1
|
Li R, Wu X, Cheng J, Zhu Z, Guo M, Hou G, Li T, Zheng Y, Ma H, Lu H, Chen X, Zhang T, Zeng W. Polyamines protect porcine sperm from lipopolysaccharide-induced mitochondrial dysfunction and apoptosis via casein kinase 2 activation. J Anim Sci 2025; 103:skae383. [PMID: 39704338 PMCID: PMC11773192 DOI: 10.1093/jas/skae383] [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: 10/09/2024] [Accepted: 12/18/2024] [Indexed: 12/21/2024] Open
Abstract
Bacterial contamination is an inevitable issue during the processing of semen preservation in pigs. As a prototypical endotoxin from Gram-negative bacteria in semen, lipopolysaccharide (LPS) undermines sperm function during liquid preservation. Spermine and spermidine could protect cells against LPS-induced injury, and the content of spermine and spermidine in seminal plasma is positively correlated with sperm quality. Thus, the present study aimed to clarify whether addition of spermine or spermidine is beneficial to porcine semen preservation and able to prevent LPS-induced sperm damage. The supplementation of spermine and spermidine in the diluent resulted in higher sperm motility, viability, acrosome integrity, and mitochondrial membrane potential (ΔΨm) after preservation in vitro at 17 °C for 7 d (P < 0.05). LPS-induced sperm quality deterioration, ΔΨm decline, cellular adenosine-triphosphate depletion, mitochondrial ultrastructure abnormality, mitochondrial permeability transition pore opening, phosphatidylserine (PS) translocation, and caspase-3 activation (P < 0.05). Interestingly, spermine and spermidine alleviated the LPS-induced changes of the aforementioned parameters and mitigated the decrease in the microtubule-associated protein light chain 3-II (LC3-II) to LC3-I ratio. Meanwhile, the α and β subunits of casein kinase 2 (CK2) were detected at the connecting piece and the tail. Significantly, addition of 4,5,6,7-tetrabromobenzotriazole, a specific CK2 inhibitor, counteracted the beneficial effects of spermine and spermidine on sperm quality, mitochondrial activity, and apoptosis. Together, these results suggest that spermine and spermidine improve sperm quality and the efficiency of liquid preservation of porcine semen. Furthermore, spermine and spermidine alleviate LPS-induced sperm mitochondrial dysfunction and apoptosis in a CK2-dependent manner.
Collapse
Affiliation(s)
- Rongnan Li
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
- College of Life Science, Shanxi Normal University, Taiyuan 030000, Shanxi, China
| | - Xiaodong Wu
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jia Cheng
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723001, Shaanxi, China
| | - Zhendong Zhu
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Ming Guo
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Guochao Hou
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Tianjiao Li
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yi Zheng
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Haidong Ma
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723001, Shaanxi, China
| | - Hongzhao Lu
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723001, Shaanxi, China
| | - Xiaoxu Chen
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Tao Zhang
- School of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong 723001, Shaanxi, China
| | - Wenxian Zeng
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| |
Collapse
|
2
|
Takai T, Asahara SI, Ikushiro H, Kobayashi K, Yano T, Kido Y, Ogawa W. Protective effect of CK2 against endoplasmic reticulum stress in pancreatic β cells. Diabetol Int 2025; 16:131-144. [PMID: 39877448 PMCID: PMC11769914 DOI: 10.1007/s13340-024-00775-w] [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: 03/20/2024] [Accepted: 10/21/2024] [Indexed: 01/31/2025]
Abstract
Endoplasmic reticulum (ER) stress due to obesity or systemic insulin resistance is an important pathogenic factor that could lead to pancreatic β-cell failure. We have previously reported that CCAAT/enhancer-binding protein β (C/EBPβ) is highly induced by ER stress in pancreatic β cells. Moreover, its accumulation hampers the response of these cells to ER stress by inhibiting the induction of the molecular chaperone 78 kDa glucose-regulated protein (GRP78). We also demonstrated that C/EBPβ is phosphorylated by CK2, which is reportedly associated with the ER stress signal. In the present study, we aimed to clarify the mechanisms underlying the effect of CK2 on pancreatic β cells using a CK2-specific inhibitor, CX4945, and shRNA-mediated knockdown and overexpression of CK2β, the regulatory subunit of CK2. The results indicate that CK2 was activated in MIN6 cells under ER stress and in pancreatic β cells of a diabetic mouse model. Under normal conditions, CK2 interacted with FL-ATF6α in MIN6 cells and regulated the expression of GRP78 and ERAD-associated proteins. Mechanistically, CK2 activation in MIN6 cells upon the overexpression of the CK2β subunit reduced ER stress signals and the accumulation of unfolded proteins via an increase in GRP78 and ERAD-associated protein levels. These results highlight the important role of CK2 in protecting against ER stress-induced apoptosis by regulating GRP78 and ERAD proteins. CK2 contributed to the clearance of unfolded or misfolded proteins in MIN6 cells under both normal and ER stress conditions. Therefore, CK2 activation could be a promising novel approach for preventing type 2 diabetes. Supplementary Information The online version contains supplementary material available at 10.1007/s13340-024-00775-w.
Collapse
Affiliation(s)
- Tomoko Takai
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
| | - Shun-ichiro Asahara
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
| | - Hiroko Ikushiro
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Japan
| | - Takato Yano
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Yoshiaki Kido
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
| |
Collapse
|
3
|
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
|
4
|
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: 25] [Impact Index Per Article: 12.5] [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
|
5
|
Bennett CF, Latorre-Muro P, Puigserver P. Mechanisms of mitochondrial respiratory adaptation. Nat Rev Mol Cell Biol 2022; 23:817-835. [PMID: 35804199 PMCID: PMC9926497 DOI: 10.1038/s41580-022-00506-6] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 02/07/2023]
Abstract
Mitochondrial energetic adaptations encompass a plethora of conserved processes that maintain cell and organismal fitness and survival in the changing environment by adjusting the respiratory capacity of mitochondria. These mitochondrial responses are governed by general principles of regulatory biology exemplified by changes in gene expression, protein translation, protein complex formation, transmembrane transport, enzymatic activities and metabolite levels. These changes can promote mitochondrial biogenesis and membrane dynamics that in turn support mitochondrial respiration. The main regulatory components of mitochondrial energetic adaptation include: the transcription coactivator peroxisome proliferator-activated receptor-γ (PPARγ) coactivator 1α (PGC1α) and associated transcription factors; mTOR and endoplasmic reticulum stress signalling; TOM70-dependent mitochondrial protein import; the cristae remodelling factors, including mitochondrial contact site and cristae organizing system (MICOS) and OPA1; lipid remodelling; and the assembly and metabolite-dependent regulation of respiratory complexes. These adaptive molecular and structural mechanisms increase respiration to maintain basic processes specific to cell types and tissues. Failure to execute these regulatory responses causes cell damage and inflammation or senescence, compromising cell survival and the ability to adapt to energetically demanding conditions. Thus, mitochondrial adaptive cellular processes are important for physiological responses, including to nutrient availability, temperature and physical activity, and their failure leads to diseases associated with mitochondrial dysfunction such as metabolic and age-associated diseases and cancer.
Collapse
Affiliation(s)
- Christopher F Bennett
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Pedro Latorre-Muro
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Pere Puigserver
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
6
|
Chen L, Zhang S, Li Q, Li J, Deng H, Zhang S, Meng R. Emerging role of Protein Kinase CK2 in Tumor immunity. Front Oncol 2022; 12:1065027. [DOI: 10.3389/fonc.2022.1065027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022] Open
Abstract
Protein kinase CK2, a conserved serine/threonine-protein kinase, is ubiquitous in cells and regulates various intracellular processes, especially in tumor cells. As one of the earliest discovered protein kinases in humans, CK2 plays a crucial role in phosphorylating or associating with hundreds of substrates to modulate several signaling pathways. Excellent reviews have reported that the overexpression of CK2 could be observed in many cancers and was closely associated with tumor occurrence and development. The elevation of CK2 is also an indicator of a poor prognosis. Recently, increasing attention has been paid to the relationship between CK2 and tumor immunity. However, there is no comprehensive description of how CK2 regulates the immune cells in the tumor microenvironment (TME). Also, the underlying mechanisms are still not very clear. In this review, we systematically summarized the correlation between CK2 and tumor immunity, primarily the effects on various immune cells, both in innate and adaptive immunity in the TME. With the comprehensive development of immunotherapy and the mounting transformation research of CK2 inhibitors from the bench to the clinic, this review will provide vital information to find new treatment options for enhancing the efficacy of immunotherapy.
