1
|
Ruggeri E, Frezzato F, Mouawad N, Pizzi M, Scarmozzino F, Capasso G, Trimarco V, Quotti Tubi L, Cellini A, Cavarretta CA, Ruocco V, Serafin A, Angotzi F, Danesin N, Manni S, Facco M, Piazza F, Trentin L, Visentin A. Protein kinase CK2α is overexpressed in classical hodgkin lymphoma, regulates key signaling pathways, PD-L1 and may represent a new target for therapy. Front Immunol 2024; 15:1393485. [PMID: 38807597 PMCID: PMC11130512 DOI: 10.3389/fimmu.2024.1393485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/25/2024] [Indexed: 05/30/2024] Open
Abstract
Introduction In classical Hodgkin lymphoma (cHL), the survival of neoplastic cells is mediated by the activation of NF-κB, JAK/STAT and PI3K/Akt signaling pathways. CK2 is a highly conserved serine/threonine kinase, consisting of two catalytic (α) and two regulatory (β) subunits, which is involved in several cellular processes and both subunits were found overexpressed in solid tumors and hematologic malignancies. Methods and results Biochemical analyses and in vitro assays showed an impaired expression of CK2 subunits in cHL, with CK2α being overexpressed and a decreased expression of CK2β compared to normal B lymphocytes. Mechanistically, CK2β was found to be ubiquitinated in all HL cell lines and consequently degraded by the proteasome pathway. Furthermore, at basal condition STAT3, NF-kB and AKT are phosphorylated in CK2-related targets, resulting in constitutive pathways activation. The inhibition of CK2 with CX-4945/silmitasertib triggered the de-phosphorylation of NF-κB-S529, STAT3-S727, AKT-S129 and -S473, leading to cHL cell lines apoptosis. Moreover, CX-4945/silmitasertib was able to decrease the expression of the immuno-checkpoint CD274/PD-L1 but not of CD30, and to synergize with monomethyl auristatin E (MMAE), the microtubule inhibitor of brentuximab vedotin. Conclusions Our data point out a pivotal role of CK2 in the survival and the activation of key signaling pathways in cHL. The skewed expression between CK2α and CK2β has never been reported in other lymphomas and might be specific for cHL. The effects of CK2 inhibition on PD-L1 expression and the synergistic combination of CX-4945/silmitasertib with MMAE pinpoints CK2 as a high-impact target for the development of new therapies for cHL.
Collapse
Affiliation(s)
- Edoardo Ruggeri
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Federica Frezzato
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Nayla Mouawad
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Marco Pizzi
- Surgical Pathology and Cytopathology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Federico Scarmozzino
- Surgical Pathology and Cytopathology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Guido Capasso
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Valentina Trimarco
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Laura Quotti Tubi
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Alessandro Cellini
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | | | - Valeria Ruocco
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Andrea Serafin
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Francesco Angotzi
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Nicolò Danesin
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Sabrina Manni
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Monica Facco
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Francesco Piazza
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Livio Trentin
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Andrea Visentin
- Hematology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| |
Collapse
|
2
|
Zhao R, Cheng S, Bai X, Zhang D, Fang H, Che W, Zhang W, Zhou Y, Duan W, Liang Q, Xiao L, Nie G, Hou Y. Development of an efficient liposomal DOX delivery formulation for HCC therapy by targeting CK2α. Biotechnol J 2024; 19:e2400050. [PMID: 38651271 DOI: 10.1002/biot.202400050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/11/2024] [Accepted: 03/23/2024] [Indexed: 04/25/2024]
Abstract
Hepatocellular carcinoma (HCC) is a digestive tract cancer with high mortality and poor prognosis, especially in China. Current chemotherapeutic drugs lead to poor prognosis, low efficacy, and high side effects due to weak targeting specificity and rapidly formed multidrug resistance (MDR). Based on the previous studies on the doxorubicin (DOX) formulation for cancer targeting therapy, we developed a novel DOX delivery formulation for the targeting chemotherapy of HCC and DOX resistant HCC. HCSP4 was previously screened and casein kinase 2α (CK2α) was predicted as its specific target on HCC cells in our lab. In the study, miR125a-5p was firstly predicted as an MDR inhibiting miRNA, and then CK2α was validated as the target of HCSP4 and miR125a-5p using CK2α-/-HepG2 cells. Based on the above, an HCC targeting and MDR inhibiting DOX delivery liposomal formulation, HCSP4/Lipo-DOX/miR125a-5p was synthesized and tested for its HCC therapeutic efficacy in vitro. The results showed that the liposomal DOX delivery formulation targeted to HCC cells specifically and sensitively, and presented the satisfied therapeutic efficacy for HCC, particularly for DOX resistant HCC. The potential therapeutic mechanism of the DOX delivery formulation was explored, and the formulation inhibited the expression of MDR-relevant genes including ATP-binding cassette subfamily B member 1 (ABCB1, also known as P-glycoprotein), ATP-binding cassette subfamily C member 5 (ABCC5), enhancer of zeste homolog 2 (EZH2), and ATPase Na+/K+ transporting subunit beta 1 (ATP1B1). Our study presents a novel targeting chemotherapeutic drug formulation for the therapy of HCC, especially for drug resistant HCC, although it is primarily and needs further study in vivo, but provided a new strategy for the development of novel anticancer drugs.
