopropyl-4-methoxy-7-methyl-5,6,7,8-tetrahydroindeno[1,2-b]indole-9,10-dione (5a-2) that decreased the intracellular CK2 activity in A431, A549, and LNCaP tumor cell lines analogous to CX-4945 (> 75% inhibition at 20 µm) and similarly blocked CK2-specific Akt phosphorylation in LNCaP cells. Cellular uptake analysis demonstrated higher intracellular concentrations of 5a-2 (408.3 nm) compared with CX-4945 (119.3 nm). This finding clarifies the comparable effects of both compounds on the intracellular CK2 activity despite their different inhibitory potency in vitro [IC50 = 25 nm (5a-2) and 3.7 nm (CX-4945)]. Examination of the effects of both CK2 inhibitors on cancer cells using live-cell imaging revealed notable differences. Whereas CX-4945 showed a stronger pro-apoptotic effect on tumor cells, 5a-2 was more effective in inhibiting tumor cell migration. Our results showed that 49% of intracellular CX-4945 was localized in the nuclear fraction, whereas 71% of 5a-2 was detectable in the cytoplasm. The different subcellular distribution, and thus the site of CK2 inhibition, provides a possible explanation for the different cellular effects. Our study indicates that investigating CK2 inhibition-mediated cellular effects in relation to the subcellular sites of CK2 inhibition may help to improve our understanding of the preferential roles of CK2 within different cancer cell compartments.
Collapse
Affiliation(s)
- Robin Birus
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universtität Münster, Germany
| | - Ehab El-Awaad
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universtität Münster, Germany.,Department of Pharmacology, Faculty of Medicine, Assiut University, Egypt
| | - Laurens Ballentin
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universtität Münster, Germany
| | - Faten Alchab
- EEA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie-ISPB, SFR Santé Lyon-Est CNRS UMS3453-INSERM US7, Université Claude Bernard Lyon 1, Université de Lyon, France.,Faculty of Pharmacy, Manara University, Latakia, Syria
| | - Dagmar Aichele
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universtität Münster, Germany
| | - Laurent Ettouati
- CNRS UMR 5246 Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), Faculté de Pharmacie, ISPB, Université Lyon 1, Université de Lyon, France
| | - Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University, Germany
| | - Marc Le Borgne
- Small Molecules for Biological Targets Team, Centre de recherche en cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, Université de Lyon, France
| | - Joachim Jose
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universtität Münster, Germany
| |
Collapse
|