Collapse
|
7
|
Caracciolo D, Juli G, Riillo C, Coricello A, Vasile F, Pollastri S, Rocca R, Scionti F, Polerà N, Grillone K, Arbitrio M, Staropoli N, Caparello B, Britti D, Loprete G, Costa G, Di Martino MT, Alcaro S, Tagliaferri P, Tassone P. Exploiting DNA Ligase III addiction of multiple myeloma by flavonoid Rhamnetin. Lab Invest 2022; 20:482. [PMID: 36273153 PMCID: PMC9588242 DOI: 10.1186/s12967-022-03705-z] [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] [Received: 09/16/2022] [Accepted: 10/11/2022] [Indexed: 11/28/2022]
Abstract
Background DNA ligases are crucial for DNA repair and cell replication since they catalyze the final steps in which DNA breaks are joined. DNA Ligase III (LIG3) exerts a pivotal role in Alternative-Non-Homologous End Joining Repair (Alt-NHEJ), an error-prone DNA repair pathway often up-regulated in genomically unstable cancer, such as Multiple Myeloma (MM). Based on the three-dimensional (3D) LIG3 structure, we performed a computational screening to identify LIG3-targeting natural compounds as potential candidates to counteract Alt-NHEJ activity in MM. Methods Virtual screening was conducted by interrogating the Phenol Explorer database. Validation of binding to LIG3 recombinant protein was performed by Saturation Transfer Difference (STD)—nuclear magnetic resonance (NMR) experiments. Cell viability was analyzed by Cell Titer-Glo assay; apoptosis was evaluated by flow cytometric analysis following Annexin V-7AAD staining. Alt-NHEJ repair modulation was evaluated using plasmid re-joining assay and Cytoscan HD. DNA Damage Response protein levels were analyzed by Western blot of whole and fractionated protein extracts and immunofluorescence analysis. The mitochondrial DNA (mtDNA) copy number was determined by qPCR. In vivo activity was evaluated in NOD-SCID mice subcutaneously engrafted with MM cells. Results Here, we provide evidence that a natural flavonoid Rhamnetin (RHM), selected by a computational approach, counteracts LIG3 activity and killed Alt-NHEJ-dependent MM cells. Indeed, Nuclear Magnetic Resonance (NMR) showed binding of RHM to LIG3 protein and functional experiments revealed that RHM interferes with LIG3-driven nuclear and mitochondrial DNA repair, leading to significant anti-MM activity in vitro and in vivo. Conclusion Taken together, our findings provide proof of concept that RHM targets LIG3 addiction in MM and may represent therefore a novel promising anti-tumor natural agent to be investigated in an early clinical setting. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03705-z.
Collapse
Affiliation(s)
- Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Giada Juli
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Caterina Riillo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Adriana Coricello
- Department of Health Science, Magna Græcia University, Catanzaro, Italy.,Net4Science Academic Spin-Off, Magna Græcia University, Campus "Salvatore Venuta", Catanzaro, Italy
| | | | - Sara Pollastri
- Department of Chemistry, University of Milan, Milan, Italy
| | - Roberta Rocca
- Department of Health Science, Magna Græcia University, Catanzaro, Italy.,Net4Science Academic Spin-Off, Magna Græcia University, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Francesca Scionti
- Institute of Research and Biomedical Innovation (IRIB), Italian National Council (CNR), Messina, Italy
| | - Nicoletta Polerà
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Katia Grillone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Mariamena Arbitrio
- Institute of Research and Biomedical Innovation (IRIB), Italian National Council (CNR), Catanzaro, Italy
| | | | - Basilio Caparello
- Presidio Ospedaliero Giovanni Paolo II Lamezia Terme, Catanzaro, Italy
| | - Domenico Britti
- Department of Health Science, Magna Græcia University, Catanzaro, Italy
| | - Giovanni Loprete
- Department of Health Science, Magna Græcia University, Catanzaro, Italy
| | - Giosuè Costa
- Department of Health Science, Magna Græcia University, Catanzaro, Italy.,Net4Science Academic Spin-Off, Magna Græcia University, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Stefano Alcaro
- Department of Health Science, Magna Græcia University, Catanzaro, Italy.,Net4Science Academic Spin-Off, Magna Græcia University, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy. .,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Gauff RPM, Lejeusne C, Greff S, Loisel S, Bohner O, Davoult D. Impact of in Situ Simulated Climate Change on Communities and Non-Indigenous Species: Two Climates, Two Responses. J Chem Ecol 2022; 48:761-771. [PMID: 36100819 DOI: 10.1007/s10886-022-01380-4] [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: 04/19/2022] [Revised: 07/21/2022] [Accepted: 08/05/2022] [Indexed: 11/24/2022]
Abstract
Climate change constitutes a major challenge for marine urban ecosystems and ocean warming will likely strongly affect local communities. Non-Indigenous Species (NIS) have been shown to often have higher heat resistance than natives, but studies investigating how forthcoming global warming might affect them in marine urban environments remain scarce, especially in Situ studies. Here we used an in Situ warming experiment in a NW Mediterranean (warm temperate) and a NE Atlantic (cold temperate) marina to see how global warming might affect recruited communities in the near future. In both marinas, warming resulted in significantly different community structure, lower biomass, and more empty space compared to control. However, while in the warm temperate marina, NIS showed an increased surface cover, it was reduced in the cold temperate one. Metabolomic analyses on Bugula neritina in the Atlantic marina revealed potential heat stress experienced by this introduced bryozoan and a potential link between heat stress and the expression of a halogenated alkaloid, Caelestine A. The present results might indicate that the effects of global warming on the prevalence of NIS may differ between geographical provinces, which could be investigated by larger scale studies.
Collapse
Affiliation(s)
- Robin P M Gauff
- Adaptation et Diversité en Milieu Marin, Sorbonne Université, CNRS, UMR 7144, Station Biologique Roscoff, Place Georges Teissier, 29680, Roscoff, France.
| | - Christophe Lejeusne
- Aix Marseille Univ, CNRS, IRD, Avignon Université, IMBE, UMR 7263, Station Marine d'Endoume, Rue de la Batterie des Lions, 13007, Marseille, France
| | - Stephane Greff
- Aix Marseille Univ, CNRS, IRD, Avignon Université, IMBE, UMR 7263, Station Marine d'Endoume, Rue de la Batterie des Lions, 13007, Marseille, France
| | - Stephane Loisel
- Adaptation et Diversité en Milieu Marin, Sorbonne Université, CNRS, UMR 7144, Station Biologique Roscoff, Place Georges Teissier, 29680, Roscoff, France
| | - Olivier Bohner
- Adaptation et Diversité en Milieu Marin, Sorbonne Université, CNRS, UMR 7144, Station Biologique Roscoff, Place Georges Teissier, 29680, Roscoff, France
| | - Dominique Davoult
- Adaptation et Diversité en Milieu Marin, Sorbonne Université, CNRS, UMR 7144, Station Biologique Roscoff, Place Georges Teissier, 29680, Roscoff, France
| |
Collapse
|
9
|
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: 18] [Impact Index Per Article: 6.0] [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
|
10
|
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
|
11
|
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: 1.0] [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
|
12
|
Liu Q, Zhang G, Ji Z, Lin H. Molecular and cellular mechanisms of spastin in neural development and disease (Review). Int J Mol Med 2021; 48:218. [PMID: 34664680 PMCID: PMC8547542 DOI: 10.3892/ijmm.2021.5051] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/29/2021] [Indexed: 12/26/2022] Open
Abstract
Spastin is a microtubule (MT)‑severing enzyme identified from mutations of hereditary spastic paraplegia in 1999 and extensive studies indicate its vital role in various cellular activities. In the past two decades, efforts have been made to understand the underlying molecular mechanisms of how spastin is linked to neural development and disease. Recent studies on spastin have unraveled the mechanistic processes of its MT‑severing activity and revealed that spastin acts as an MT amplifier to mediate its remodeling, thus providing valuable insight into the molecular roles of spastin under physiological conditions. In addition, recent research has revealed multiple novel molecular mechanisms of spastin in cellular biological pathways, including endoplasmic reticulum shaping, calcium trafficking, fatty acid trafficking, as well as endosomal fission and trafficking. These processes are closely involved in axonal and dendritic development and maintenance. The current review presents recent biological advances regarding the molecular mechanisms of spastin at the cellular level and provides insight into how it affects neural development and disease.