Collapse
Affiliation(s)
- Ruixia Zhao
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Sinan Cheng
- Changzhi Medical College, Changzhi, Shanxi, China
| | - Xue Bai
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Danying Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Hongming Fang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Wanlin Che
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Wenxuan Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Yujuan Zhou
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Wei Duan
- School of Medicine, Deakin University, Waurn Ponds, VIC, Australia
| | - Qiumin Liang
- Guangxi Key Laboratory of Agricultural Resource Chemistry and Biotechnology, Yulin, Guangxi, China
| | - Li Xiao
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
- Xi'an Medical University, Xi'an, Shaanxi, China
| | - Guochao Nie
- Guangxi Key Laboratory of Agricultural Resource Chemistry and Biotechnology, Yulin, Guangxi, China
| | - Yingchun Hou
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| |
Collapse
|
3
|
Kanaoka D, Yamada M, Yokoyama H, Nishino S, Kunimura N, Satoyoshi H, Wakabayashi S, Urabe K, Ishii T, Nakanishi M. FPFT-2216, a Novel Anti-lymphoma Compound, Induces Simultaneous Degradation of IKZF1/3 and CK1α to Activate p53 and Inhibit NFκB Signaling. CANCER RESEARCH COMMUNICATIONS 2024; 4:312-327. [PMID: 38265263 PMCID: PMC10846380 DOI: 10.1158/2767-9764.crc-23-0264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/03/2023] [Accepted: 01/19/2024] [Indexed: 01/25/2024]
Abstract
Reducing casein kinase 1α (CK1α) expression inhibits the growth of multiple cancer cell lines, making it a potential therapeutic target for cancer. Herein, we evaluated the antitumor activity of FPFT-2216-a novel low molecular weight compound-in lymphoid tumors and elucidated its molecular mechanism of action. In addition, we determined whether targeting CK1α with FPFT-2216 is useful for treating hematopoietic malignancies. FPFT-2216 strongly degraded CK1α and IKAROS family zinc finger 1/3 (IKZF1/3) via proteasomal degradation. FPFT-2216 exhibited stronger inhibitory effects on human lymphoma cell proliferation than known thalidomide derivatives and induced upregulation of p53 and its transcriptional targets, namely, p21 and MDM2. Combining FPFT-2216 with an MDM2 inhibitor exhibited synergistic antiproliferative activity and induced rapid tumor regression in immunodeficient mice subcutaneously transplanted with a human lymphoma cell line. Nearly all tumors in mice disappeared after 10 days; this was continuously observed in 5 of 7 mice up to 24 days after the final FPFT-2216 administration. FPFT-2216 also enhanced the antitumor activity of rituximab and showed antitumor activity in a patient-derived diffuse large B-cell lymphoma xenograft model. Furthermore, FPFT-2216 decreased the activity of the CARD11/BCL10/MALT1 (CBM) complex and inhibited IκBα and NFκB phosphorylation. These effects were mediated through CK1α degradation and were stronger than those of known IKZF1/3 degraders. In conclusion, FPFT-2216 inhibits tumor growth by activating the p53 signaling pathway and inhibiting the CBM complex/NFκB pathway via CK1α degradation. Therefore, FPFT-2216 may represent an effective therapeutic agent for hematopoietic malignancies, such as lymphoma. SIGNIFICANCE We found potential vulnerability to CK1α degradation in certain lymphoma cells refractory to IKZF1/3 degraders. Targeting CK1α with FPFT-2216 could inhibit the growth of these cells by activating p53 signaling. Our study demonstrates the potential therapeutic application of CK1α degraders, such as FPFT-2216, for treating lymphoma.