Collapse
Affiliation(s)
- Qiuling Liu
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Guowei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhisheng Ji
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Hongsheng Lin
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
13
|
Protein kinase CK2: a potential therapeutic target for diverse human diseases. Signal Transduct Target Ther 2021; 6:183. [PMID: 33994545 PMCID: PMC8126563 DOI: 10.1038/s41392-021-00567-7] [Citation(s) in RCA: 195] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/04/2023] Open
Abstract
CK2 is a constitutively active Ser/Thr protein kinase, which phosphorylates hundreds of substrates, controls several signaling pathways, and is implicated in a plethora of human diseases. Its best documented role is in cancer, where it regulates practically all malignant hallmarks. Other well-known functions of CK2 are in human infections; in particular, several viruses exploit host cell CK2 for their life cycle. Very recently, also SARS-CoV-2, the virus responsible for the COVID-19 pandemic, has been found to enhance CK2 activity and to induce the phosphorylation of several CK2 substrates (either viral and host proteins). CK2 is also considered an emerging target for neurological diseases, inflammation and autoimmune disorders, diverse ophthalmic pathologies, diabetes, and obesity. In addition, CK2 activity has been associated with cardiovascular diseases, as cardiac ischemia-reperfusion injury, atherosclerosis, and cardiac hypertrophy. The hypothesis of considering CK2 inhibition for cystic fibrosis therapies has been also entertained for many years. Moreover, psychiatric disorders and syndromes due to CK2 mutations have been recently identified. On these bases, CK2 is emerging as an increasingly attractive target in various fields of human medicine, with the advantage that several very specific and effective inhibitors are already available. Here, we review the literature on CK2 implication in different human pathologies and evaluate its potential as a pharmacological target in the light of the most recent findings.
Collapse
|
14
|
Spinello Z, Fregnani A, Quotti Tubi L, Trentin L, Piazza F, Manni S. Targeting Protein Kinases in Blood Cancer: Focusing on CK1α and CK2. Int J Mol Sci 2021; 22:ijms22073716. [PMID: 33918307 PMCID: PMC8038136 DOI: 10.3390/ijms22073716] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023] Open
Abstract
Disturbance of protein kinase activity may result in dramatic consequences that often lead to cancer development and progression. In tumors of blood origin, both tyrosine kinases and serine/threonine kinases are altered by different types of mutations, critically regulating cancer hallmarks. CK1α and CK2 are highly conserved, ubiquitously expressed and constitutively active pleiotropic kinases, which participate in multiple biological processes. The involvement of these kinases in solid and blood cancers is well documented. CK1α and CK2 are overactive in multiple myeloma, leukemias and lymphomas. Intriguingly, they are not required to the same degree for the viability of normal cells, corroborating the idea of “druggable” kinases. Different to other kinases, mutations on the gene encoding CK1α and CK2 are rare or not reported. Actually, these two kinases are outside the paradigm of oncogene addiction, since cancer cells’ dependency on these proteins resembles the phenomenon of “non-oncogene” addiction. In this review, we will summarize the general features of CK1α and CK2 and the most relevant oncogenic and stress-related signaling nodes, regulated by kinase phosphorylation, that may lead to tumor progression. Finally, we will report the current data, which support the positioning of these two kinases in the therapeutic scene of hematological cancers.
Collapse
Affiliation(s)
- Zaira Spinello
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
| | - Anna Fregnani
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
| | - Laura Quotti Tubi
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
| | - Livio Trentin
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
| | - Francesco Piazza
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
- Correspondence: (F.P.); (S.M.); Tel.: +39-049-792-3263 (F.P. & S.M.); Fax: +39-049-792-3250 (F.P. & S.M.)
| | - Sabrina Manni
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
- Correspondence: (F.P.); (S.M.); Tel.: +39-049-792-3263 (F.P. & S.M.); Fax: +39-049-792-3250 (F.P. & S.M.)
| |
Collapse
|
15
|
Rosales M, Rodríguez-Ulloa A, Besada V, Ramón AC, Pérez GV, Ramos Y, Guirola O, González LJ, Zettl K, Wiśniewski JR, Perera Y, Perea SE. Phosphoproteomic Landscape of AML Cells Treated with the ATP-Competitive CK2 Inhibitor CX-4945. Cells 2021; 10:cells10020338. [PMID: 33562780 PMCID: PMC7915770 DOI: 10.3390/cells10020338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/30/2021] [Accepted: 02/02/2021] [Indexed: 12/13/2022] Open
Abstract
Casein kinase 2 (CK2) regulates a plethora of proteins with pivotal roles in solid and hematological neoplasia. Particularly, in acute myeloid leukemia (AML) CK2 has been pointed as an attractive therapeutic target and prognostic marker. Here, we explored the impact of CK2 inhibition over the phosphoproteome of two cell lines representing major AML subtypes. Quantitative phosphoproteomic analysis was conducted to evaluate changes in phosphorylation levels after incubation with the ATP-competitive CK2 inhibitor CX-4945. Functional enrichment, network analysis, and database mining were performed to identify biological processes, signaling pathways, and CK2 substrates that are responsive to CX-4945. A total of 273 and 1310 phosphopeptides were found differentially modulated in HL-60 and OCI-AML3 cells, respectively. Despite regulated phosphopeptides belong to proteins involved in multiple biological processes and signaling pathways, most of these perturbations can be explain by direct CK2 inhibition rather than off-target effects. Furthermore, CK2 substrates regulated by CX-4945 are mainly related to mRNA processing, translation, DNA repair, and cell cycle. Overall, we evidenced that CK2 inhibitor CX-4945 impinge on mediators of signaling pathways and biological processes essential for primary AML cells survival and chemosensitivity, reinforcing the rationale behind the pharmacologic blockade of protein kinase CK2 for AML targeted therapy.
Collapse
Affiliation(s)
- Mauro Rosales
- Department of Animal and Human Biology, Faculty of Biology, University of Havana (UH), Havana 10400, Cuba;
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, Center for Genetic Engineering and Biotechnology (CIGB), Havana 10600, Cuba; (A.C.R.); (G.V.P.)
| | - Arielis Rodríguez-Ulloa
- Mass Spectrometry Laboratory, Proteomics Group, Department of Systems Biology, Biomedical Research Division, CIGB, Havana 10600, Cuba; (A.R.-U.); (V.B.); (Y.R.); (L.J.G.)
| | - Vladimir Besada
- Mass Spectrometry Laboratory, Proteomics Group, Department of Systems Biology, Biomedical Research Division, CIGB, Havana 10600, Cuba; (A.R.-U.); (V.B.); (Y.R.); (L.J.G.)
| | - Ailyn C. Ramón
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, Center for Genetic Engineering and Biotechnology (CIGB), Havana 10600, Cuba; (A.C.R.); (G.V.P.)
| | - George V. Pérez
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, Center for Genetic Engineering and Biotechnology (CIGB), Havana 10600, Cuba; (A.C.R.); (G.V.P.)
| | - Yassel Ramos
- Mass Spectrometry Laboratory, Proteomics Group, Department of Systems Biology, Biomedical Research Division, CIGB, Havana 10600, Cuba; (A.R.-U.); (V.B.); (Y.R.); (L.J.G.)
| | - Osmany Guirola
- Bioinformatics Group, Department of Systems Biology, Biomedical Research Division, CIGB, Havana 10600, Cuba;
| | - Luis J. González
- Mass Spectrometry Laboratory, Proteomics Group, Department of Systems Biology, Biomedical Research Division, CIGB, Havana 10600, Cuba; (A.R.-U.); (V.B.); (Y.R.); (L.J.G.)
| | - Katharina Zettl
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, 82152 Munich, Germany; (K.Z.); (J.R.W.)
| | - Jacek R. Wiśniewski
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, 82152 Munich, Germany; (K.Z.); (J.R.W.)
| | - Yasser Perera
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, Center for Genetic Engineering and Biotechnology (CIGB), Havana 10600, Cuba; (A.C.R.); (G.V.P.)
- China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Zhong Gu Biotechnology Co., Ltd, Lengshuitan District, Yongzhou 425000, China
- Correspondence: (Y.P.); (S.E.P.)
| | - Silvio E. Perea
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, Center for Genetic Engineering and Biotechnology (CIGB), Havana 10600, Cuba; (A.C.R.); (G.V.P.)
- Correspondence: (Y.P.); (S.E.P.)
| |
Collapse
|
16
|
Protein kinase CK2 inhibition as a pharmacological strategy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 124:23-46. [PMID: 33632467 DOI: 10.1016/bs.apcsb.2020.09.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
CK2 is a constitutively active Ser/Thr protein kinase which phosphorylates hundreds of substrates. Since they are primarily related to survival and proliferation pathways, the best-known pathological roles of CK2 are in cancer, where its targeting is currently being considered as a possible therapy. However, CK2 activity has been found instrumental in many other human pathologies, and its inhibition will expectably be extended to different purposes in the near future. Here, after a description of CK2 features and implications in diseases, we analyze the different inhibitors and strategies available to target CK2, and update the results so far obtained by their in vivo application.
Collapse
|
17
|
Stengel ST, Fazio A, Lipinski S, Jahn MT, Aden K, Ito G, Wottawa F, Kuiper JW, Coleman OI, Tran F, Bordoni D, Bernardes JP, Jentzsch M, Luzius A, Bierwirth S, Messner B, Henning A, Welz L, Kakavand N, Falk-Paulsen M, Imm S, Hinrichsen F, Zilbauer M, Schreiber S, Kaser A, Blumberg R, Haller D, Rosenstiel P. Activating Transcription Factor 6 Mediates Inflammatory Signals in Intestinal Epithelial Cells Upon Endoplasmic Reticulum Stress. Gastroenterology 2020; 159:1357-1374.e10. [PMID: 32673694 PMCID: PMC7923714 DOI: 10.1053/j.gastro.2020.06.088] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 05/18/2020] [Accepted: 06/18/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Excess and unresolved endoplasmic reticulum (ER) stress in intestinal epithelial cells (IECs) promotes intestinal inflammation. Activating transcription factor 6 (ATF6) is one of the signaling mediators of ER stress. We studied the pathways that regulate ATF6 and its role for inflammation in IECs. METHODS We performed an RNA interference screen, using 23,349 unique small interfering RNAs targeting 7783 genes and a luciferase reporter controlled by an ATF6-dependent ERSE (ER stress-response element) promoter, to identify proteins that activate or inhibit the ATF6 signaling pathway in HEK293 cells. To validate the screening results, intestinal epithelial cell lines (Caco-2 cells) were transfected with small interfering RNAs or with a plasmid overexpressing a constitutively active form of ATF6. Caco-2 cells with a CRISPR-mediated disruption of autophagy related 16 like 1 gene (ATG16L1) were used to study the effect of ATF6 on ER stress in autophagy-deficient cells. We also studied intestinal organoids derived from mice that overexpress constitutively active ATF6, from mice with deletion of the autophagy related 16 like 1 or X-Box binding protein 1 gene in IECs (Atg16l1ΔIEC or Xbp1ΔIEC, which both develop spontaneous ileitis), from patients with Crohn's disease (CD) and healthy individuals (controls). Cells and organoids were incubated with tunicamycin to induce ER stress and/or chemical inhibitors of newly identified activator proteins of ATF6 signaling, and analyzed by real-time polymerase chain reaction and immunoblots. Atg16l1ΔIEC and control (Atg16l1fl/fl) mice were given intraperitoneal injections of tunicamycin and were treated with chemical inhibitors of ATF6 activating proteins. RESULTS We identified and validated 15 suppressors and 7 activators of the ATF6 signaling pathway; activators included the regulatory subunit of casein kinase 2 (CSNK2B) and acyl-CoA synthetase long chain family member 1 (ACSL1). Knockdown or chemical inhibition of CSNK2B and ACSL1 in Caco-2 cells reduced activity of the ATF6-dependent ERSE reporter gene, diminished transcription of the ATF6 target genes HSP90B1 and HSPA5 and reduced NF-κB reporter gene activation on tunicamycin stimulation. Atg16l1ΔIEC and or Xbp1ΔIEC organoids showed increased expression of ATF6 and its target genes. Inhibitors of ACSL1 or CSNK2B prevented activation of ATF6 and reduced CXCL1 and tumor necrosis factor (TNF) expression in these organoids on induction of ER stress with tunicamycin. Injection of mice with inhibitors of ACSL1 or CSNK2B significantly reduced tunicamycin-mediated intestinal inflammation and IEC death and expression of CXCL1 and TNF in Atg16l1ΔIEC mice. Purified ileal IECs from patients with CD had higher levels of ATF6, CSNK2B, and HSPA5 messenger RNAs than controls; early-passage organoids from patients with active CD show increased levels of activated ATF6 protein, incubation of these organoids with inhibitors of ACSL1 or CSNK2B reduced transcription of ATF6 target genes, including TNF. CONCLUSIONS Ileal IECs from patients with CD have higher levels of activated ATF6, which is regulated by CSNK2B and HSPA5. ATF6 increases expression of TNF and other inflammatory cytokines in response to ER stress in these cells and in organoids from Atg16l1ΔIEC and Xbp1ΔIEC mice. Strategies to inhibit the ATF6 signaling pathway might be developed for treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Stephanie T. Stengel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Antonella Fazio
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Simone Lipinski
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Martin T. Jahn
- RD3 Marine Microbiology, GEOMAR Helmholtz Centre for Ocean Research Kiel, Germany
| | - Konrad Aden
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany,Department of Internal Medicine I., Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Go Ito
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany,Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Felix Wottawa
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Jan W.P. Kuiper
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Olivia I. Coleman
- Chair of Nutrition and Immunology, Technische Universität München, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Florian Tran
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany,Department of Internal Medicine I., Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Dora Bordoni
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Joana P. Bernardes
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Marlene Jentzsch
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Anne Luzius
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Sandra Bierwirth
- Chair of Nutrition and Immunology, Technische Universität München, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Berith Messner
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Anna Henning
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Lina Welz
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Nassim Kakavand
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Maren Falk-Paulsen
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Simon Imm
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Finn Hinrichsen
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Matthias Zilbauer
- Department of Pediatrics, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, England, UK MA
| | - Stefan Schreiber
- Department of Internal Medicine I., Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, England, UK MA
| | - Richard Blumberg
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, US
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technische Universität München, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
| |
Collapse
|
18
|
Manni S, Fregnani A, Barilà G, Zambello R, Semenzato G, Piazza F. Actionable Strategies to Target Multiple Myeloma Plasma Cell Resistance/Resilience to Stress: Insights From "Omics" Research. Front Oncol 2020; 10:802. [PMID: 32500036 PMCID: PMC7243738 DOI: 10.3389/fonc.2020.00802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/23/2020] [Indexed: 12/20/2022] Open
Abstract
While the modern therapeutic armamentarium to treat multiple myeloma (MM) patients allows a longer control of the disease, this second-most-frequent hematologic cancer is still uncurable in the vast majority of cases. Since MM plasma cells are subjected to various types of chronic cellular stress and the integrity of specific stress-coping pathways is essential to ensure MM cell survival, not surprisingly the most efficacious anti-MM therapy are those that make use of proteasome inhibitors and/or immunomodulatory drugs, which target the biochemical mechanisms of stress management. Based on this notion, the recently realized discoveries on MM pathobiology through high-throughput techniques (genomic, transcriptomic, and other "omics"), in order for them to be clinically useful, should be elaborated to identify novel vulnerabilities in this disease. This groundwork of information will likely allow the design of novel therapies against targetable molecules/pathways, in an unprecedented opportunity to change the management of MM according to the principle of "precision medicine." In this review, we will discuss some examples of therapeutically actionable molecules and pathways related to the regulation of cellular fitness and stress resistance in MM.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Foundation for Advanced Biomedical Research – Veneto Institute of Molecular Medicine (FABR-VIMM), Padova, Italy