Collapse
Affiliation(s)
- Daiki Kanaoka
- Department of Scientific Research, Fujimoto Pharmaceutical Corporation, Nishi-otsuka, Matsubara, Osaka, Japan
| | - Mitsuo Yamada
- Department of Scientific Research, Fujimoto Pharmaceutical Corporation, Nishi-otsuka, Matsubara, Osaka, Japan
| | - Hironori Yokoyama
- Department of Scientific Research, Fujimoto Pharmaceutical Corporation, Nishi-otsuka, Matsubara, Osaka, Japan
| | - Satoko Nishino
- Department of Scientific Research, Fujimoto Pharmaceutical Corporation, Nishi-otsuka, Matsubara, Osaka, Japan
| | - Naoshi Kunimura
- Department of Scientific Research, Fujimoto Pharmaceutical Corporation, Nishi-otsuka, Matsubara, Osaka, Japan
| | - Hiroshi Satoyoshi
- Department of Scientific Research, Fujimoto Pharmaceutical Corporation, Nishi-otsuka, Matsubara, Osaka, Japan
| | - Shota Wakabayashi
- Department of Scientific Research, Fujimoto Pharmaceutical Corporation, Nishi-otsuka, Matsubara, Osaka, Japan
| | - Kazunori Urabe
- Department of Scientific Research, Fujimoto Pharmaceutical Corporation, Nishi-otsuka, Matsubara, Osaka, Japan
| | - Takafumi Ishii
- Department of Scientific Research, Fujimoto Pharmaceutical Corporation, Nishi-otsuka, Matsubara, Osaka, Japan
| | - Masato Nakanishi
- Department of Scientific Research, Fujimoto Pharmaceutical Corporation, Nishi-otsuka, Matsubara, Osaka, Japan
| |
Collapse
|
4
|
Ahsan R, Khan MM, Mishra A, Noor G, Ahmad U. Protein Kinases and their Inhibitors Implications in Modulating Disease Progression. Protein J 2023; 42:621-632. [PMID: 37768476 DOI: 10.1007/s10930-023-10159-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2023] [Indexed: 09/29/2023]
Abstract
Protein phosphorylation plays an important role in cellular pathways, including cell cycle regulation, metabolism, differentiation and survival. The protein kinase superfamily network consists of 518 members involved in intrinsic or extrinsic interaction processes. Protein kinases are divided into two categories based on their ability to phosphorylate tyrosine, serine, and threonine residues. The complexity of the system implies its vulnerability. Any changes in the pathways of protein kinases may be implicated in pathological processes. Therefore, they are regarded as having an important role in human diseases and represent prospective therapeutic targets. This article provides a review of the protein kinase inhibitors approved by the FDA. Finally, we summarize the mechanism of action of protein kinases, including their role in the development and progression of protein kinase-related roles in various pathological conditions and the future therapeutic potential of protein kinase inhibitors, along with links to protein kinase databases. Further clinical studies aimed at examining the sequence of protein kinase inhibitor availability would better utilize current protein kinase inhibitors in diseases. Additionally, this review may help researchers and biochemists find new potent and selective protein kinase inhibitors and provide more indications for using existing drugs.
Collapse
Affiliation(s)
- Rabiya Ahsan
- Department of pharmacology, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Mohd Muazzam Khan
- Department of pharmacology, Faculty of Pharmacy, Integral University, Lucknow, India.
| | - Anuradha Mishra
- Department of pharmacology, Amity Institute of Pharmacy, Amity University, sector 125, Noida, Uttar Pradesh, 201313, India
| | - Gazala Noor
- Department of pharmacology, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Usama Ahmad
- Department of pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow, India
| |
Collapse
|
5
|
Yang WC, Gong DH, Hong Wu, Gao YY, Hao GF. Grasping cryptic binding sites to neutralize drug resistance in the field of anticancer. Drug Discov Today 2023; 28:103705. [PMID: 37453458 DOI: 10.1016/j.drudis.2023.103705] [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: 02/27/2023] [Revised: 06/09/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Drug resistance is a significant obstacle to successful cancer treatment. The utilization and development of cryptic binding sites (CBSs) in proteins involved in cancer-related drug-resistance (CRDR) could help to overcome that drug resistance. However, there is no comprehensive review of the successful use of CBSs in addressing CRDR. Here, we have systematically summarized and analyzed the opportunities and challenges of using CBSs in addressing CRDR and revealed the key role that CBSs have in targeting CRDR. First, we have identified the CRDR targets and the corresponding CBSs. Second, we discuss the mechanisms by which CBSs can overcome CRDR. Finally, we have provided examples of successful CBS applications in addressing CRDR. We hope that this approach will provide guidance to biologists and chemists in effectively utilizing CBSs for the development of new drugs to alleviate CRDR.
Collapse
Affiliation(s)
- Wei-Cheng Yang
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, China
| | - Dao-Hong Gong
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, China
| | - Hong Wu
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, China
| | - Yang-Yang Gao
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, China.
| | - Ge-Fei Hao
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, China; National Key Laboratory of Green Pesticide, Central China Normal University, Wuhan 430079, China.
| |
Collapse
|
6
|
Gu L, Jin X, Liang H, Yang C, Zhang Y. Upregulation of CSNK1A1 induced by ITGB5 confers to hepatocellular carcinoma resistance to sorafenib in vivo by disrupting the EPS15/EGFR complex. Pharmacol Res 2023; 192:106789. [PMID: 37149115 DOI: 10.1016/j.phrs.2023.106789] [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: 01/03/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023]
Abstract
Oral multitarget tyrosine kinase inhibitors (TKIs), such as sorafenib, which suppress tumor cell proliferation and tumor angiogenesis, have been approved to treat patients with hepatocellular carcinoma (HCC). Of note, only approximately 30% of patients can benefit from TKIs, and this population usually acquires drug resistance within 6 months. In this study, we intended to explore the mechanism associated with regulating the sensitivity of HCC to TKIs. We revealed that integrin subunit β 5 (ITGB5) is abnormally expressed in HCC and contributes to decreased the sensitivity of HCC to sorafenib. Mechanistically, unbiased mass spectrometry analysis using ITGB5 antibodies revealed that ITGB5 interacts with EPS15 to prevent the degradation of EGFR in HCC cells, which activates AKT-mTOR signaling and the MAPK pathway to reduce the sensitivity of HCC cells to sorafenib. In addition, mass spectrometry analysis showed that CSNK1A1 binds to ITGB5 in HCC cells. Further study indicated that ITGB5 increased the protein level of CSNK1A1 through the EGFR-AKT-mTOR pathway in HCC. Upregulated CSNK1A1 phosphorylates ITGB5 to enhance the interaction between ITGB5 and EPS15 and activate EGFR in HCC cells. Thus, we identified a positive feedback loop between ITGB5-EPS15-EGFR-CSNK1A1 in HCC cells. This finding provides a theoretical basis for the future development of therapeutic strategies to improve the anti-HCC efficacy of sorafenib.
Collapse
Affiliation(s)
- Li Gu
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xin Jin
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Uro-Oncology Institute of Central South University, Changsha, Hunan, 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Huaiyuan Liang
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Uro-Oncology Institute of Central South University, Changsha, Hunan, 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Chong Yang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province & Organ Transplantation Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China.
| | - Yu Zhang
- Hepatobiliary and Pancreatic Surgery Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
7
|
Dai XJ, Xue LP, Ji SK, Zhou Y, Gao Y, Zheng YC, Liu HM, Liu HM. Triazole-fused pyrimidines in target-based anticancer drug discovery. Eur J Med Chem 2023; 249:115101. [PMID: 36724635 DOI: 10.1016/j.ejmech.2023.115101] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/31/2022] [Accepted: 01/06/2023] [Indexed: 01/12/2023]
Abstract
In recent decades, the development of targeted drugs has featured prominently in the treatment of cancer, which is among the major causes of mortality globally. Triazole-fused pyrimidines, a widely-used class of heterocycles in medicinal chemistry, have attracted considerable interest as potential anticancer agents that target various cancer-associated targets in recent years, demonstrating them as valuable templates for discovering novel anticancer candidates. The current review concentrates on the latest advancements of triazole-pyrimidines as target-based anticancer agents, including works published between 2007 and the present (2007-2022). The structure-activity relationships (SARs) and multiple pathways are also reviewed to shed light on the development of more effective and biotargeted anticancer candidates.