| | - Anna Fregnani
- Foundation for Advanced Biomedical Research – Veneto Institute of Molecular Medicine (FABR-VIMM), Padova, Italy
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padova, Padova, Italy
| | - Gregorio Barilà
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Foundation for Advanced Biomedical Research – Veneto Institute of Molecular Medicine (FABR-VIMM), Padova, Italy
| | - Renato Zambello
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Foundation for Advanced Biomedical Research – Veneto Institute of Molecular Medicine (FABR-VIMM), Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Foundation for Advanced Biomedical Research – Veneto Institute of Molecular Medicine (FABR-VIMM), Padova, Italy
| | - Francesco Piazza
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
- Foundation for Advanced Biomedical Research – Veneto Institute of Molecular Medicine (FABR-VIMM), Padova, Italy
| |
Collapse
|
19
|
Al-Romaiyan A, Liu B, Persaud S, Jones P. A novel Gymnema sylvestre extract protects pancreatic beta-cells from cytokine-induced apoptosis. Phytother Res 2019; 34:161-172. [PMID: 31515869 DOI: 10.1002/ptr.6512] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 08/04/2019] [Accepted: 08/29/2019] [Indexed: 01/17/2023]
Abstract
Inflammatory cytokines such as interleukin-1β, TNF-α, and interferon-γ are known to be involved in mediating β-cells death in diabetes mellitus (DM). Thus, protecting from β-cells death in patients with DM may be a useful target in alleviating symptoms of hyperglycemia. Traditional plant-based remedies have been used to treat DM for many centuries and may play a role in protecting β-cell from death. An example of these remedies is Gymnema sylvestre (GS) extract. In this study, we investigated the effect of this plant extract on β-cells apoptosis. Om Santal Adivasi (OSA®) maintained cell membrane integrity in MIN6 cells and mouse islets. Om Santal Adivasi significantly protected MIN6 cells and mouse islets from cytokine-induced apoptosis. In the presence of cytokines, OSA® significantly reduced the expression and activity of caspase-3. The antiapoptotic effect of OSA® as shown by microarray analysis is largely mediated by activating pathways involved in cell survival (mainly casein kinase II pathway) and the free radical scavenger system (specifically superoxide dismutase and catalase). This study indicates that the GS isolate OSA® protects against cytokine-induced apoptosis of β-cells by increasing the expression of cell survival pathways and free radical scavenger system.
Collapse
Affiliation(s)
- Altaf Al-Romaiyan
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University, Kuwait
| | - Bo Liu
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, King's College London, London, UK
| | - Shanta Persaud
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, King's College London, London, UK
| | - Peter Jones
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, King's College London, London, UK
| |
Collapse
|
20
|
Lind J, Czernilofsky F, Vallet S, Podar K. Emerging protein kinase inhibitors for the treatment of multiple myeloma. Expert Opin Emerg Drugs 2019; 24:133-152. [PMID: 31327278 DOI: 10.1080/14728214.2019.1647165] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Significant advances have been made during the last two decades in terms of new therapeutic options but also of innovative approaches to diagnosis and management of multiple myeloma (MM). While patient survival has been significantly prolonged, most patients relapse. Including the milestone approval of the first kinase inhibitor imatinib mesylate for CML in 2001, 48 small molecule protein kinase (PK) inhibitors have entered clinical practice until now. However, no PK inhibitor has been approved for MM therapy yet. Areas covered: This review article summarizes up-to-date knowledge on the pathophysiologic role of PKs in MM. Derived small molecules targeting receptor tyrosine kinases (RTKs), the Ras/Raf/MEK/MAPK- pathway, the PI3K/Akt/mTOR- pathway as well as Bruton tyrosine kinase (BTK), Aurora kinases (AURK), and cyclin-dependent kinases (CDKs) are most promising. Preclinical as well as early clinical data focusing on these molecules will be presented and critically reviewed. Expert opinion: Current MM therapy is directed against general vulnerabilities. Novel therapeutic strategies, inhibition of PKs in particular, are directed to target tumor-specific driver aberrations such as genetic abnormalities and microenvironment-driven deregulations. Results of ongoing Precision Medicine trials with PK inhibitors alone or in combination with other agents are eagerly awaited and hold the promise of once more improving MM patient outcome.
Collapse
Affiliation(s)
- Judith Lind
- Department of Internal Medicine II, University Hospital Krems, Karl Landsteiner University of Health Sciences , Krems an der Donau , Austria
| | - Felix Czernilofsky
- Department of Internal Medicine II, University Hospital Krems, Karl Landsteiner University of Health Sciences , Krems an der Donau , Austria
| | - Sonia Vallet
- Department of Internal Medicine II, University Hospital Krems, Karl Landsteiner University of Health Sciences , Krems an der Donau , Austria
| | - Klaus Podar
- Department of Internal Medicine II, University Hospital Krems, Karl Landsteiner University of Health Sciences , Krems an der Donau , Austria
| |
Collapse
|
21
|
Borgo C, Ruzzene M. Role of protein kinase CK2 in antitumor drug resistance. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:287. [PMID: 31277672 PMCID: PMC6612148 DOI: 10.1186/s13046-019-1292-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 06/25/2019] [Indexed: 01/21/2023]
Abstract
Drug resistance represents the major reason of pharmacological treatment failure. It is supported by a broad spectrum of mechanisms, whose molecular bases have been frequently correlated to aberrant protein phosphorylation. CK2 is a constitutively active protein kinase which phosphorylates hundreds of substrates; it is expressed in all cells, but its level is commonly found higher in cancer cells, where it plays anti-apoptotic, pro-migration and pro-proliferation functions. Several evidences support a role for CK2 in processes directly responsible of drug resistance, such as drug efflux and DNA repair; moreover, CK2 intervenes in signaling pathways which are crucial to evade drug response (as PI3K/AKT/PTEN, NF-κB, β-catenin, hedgehog signaling, p53), and controls the activity of chaperone machineries fundamental in resistant cells. Interestingly, a panel of specific and effective inhibitors of CK2 is available, and several examples are known of their efficacy in resistant cells, with synergistic effect when used in combination with conventional drugs, also in vivo. Here we analyze and discuss evidences supporting the hypothesis that CK2 targeting represents a valuable strategy to overcome drug resistance.
Collapse
Affiliation(s)
- Christian Borgo
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58b, 35131, Padova, Italy
| | - Maria Ruzzene
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58b, 35131, Padova, Italy.
| |
Collapse
|
22
|
Peli1 induction impairs cardiac microvascular endothelium through Hsp90 dissociation from IRE1α. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2606-2617. [PMID: 31260751 DOI: 10.1016/j.bbadis.2019.06.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/10/2019] [Accepted: 06/25/2019] [Indexed: 12/11/2022]
Abstract
Ameliorating cardiac microvascular injury is the most effective means to mitigate diabetes-induced cardiovascular complications. Inositol-requiring 1α (IRE1α), a sensor of endoplasmic reticulum stress, is activated by Toll like receptors (TLRs), and then promotes cardiac microvascular injury. Peli1 is a master regulator of TLRs and activates IRE1α. This study aims to investigate whether Peli1 in endothelial cells promotes diabetes-induced cardiac microvascular injury through activating IRE1α. Here we found that Peli1 was markedly up-regulated in cardiac endothelial cells of both diabetic mice and in AGEs-treated cardiac microvascular endothelial cells (CMECs). Peli1 deficiency in endothelial cells significantly alleviated diabetes-induced cardiac microvascular permeability, promoted microvascular regeneration, and suppressed apoptosis, accompanied by the attenuation of adverse cardiac remodeling. Furthermore, Peli1 deletion in CMECs ameliorated AGEs-induced damages in vitro. We identified heat shock protein 90 (Hsp90) as a potential binding partner for Peli1, and the Ring domain of Peli1 directly bound with Hsp90 to enhance IRE1α phosphorylation. Our study suggests that blocking Peli1 in endothelial cells may protect against diabetes-induced cardiac microvascular injury by restraining ER stress.