Collapse
Affiliation(s)
- Xing-Jie Dai
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan Province, China
| | - Lei-Peng Xue
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan Province, China
| | - Shi-Kun Ji
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan Province, China
| | - Ying Zhou
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan Province, China
| | - Ya Gao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan Province, China
| | - Yi-Chao Zheng
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan Province, China
| | - Hui-Min Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan Province, China.
| | - Hong-Min Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan Province, China
| |
Collapse
|
8
|
Quotti Tubi L, Mandato E, Canovas Nunes S, Arjomand A, Zaffino F, Manni S, Casellato A, Macaccaro P, Vitulo N, Zumerle S, Filhol O, Boldyreff B, Siebel CW, Viola A, Valle G, Mainoldi F, Casola S, Cancila V, Gulino A, Tripodo C, Pizzi M, Dei Tos AP, Trentin L, Semenzato G, Piazza F. CK2β-regulated signaling controls B cell differentiation and function. Front Immunol 2023; 13:959138. [PMID: 36713383 PMCID: PMC9874936 DOI: 10.3389/fimmu.2022.959138] [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: 06/01/2022] [Accepted: 12/08/2022] [Indexed: 01/13/2023] Open
Abstract
Serine-Threonine kinase CK2 supports malignant B-lymphocyte growth but its role in B-cell development and activation is largely unknown. Here, we describe the first B-cell specific knockout (KO) mouse model of the β regulatory subunit of CK2. CK2βKO mice present an increase in marginal zone (MZ) and a reduction in follicular B cells, suggesting a role for CK2 in the regulation of the B cell receptor (BCR) and NOTCH2 signaling pathways. Biochemical analyses demonstrate an increased activation of the NOTCH2 pathway in CK2βKO animals, which sustains MZ B-cell development. Transcriptomic analyses indicate alterations in biological processes involved in immune response and B-cell activation. Upon sheep red blood cells (SRBC) immunization CK2βKO mice exhibit enlarged germinal centers (GCs) but display a limited capacity to generate class-switched GC B cells and immunoglobulins. In vitro assays highlight that B cells lacking CK2β have an impaired signaling downstream of BCR, Toll-like receptor, CD40, and IL-4R all crucial for B-cell activation and antigen presenting efficiency. Somatic hypermutations analysis upon 4-Hydroxy-3-nitrophenylacetyl hapten conjugated to Chicken Gamma Globulin (NP-CGG) evidences a reduced NP-specific W33L mutation frequency in CK2βKO mice suggesting the importance of the β subunit in sustaining antibody affinity maturation. Lastly, since diffuse large B cell lymphoma (DLBCL) cells derive from GC or post-GC B cells and rely on CK2 for their survival, we sought to investigate the consequences of CK2 inhibition on B cell signaling in DLBCL cells. In line with the observations in our murine model, CK2 inactivation leads to signaling defects in pathways that are essential for malignant B-lymphocyte activation.
Collapse
Affiliation(s)
- Laura Quotti Tubi
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Elisa Mandato
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Sara Canovas Nunes
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy,Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Arash Arjomand
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Fortunato Zaffino
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Sabrina Manni
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Alessandro Casellato
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Paolo Macaccaro
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Nicola Vitulo
- Department of Biology, Interdepartmental Research Center for Biotechnologies (CRIBI) Biotechnology Center, University of Padova, Padova, Italy
| | - Sara Zumerle
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Odile Filhol
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1036, Institute de Recherches en Technologies et Sciences pour le Vivant/Biologie du Cancer et de l’Infection, Grenoble, France
| | | | - Christian W. Siebel
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, CA, United States
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Giorgio Valle
- Department of Biology, Interdepartmental Research Center for Biotechnologies (CRIBI) Biotechnology Center, University of Padova, Padova, Italy
| | | | - Stefano Casola
- IFOM-ETS-The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, University of Palermo, Palermo, Italy
| | | | - Claudio Tripodo
- IFOM-ETS-The AIRC Institute of Molecular Oncology, Milan, Italy,Tumor Immunology Unit, University of Palermo, Palermo, Italy
| | - Marco Pizzi
- Department of Medicine, Cytopathology and Surgical Pathology Unit, University of Padova, Padova, Italy
| | - Angelo Paolo Dei Tos
- Department of Medicine, Cytopathology and Surgical Pathology Unit, University of Padova, Padova, Italy
| | - Livio Trentin
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Francesco Piazza
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy,*Correspondence: Francesco Piazza,
| |
Collapse
|
9
|
MDM4: What do we know about the association between its polymorphisms and cancer? MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:61. [PMID: 36566308 DOI: 10.1007/s12032-022-01929-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022]
Abstract
MDM4 is an important p53-negative regulator, consequently, it is involved in cell proliferation, DNA repair, and apoptosis regulation. MDM4 overexpression and amplification are described to lead to cancer formation, metastasis, and poor disease prognosis. Several MDM4 SNPs are in non-coding regions, and some affect the MDM4 regulation by disrupting the micro RNA binding site in 3'UTR (untranslated region). Here, we gathered several association studies with different MDM4 SNPs and populations to understand the relationship between its SNPs and solid tumor risk. Many studies failed to replicate their results regarding different populations, cancer types, and risk genotypes, leading to conflicting conclusions. We suggested that distinct haplotype patterns in different populations might affect the association between MDM4 SNPs and cancer risk. Thus, we propose to investigate some linkage SNPs in specific haplotypes to provide informative MDM4 markers for association studies with cancer.