Collapse
|
23
|
CK2 Pro-Survival Role in Prostate Cancer Is Mediated via Maintenance and Promotion of Androgen Receptor and NFκB p65 Expression. Pharmaceuticals (Basel) 2019; 12:ph12020089. [PMID: 31197122 PMCID: PMC6631211 DOI: 10.3390/ph12020089] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 06/05/2019] [Accepted: 06/13/2019] [Indexed: 12/25/2022] Open
Abstract
The prosurvival protein kinase CK2, androgen receptor (AR), and nuclear factor kappa B (NFκB) interact in the function of prostate cells, and there is evidence of crosstalk between these signals in the pathobiology of prostate cancer (PCa). As CK2 is elevated in PCa, and AR and NFκB are involved in the development and progression of prostate cancer, we investigated their interaction in benign and malignant prostate cells in the presence of altered CK2 expression. Our results show that elevation of CK2 levels caused increased levels of AR and NFκB p65 in prostate cells of different phenotypes. Analysis of TCGA PCa data indicated that AR and CK2α RNA expression are strongly correlated. Small molecule inhibition or molecular down-regulation of CK2 caused reduction in AR mRNA expression and protein levels in PCa cells and in orthotopic xenograft tumors by various pathways. Among these, regulation of AR protein stability plays a unifying role in CK2 maintenance of AR protein levels. Our results show induction of various endoplasmic reticulum stress signals after CK2 inhibition, which may play a role in the PCa cell death response. Of note, CK2 inhibition caused loss of cell viability in both parental and enzalutamide-resistant castrate-resistant PCa cells. The present work elucidates the specific link of CK2 to the pathogenesis of PCa in association with AR and NFκB expression; further, the observation that inhibition of CK2 can exert a growth inhibitory effect on therapy-resistant PCa cells emphasizes the potential utility of CK2 inhibition in patients who are on enzalutamide treatment for advanced cancer.
Collapse
|
24
|
Carrino M, Quotti Tubi L, Fregnani A, Canovas Nunes S, Barilà G, Trentin L, Zambello R, Semenzato G, Manni S, Piazza F. Prosurvival autophagy is regulated by protein kinase CK1 alpha in multiple myeloma. Cell Death Discov 2019; 5:98. [PMID: 31123604 PMCID: PMC6529432 DOI: 10.1038/s41420-019-0179-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/09/2019] [Accepted: 04/23/2019] [Indexed: 01/09/2023] Open
Abstract
Multiple myeloma (MM) is a tumor of plasma cells (PCs). Due to the intense immunoglobulin secretion, PCs are prone to endoplasmic reticulum stress and activate several stress-managing pathways, including autophagy. Indeed, autophagy deregulation is maladaptive for MM cells, resulting in cell death. CK1α, a pro-survival kinase in MM, has recently been involved as a regulator of the autophagic flux and of the transcriptional competence of the autophagy-related transcription factor FOXO3a in several cancers. In this study, we investigated the role of CK1α in autophagy in MM. To study the autophagic flux we generated clones of MM cell lines expressing the mCherry-eGFP-LC3B fusion protein. We observed that CK1 inhibition with the chemical ATP-competitive CK1 α/δ inhibitor D4476 resulted in an impaired autophagic flux, likely due to an alteration of lysosomes acidification. However, D4476 caused the accumulation of the transcription factor FOXO3a in the nucleus, and this was paralleled by the upregulation of mRNA coding for autophagic genes. Surprisingly, silencing of CK1α by RNA interference triggered the autophagic flux. However, FOXO3a did not shuttle into the nucleus and the transcription of autophagy-related FOXO3a-dependent genes was not observed. Thus, while the chemical inhibition with the dual CK1α/δ inhibitor D4476 induced cell death as a consequence of an accumulation of ineffective autophagic vesicles, on the opposite, CK1α silencing, although it also determined apoptosis, triggered a full activation of the early autophagic flux, which was then not supported by the upregulation of autophagic genes. Taken together, our results indicate that the family of CK1 kinases may profoundly influence MM cells survival also through the modulation of the autophagic pathway.
Collapse
Affiliation(s)
- Marilena Carrino
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Laura Quotti Tubi
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Anna Fregnani
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Sara Canovas Nunes
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy.,Boston Children's Hospital/Harvard Medical School, Boston, MA USA
| | - Gregorio Barilà
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Livio Trentin
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Renato Zambello
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Gianpietro Semenzato
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Sabrina Manni
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Francesco Piazza
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| |
Collapse
|
25
|
Okayama M, Kitabatake S, Sato M, Fujimori K, Ichikawa D, Matsushita M, Suto Y, Iwasaki G, Yamada T, Kiuchi F, Hirao M, Kunieda H, Osada M, Okamoto S, Hattori Y. A novel derivative (GTN024) from a natural product, komaroviquinone, induced the apoptosis of high-risk myeloma cells via reactive oxygen production and ER stress. Biochem Biophys Res Commun 2018; 505:787-793. [DOI: 10.1016/j.bbrc.2018.09.177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 09/28/2018] [Indexed: 01/16/2023]
|
26
|
Taalab YM, Ibrahim N, Maher A, Hassan M, Mohamed W, Moustafa AA, Salama M, Johar D, Bernstein L. Mechanisms of disordered neurodegenerative function: concepts and facts about the different roles of the protein kinase RNA-like endoplasmic reticulum kinase (PERK). Rev Neurosci 2018; 29:387-415. [PMID: 29303785 DOI: 10.1515/revneuro-2017-0071] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/20/2017] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases, such as Alzheimer's disease, Huntington's disease, Parkinson's disease, prion disease, and amyotrophic lateral sclerosis, are a dissimilar group of disorders that share a hallmark feature of accumulation of abnormal intraneuronal or extraneuronal misfolded/unfolded protein and are classified as protein misfolding disorders. Cellular and endoplasmic reticulum (ER) stress activates multiple signaling cascades of the unfolded protein response (UPR). Consequently, translational and transcriptional alterations in target gene expression occur in response directed toward restoring the ER capacity of proteostasis and reestablishing the cellular homeostasis. Evidences from in vitro and in vivo disease models indicate that disruption of ER homeostasis causes abnormal protein aggregation that leads to synaptic and neuronal dysfunction. However, the exact mechanism by which it contributes to disease progression and pathophysiological changes remains vague. Downstream signaling pathways of UPR are fully integrated, yet with diverse unexpected outcomes in different disease models. Three well-identified ER stress sensors have been implicated in UPR, namely, inositol requiring enzyme 1, protein kinase RNA-activated-like ER kinase (PERK), and activating transcription factor 6. Although it cannot be denied that each of the involved stress sensor initiates a distinct downstream signaling pathway, it becomes increasingly clear that shared pathways are crucial in determining whether or not the UPR will guide the cells toward adaptive prosurvival or proapoptotic responses. We review a body of work on the mechanism of neurodegenerative diseases based on oxidative stress and cell death pathways with emphasis on the role of PERK.
Collapse
Affiliation(s)
- Yasmeen M Taalab
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Al-Mansoura University, Al-Mansoura, 35111, Egypt
| | - Nour Ibrahim
- Faculty of Medicine, Ain Shams University, Cairo, 11591, Egypt
| | - Ahmed Maher
- Zoonotic Disease Department, National Research Center, Dokki, Giza, 25200, Egypt
| | - Mubashir Hassan
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju-do 32588, South Korea
| | - Wael Mohamed
- Department of Clinical Pharmacology, Faculty of Medicine, Al-Menoufia University, Al-Menoufia, 25200 Egypt.,Basic Medical Science Department, Kulliyyah of Medicine, International Islamic University Malaysia, Kunatan Pahang, Malaysia
| | - Ahmed A Moustafa
- School of Social Sciences and Psychology and MARCS Institute for Brain and Behaviour, Western Sydney University, Sydney, New South Wales, 2751 Australia
| | - Mohamed Salama
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Al-Mansoura University, Al-Mansoura, 35111, Egypt.,Medical Experimental Research Center (MERC), Al-Mansoura University, Al-Mansoura, Egypt
| | - Dina Johar
- Department of Biochemistry and Nutrition, Faculty of Women for Arts, Sciences and Education, Ain Shams University, Heliopolis, Cairo, 11291, Egypt.,Max Rady College of Medicine, Rady Faculty of Health Sciences, Department of Physiology & Pathophysiology 432 Basic Medical Sciences Building, 745 Bannatyne Avenue University of Manitoba, Winnipeg, MB R3E 0J9, Canada, e-mail:
| | - Larry Bernstein
- Triplex Consulting, 54 Firethorn Lane, Northampton, MA 01060, USA
| |
Collapse
|
27
|
Canedo-Antelo M, Serrano MP, Manterola A, Ruiz A, Llavero F, Mato S, Zugaza JL, Pérez-Cerdá F, Matute C, Sánchez-Gómez MV. Inhibition of Casein Kinase 2 Protects Oligodendrocytes From Excitotoxicity by Attenuating JNK/p53 Signaling Cascade. Front Mol Neurosci 2018; 11:333. [PMID: 30271323 PMCID: PMC6146035 DOI: 10.3389/fnmol.2018.00333] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/27/2018] [Indexed: 12/12/2022] Open
Abstract
Oligodendrocytes are highly vulnerable to glutamate excitotoxicity, a central mechanism involved in tissue damage in Multiple Sclerosis (MS). Sustained activation of AMPA receptors in rat oligodendrocytes induces cytosolic calcium overload, mitochondrial depolarization, increase of reactive oxygen species, and activation of intracelular pathways resulting in apoptotic cell death. Although many signals driven by excitotoxicity have been identified, some of the key players are still under investigation. Casein kinase 2 (CK2) is a serine/threonine kinase, constitutively expressed in all eukaryotic tissues, involved in cell proliferation, malignant transformation and apoptosis. In this study, we identify CK2 as a critical regulator of oligodendrocytic death pathways and elucidate its role as a signal inductor following excitotoxic insults. We provide evidence that CK2 activity is up-regulated in AMPA-treated oligodendrocytes and CK2 inhibition significantly diminished AMPA receptor-induced oligodendroglial death. In addition, we analyzed mitogen-activated protein kinase (MAPK) signaling after excitotoxic insult. We observed that AMPA receptor activation induced a rapid increase in c-Jun N-terminal kinase (JNK) and p38 phosphorylation that was reduced after CK2 inhibition. Moreover, blocking their phosphorylation, we enhanced oligodendrocyte survival after excitotoxic insult. Finally, we observed that the tumor suppressor p53 is activated during AMPA receptor-induced cell death and, interestingly, down-regulated by JNK or CK2 inhibition. Together, these data indicate that the increase in CK2 activity induced by excitotoxic insults regulates MAPKs, triggers p53 activation and mediates subsequent oligodendroglial loss. Therefore, targeting CK2 may be a useful strategy to prevent oligodendrocyte death in MS and other diseases involving central nervous system (CNS) white matter.
Collapse
Affiliation(s)
- Manuel Canedo-Antelo
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Mari Paz Serrano
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Andrea Manterola
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Asier Ruiz
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Francisco Llavero
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Genética, Antropología Física y Fisiología Animal, Universidad del País Vasco (UPV/EHU), Leioa, Spain
| | - Susana Mato
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - José Luis Zugaza
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Genética, Antropología Física y Fisiología Animal, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Fernando Pérez-Cerdá
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Carlos Matute
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - María Victoria Sánchez-Gómez
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| |
Collapse
|
28
|
Manni S, Carrino M, Semenzato G, Piazza F. Old and Young Actors Playing Novel Roles in the Drama of Multiple Myeloma Bone Marrow Microenvironment Dependent Drug Resistance. Int J Mol Sci 2018; 19:ijms19051512. [PMID: 29783691 PMCID: PMC5983700 DOI: 10.3390/ijms19051512] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma (MM) is the second most frequent hematologic cancer. In addition to the deleterious effects of neoplastic plasma cell growth and spreading during the disease evolution, this tumor is characterized by the serious pathological consequences due to the massive secretion of monoclonal immunoglobulins and by the derangement of bone physiology with progressive weakening of the skeleton. Despite significant progresses having been made in the last two decades in the therapeutic management of this plasma cell tumor, MM remains invariably lethal, due to its extremely complex genetic architecture and to the constant protection it receives from the tumor niche, which is represented by the bone marrow microenvironment. While it is predictable that the discovery of novel therapies against the first of these two pathobiological features will take a longer time, the identification of the cellular and molecular mechanisms underlying the pro-growth effects of the myeloma milieu is a task that could lead to the development of novel treatments in a shorter timeframe. In this regard, aside from known “old” determinants of the cross-talk between bone marrow and MM cells, “young” cellular and molecular factors are now emerging, taking the scene of this complex neoplastic setting. In this review we aimed at giving insights on the latest evidence of potentially-targetable modes that MM cells exploit to increase fitness and gain a survival advantage. The benefits coming from the derangements of stress-managing pathways, autophagy, transcriptional rewiring, and non-coding RNAs are examples of such methods that MM cells utilize to escape cell death, but that hopefully will offer novel targets for the ever-increasing anti-MM therapeutic armamentarium.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine, Hematology Section, University of Padova, Via N.Giustiniani 2, 35128 Padova, Italy.
- Venetian Institute of Molecular Medicine, Via G.Orus 2, 35129 Padova, Italy.
| | - Marilena Carrino
- Department of Medicine, Hematology Section, University of Padova, Via N.Giustiniani 2, 35128 Padova, Italy.
- Venetian Institute of Molecular Medicine, Via G.Orus 2, 35129 Padova, Italy.
| | - Gianpietro Semenzato
- Department of Medicine, Hematology Section, University of Padova, Via N.Giustiniani 2, 35128 Padova, Italy.
- Venetian Institute of Molecular Medicine, Via G.Orus 2, 35129 Padova, Italy.
| | - Francesco Piazza
- Department of Medicine, Hematology Section, University of Padova, Via N.Giustiniani 2, 35128 Padova, Italy.
- Venetian Institute of Molecular Medicine, Via G.Orus 2, 35129 Padova, Italy.
| |
Collapse
|
29
|
Takai T, Matsuda T, Matsuura Y, Inoue K, Suzuki E, Kanno A, Kimura-Koyanagi M, Asahara SI, Hatano N, Ogawa W, Kido Y. Casein kinase 2 phosphorylates and stabilizes C/EBPβ in pancreatic β cells. Biochem Biophys Res Commun 2018; 497:451-456. [PMID: 29448105 DOI: 10.1016/j.bbrc.2018.02.108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 02/11/2018] [Indexed: 12/27/2022]
Abstract
During the development of type 2 diabetes, endoplasmic reticulum (ER) stress leads to pancreatic β cell failure. CCAAT/enhancer-binding protein (C/EBP) β is highly induced by ER stress and AMP-activated protein kinase (AMPK) suppression in pancreatic β cells, and its accumulation reduces pancreatic β cell mass. We investigated the phosphorylation state of C/EBPβ under these conditions. Casein kinase 2 (CK2) was found to co-localize with C/EBPβ in MIN6 cells. It phosphorylated S222 of C/EBPβ, a previously unidentified phosphorylation site. We found that C/EBPβ is phosphorylated by CK2 under AMPK suppression and ER stress, which are important from the viewpoint of the worsening pathological condition of type 2 diabetes, such as decreased insulin secretion and apoptosis of pancreatic β cells.