Collapse
|
10
|
Yeast Protein Asf1 Possesses Modulating Activity towards Protein Kinase CK2. Int J Mol Sci 2022; 23:ijms232415764. [PMID: 36555405 PMCID: PMC9779303 DOI: 10.3390/ijms232415764] [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: 10/20/2022] [Revised: 11/30/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
Protein kinase CK2 plays an important role in cell survival and protects regulatory proteins from caspase-mediated degradation during apoptosis. The consensus sequence of proteins phosphorylated by CK2 contains a cluster of acidic amino acids around the phosphorylation site. The poly-acidic sequence in yeast protein Asf1 is similar to the acidic loop in CK2β, which possesses a regulatory function. We observed that the overexpression of Asf1 in yeast cells influences cell growth. Experiments performed in vitro and in vivo indicate that yeast protein Asf1 inhibits protein kinase CK2. Our data suggest that each CK2 isoform might be regulated in a different way. Deletion of the amino or carboxyl end of Asf1 reveals that the acidic cluster close to the C-terminus is responsible for the activation or inhibition of CK2 activity.
Collapse
|
11
|
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: 11] [Impact Index Per Article: 5.5] [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
|
12
|
Fregnani A, Saggin L, Gianesin K, Quotti Tubi L, Carraro M, Barilà G, Scapinello G, Bonetto G, Pesavento M, Berno T, Branca A, Gurrieri C, Zambello R, Semenzato G, Trentin L, Manni S, Piazza F. CK1α/RUNX2 Axis in the Bone Marrow Microenvironment: A Novel Therapeutic Target in Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14174173. [PMID: 36077711 PMCID: PMC9454895 DOI: 10.3390/cancers14174173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/10/2022] [Accepted: 08/22/2022] [Indexed: 11/18/2022] Open
Abstract
Simple Summary Multiple myeloma (MM) is an incurable disease for which novel therapeutic approaches targeting the malignant cells and the associated bone disease are urgently needed. CK1α is a protein kinase that plays a crucial role in the signaling network that sustains plasma cell (PC) survival and bone disease. This protein regulates Wnt/β-catenin signaling, which is fundamental for both MM cell survival and mesenchymal stromal cell (MSC) osteogenic differentiation. In this study, we investigated its involvement in MM–MSC cross-talk. We found that, by lowering CK1α expression levels in co-cultures of MM and MSC cells, expression of RUNX2—the master regulator of osteogenic differentiation—was regulated differently in the two cell types. Our data suggest the possibility of using a specific CK1α inhibitor as part of a novel therapeutic approach to selectively kill malignant PCs and overcome the blocking of osteogenic differentiation induced by MM cells in MSCs. Abstract Multiple myeloma (MM) is a malignant plasma cell (PC) neoplasm, which also displays pathological bone involvement. Clonal expansion of MM cells in the bone marrow causes a perturbation of bone homeostasis that culminates in MM-associated bone disease (MMABD). We previously demonstrated that the S/T kinase CK1α sustains MM cell survival through the activation of AKT and β-catenin signaling. CK1α is a negative regulator of the Wnt/β-catenin cascade, the activation of which promotes osteogenesis by directly stimulating the expression of RUNX2, the master gene regulator of osteoblastogenesis. In this study, we investigated the role of CK1α in the osteoblastogenic potential of mesenchymal stromal cells (MSCs) and its involvement in MM–MSC cross-talk. We found that CK1α silencing in in vitro co-cultures of MMs and MSCs modulated RUNX2 expression differently in PCs and in MSCs, mainly through the regulation of Wnt/β-catenin signaling. Our findings suggest that the CK1α/RUNX2 axis could be a potential therapeutic target for constraining malignant PC expansion and supporting the osteoblastic transcriptional program of MSCs, with potential for ameliorating MMABD. Moreover, considering that Lenalidomide treatment leads to MM cell death through Ikaros, Aiolos and CK1α proteasomal degradation, we examined its effects on the osteoblastogenic potential of MSC compartments.
Collapse
Affiliation(s)
- Anna Fregnani
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Lara Saggin
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Ketty Gianesin
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Laura Quotti Tubi
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Marco Carraro
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Gregorio Barilà
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Greta Scapinello
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Giorgia Bonetto
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Maria Pesavento
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Tamara Berno
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Antonio Branca
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Carmela Gurrieri
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Renato Zambello
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Gianpietro Semenzato
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Livio Trentin
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Sabrina Manni
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
- Correspondence: (S.M.); (F.P.); Tel.: +39-049-7923263 (S.M. & F.P.); Fax: +39-049-7923250 (S.M. & F.P.)
| | - Francesco Piazza
- Hematology and Clinical Immunology Branch, Department of Medicine, University of Padova, 35128 Padova, Italy
- Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
- Correspondence: (S.M.); (F.P.); Tel.: +39-049-7923263 (S.M. & F.P.); Fax: +39-049-7923250 (S.M. & F.P.)
| |
Collapse
|
13
|
Manni S, Pesavento M, Spinello Z, Saggin L, Arjomand A, Fregnani A, Quotti Tubi L, Scapinello G, Gurrieri C, Semenzato G, Trentin L, Piazza F. Protein Kinase CK2 represents a new target to boost Ibrutinib and Venetoclax induced cytotoxicity in mantle cell lymphoma. Front Cell Dev Biol 2022; 10:935023. [PMID: 36035991 PMCID: PMC9403710 DOI: 10.3389/fcell.2022.935023] [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: 05/03/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an incurable B cell non-Hodgkin lymphoma, characterized by frequent relapses. In the last decade, the pro-survival pathways related to BCR signaling and Bcl-2 have been considered rational therapeutic targets in B cell derived lymphomas. The BTK inhibitor Ibrutinib and the Bcl-2 inhibitor Venetoclax are emerging as effective drugs for MCL. However, primary and acquired resistance also to these agents may occur. Protein Kinase CK2 is a S/T kinase overexpressed in many solid and blood-derived tumours. CK2 promotes cancer cell growth and clonal expansion, sustaining pivotal survival signaling cascades, such as the ones dependent on AKT, NF-κB, STAT3 and others, counteracting apoptosis through a “non-oncogene” addiction mechanism. We previously showed that CK2 is overexpressed in MCL and regulates the levels of activating phosphorylation on S529 of the NF-κB family member p65/RelA. In the present study, we investigated the effects of CK2 inactivation on MCL cell proliferation, survival and apoptosis and this kinase’s involvement in the BCR and Bcl-2 related signaling. By employing CK2 loss of function MCL cell models, we demonstrated that CK2 sustains BCR signaling (such as BTK, NF-κB and AKT) and the Bcl-2-related Mcl-1 expression. CK2 inactivation enhanced Ibrutinib and Venetoclax-induced cytotoxicity. The demonstration of a CK2-dependent upregulation of pathways that may antagonize the effect of these drugs may offer a novel strategy to overcome primary and secondary resistance.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
- *Correspondence: Sabrina Manni, ; Francesco Piazza,
| | - Maria Pesavento
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Zaira Spinello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Lara Saggin
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Arash Arjomand
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Anna Fregnani
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Laura Quotti Tubi
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Greta Scapinello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Carmela Gurrieri
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Livio Trentin
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Francesco Piazza
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
- *Correspondence: Sabrina Manni, ; Francesco Piazza,
| |
Collapse
|
14
|
Cipak L. Protein Kinases: Function, Substrates, and Implication in Diseases. Int J Mol Sci 2022; 23:ijms23073560. [PMID: 35408921 PMCID: PMC8998185 DOI: 10.3390/ijms23073560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Lubos Cipak
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| |
Collapse
|
15
|
Roth A, Gihring A, Bischof J, Pan L, Oswald F, Knippschild U. CK1 Is a Druggable Regulator of Microtubule Dynamics and Microtubule-Associated Processes. Cancers (Basel) 2022; 14:1345. [PMID: 35267653 PMCID: PMC8909099 DOI: 10.3390/cancers14051345] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/25/2022] [Accepted: 03/03/2022] [Indexed: 02/05/2023] Open
Abstract
Protein kinases of the Casein Kinase 1 family play a vital role in the regulation of numerous cellular processes. Apart from functions associated with regulation of proliferation, differentiation, or apoptosis, localization of several Casein Kinase 1 isoforms to the centrosome and microtubule asters also implicates regulatory functions in microtubule dynamic processes. Being localized to the spindle apparatus during mitosis Casein Kinase 1 directly modulates microtubule dynamics by phosphorylation of tubulin isoforms. Additionally, site-specific phosphorylation of microtubule-associated proteins can be related to the maintenance of genomic stability but also microtubule stabilization/destabilization, e.g., by hyper-phosphorylation of microtubule-associated protein 1A and RITA1. Consequently, approaches interfering with Casein Kinase 1-mediated microtubule-specific functions might be exploited as therapeutic strategies for the treatment of cancer. Currently pursued strategies include the development of Casein Kinase 1 isoform-specific small molecule inhibitors and therapeutically useful peptides specifically inhibiting kinase-substrate interactions.
Collapse
Affiliation(s)
- Aileen Roth
- University Medical Center Ulm, Department of General, and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (A.R.); (A.G.); (J.B.)
| | - Adrian Gihring
- University Medical Center Ulm, Department of General, and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (A.R.); (A.G.); (J.B.)
| | - Joachim Bischof
- University Medical Center Ulm, Department of General, and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (A.R.); (A.G.); (J.B.)
| | - Leiling Pan
- University Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine I, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany;
| | - Franz Oswald
- University Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine I, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany;
| | - Uwe Knippschild
- University Medical Center Ulm, Department of General, and Visceral Surgery, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (A.R.); (A.G.); (J.B.)
| |
Collapse
|
16
|
Manni S, Fregnani A, Quotti Tubi L, Spinello Z, Carraro M, Scapinello G, Visentin A, Barilà G, Pizzi M, Dei Tos AP, Vianello F, Zambello R, Gurrieri C, Semenzato G, Trentin L, Piazza F. Protein Kinase CK1α Sustains B-Cell Receptor Signaling in Mantle Cell Lymphoma. Front Oncol 2021; 11:733848. [PMID: 34722279 PMCID: PMC8551451 DOI: 10.3389/fonc.2021.733848] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/06/2021] [Indexed: 12/25/2022] Open
Abstract
Mantle Cell Lymphoma (MCL) is still an incurable B-cell malignancy characterized by poor prognosis and frequent relapses. B Cell Receptor (BCR) signaling inhibitors, in particular of the kinases BTK and PI3Kγ/δ, have demonstrated clinically meaningful anti-proliferative effects in B cell tumors. However, refractoriness to these drugs may develop, portending a dismal prognosis. Protein kinase CK1α is an emerging pro-growth enzyme in B cell malignancies. In multiple myeloma, this kinase sustains β-catenin and AKT-dependent survival and is involved in the activation of NF-κB in B cells. In this study, we analyzed the role of CK1α on MCL cell survival and proliferation, on the regulation of BCR-related BTK, NF-κB, PI3K/AKT signaling cascades and the effects of CK1α chemical inhibition or gene silencing in association with the BTK inhibitor Ibrutinib or the PI3Kγ/δ inhibitor Duvelisib. CK1α was found highly expressed in MCL cells as compared to normal B cells. The inactivation/loss of CK1α caused MCL cell apoptosis and proliferation arrest. CK1α sustained BCR signaling, in particular the NF-κB, AKT and BTK pathways by modulating the phosphorylation of Ser 652 on CARD11, Ser 536 p65 on NF-κB, Ser 473 on AKT, Tyr 223 on BTK, as well as the protein levels. We also provided evidence that CK1α-mediated regulation of CARD11 and BTK likely implicates a physical interaction. The combination of CK1α inhibition with Ibrutinib or Duvelisib synergistically increased cytotoxicity, leading to a further decrease of the activation of BCR signaling pathways. Therefore, CK1α sustains MCL growth through the regulation of BCR-linked survival signaling cascades and protects from Ibrutinib/Duvelisib-induced apoptosis. Thus, CK1α could be considered as a rational molecular target for the treatment of MCL, in association with novel agents.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Anna Fregnani
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Laura Quotti Tubi
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Zaira Spinello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Marco Carraro
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Greta Scapinello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Andrea Visentin
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Gregorio Barilà
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Marco Pizzi
- Department of Medicine-DIMED, Surgical Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | - Angelo Paolo Dei Tos
- Department of Medicine-DIMED, Surgical Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | - Fabrizio Vianello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
| | - Renato Zambello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Carmela Gurrieri
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Livio Trentin
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Francesco Piazza
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Laboratory of Myeloma and Lymphoma Pathobiology, Veneto Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|