Collapse
Affiliation(s)
- Tomoko Takai
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomokazu Matsuda
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuki Matsuura
- Division of Metabolism and Disease, Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Kaho Inoue
- Division of Metabolism and Disease, Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Emi Suzuki
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ayumi Kanno
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Maki Kimura-Koyanagi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shun-Ichiro Asahara
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoya Hatano
- The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshiaki Kido
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Metabolism and Disease, Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan.
| |
Collapse
|
30
|
CK2 blockade causes MPNST cell apoptosis and promotes degradation of β-catenin. Oncotarget 2018; 7:53191-53203. [PMID: 27448963 PMCID: PMC5288178 DOI: 10.18632/oncotarget.10668] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/07/2016] [Indexed: 12/24/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are soft tissue sarcomas that are a major cause of mortality of Neurofibromatosis type 1 (NF1) patients. MPNST patients have few therapeutic options available and only complete surgical resection can be curative. MPNST formation and survival are dependent on activated β-catenin signaling. The goal of this study was to determine if inhibition of the CK2 enzyme can be therapeutically exploited in MPNSTs, given CK2's role in mainta ining oncogenic phenotypes including stabilization of β-catenin. We found that CK2α is over-expressed in MPNSTs and is critical for maintaining cell survival, as the CK2 inhibitor, CX-4945 (Silmitasertib), and shRNA targeting CK2α each significantly reduce MPNST cell viability. These effects were preceded by loss of critical signaling pathways in MPNSTs, including destabilization of β-catenin and TCF8. CX-4945 administration in vivo slowed tumor growth and extends survival time. We conclude that CK2 inhibition is a promising approach to blocking β-catenin in MPNST cells, although combinatorial therapies may be required for maximal efficacy.
Collapse
|
31
|
Wu F, Qiu J, Fan Y, Zhang Q, Cheng B, Wu Y, Bai B. Apelin-13 attenuates ER stress-mediated neuronal apoptosis by activating Gα i/Gα q-CK2 signaling in ischemic stroke. Exp Neurol 2018; 302:136-144. [PMID: 29337146 DOI: 10.1016/j.expneurol.2018.01.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/28/2017] [Accepted: 01/08/2018] [Indexed: 11/26/2022]
Abstract
Cerebral ischemia/reperfusion (I/R) injury-induced neuronal apoptosis contributes to the death and disability in patients with ischemic stroke. However, underlying mechanisms remain elusive and it lacks effective treatment. Here we reported that the expression of casein kinase 2 (CK2) was significantly reduced in brains of middle cerebral artery occlusion/reperfusion (MACO/R) model rats and oxygen-glucose deprivation/reperfusion (OGD/R) model neurons, which was associated with the activation of eIF2-ATF4-CHOP signaling pathway, leading to neuronal apoptosis. Moreover, we found that apelin-13 significantly upregulated CK2 expression and inhibited eIF2-ATF4-CHOP activation, attenuating cerebral I/R injury-induced infarct and neuronal apoptosis in MACO/R model rats and OGD/R model neurons. Furthermore, we demonstrated that the rescue effect of apelin-13 on I/R injury-induced neuronal apoptosis was mediated by Gαi/Gαq-CK2-dependent inhibition of eIF2-ATF4-CHOP activation. These data indicated cerebral I/R injury reduced CK2 expression and activated eIF2-ATF4-CHOP signaling contributing to neuronal apoptosis, and apelin-13 can activate Gαi/Gαq-CK2 signaling attenuating eIF2-ATF4-CHOP-mediated neuronal apoptosis. It provides a novel insight that not only apelin-13 but also CK2 agonists may have therapeutic potential for protecting neurons from I/R injury-induced apoptosis, facilitating post-stroke recovery.
Collapse
Affiliation(s)
- Fei Wu
- College of Life Science, Shandong Agricultural University, Taian, Shandong, China; Institute of Neurobiology, Jining Medical University, Jining, Shandong, China
| | - Jian Qiu
- Institute of Neurobiology, Jining Medical University, Jining, Shandong, China; Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yang Fan
- Institute of Neurobiology, Jining Medical University, Jining, Shandong, China
| | - Qiuling Zhang
- Life Science Research Center, Taishan Medical College, Taian, Shandong, China
| | - Baohua Cheng
- Institute of Neurobiology, Jining Medical University, Jining, Shandong, China
| | - Yili Wu
- Department of Psychiatry, Jining Medical University, Jining, Shandong, China; Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, Shandong, China; Shandong Key Laboratory of Behavioral Medicine, Jining Medical University, Jining, Shandong, China.
| | - Bo Bai
- Institute of Neurobiology, Jining Medical University, Jining, Shandong, China; Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, Shandong, China; Shandong Key Laboratory of Behavioral Medicine, Jining Medical University, Jining, Shandong, China.
| |
Collapse
|
32
|
Pitale PM, Gorbatyuk O, Gorbatyuk M. Neurodegeneration: Keeping ATF4 on a Tight Leash. Front Cell Neurosci 2017; 11:410. [PMID: 29326555 PMCID: PMC5736573 DOI: 10.3389/fncel.2017.00410] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/05/2017] [Indexed: 12/15/2022] Open
Abstract
Activation of the endoplasmic reticulum (ER) stress and ER stress response, also known as the unfolded protein response (UPR), is common to various degenerative disorders. Therefore, signaling components of the UPR are currently emerging as potential targets for intervention and treatment of human diseases. One UPR signaling member, activating transcription factor 4 (ATF4), has been found up-regulated in many pathological conditions, pointing to therapeutic potential in targeting its expression. In cells, ATF4 governs multiple signaling pathways, including autophagy, oxidative stress, inflammation, and translation, suggesting a multifaceted role of ATF4 in the progression of various pathologies. However, ATF4 has been shown to trigger both pro-survival and pro-death pathways, and this, perhaps, can explain the contradictory opinions in current literature regarding targeting ATF4 for clinical application. In this review, we summarized recent published studies from our labs and others that focus on the therapeutic potential of the strategy controlling ATF4 expression in different retinal and neurodegenerative disorders.
Collapse
Affiliation(s)
- Priyamvada M Pitale
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Oleg Gorbatyuk
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Marina Gorbatyuk
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
33
|
Manni S, Carrino M, Piazza F. Role of protein kinases CK1α and CK2 in multiple myeloma: regulation of pivotal survival and stress-managing pathways. J Hematol Oncol 2017; 10:157. [PMID: 28969692 PMCID: PMC5625791 DOI: 10.1186/s13045-017-0529-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/22/2017] [Indexed: 01/07/2023] Open
Abstract
Multiple myeloma (MM) is a malignant tumor of transformed plasma cells. MM pathogenesis is a multistep process. This cancer can occur de novo (rarely) or it can develop from monoclonal gammopathy of undetermined significance (most of the cases). MM can be asymptomatic (smoldering myeloma) or clinically active. Malignant plasma cells exploit intrinsic and extrinsic bone marrow microenvironment-derived growth signals. Upregulation of stress-coping pathways is also instrumental to maintain MM cell growth. The phylogenetically related Ser/Thr kinases CSNK1A1 (CK1α) and CSNK2 (CK2) have recently gained a growing importance in hematologic malignancies arising both from precursors and from mature blood cells. In multiple myeloma, CK1α or CK2 sustain oncogenic cascades, such as the PI3K/AKT, JAK/STAT, and NF-κB, as well as propel stress-related signaling that help in coping with different noxae. Data also suggest that these kinases modulate the delivery of growth factors and cytokines from the bone marrow stroma. The “non-oncogene addiction” phenotype generated by the increased activity of CK1α and CK2 in multiple myeloma contributes to malignant plasma cell proliferation and survival and represents an Achilles’ heel for the activity of small ATP competitive CK1α or CK2 inhibitors.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine, Hematology Section, University of Padova, Via Giustiniani 2, 35128, Padova, Italy. .,Venetian Institute of Molecular Medicine, Padova, Italy.
| | - Marilena Carrino
- Department of Medicine, Hematology Section, University of Padova, Via Giustiniani 2, 35128, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Francesco Piazza
- Department of Medicine, Hematology Section, University of Padova, Via Giustiniani 2, 35128, Padova, Italy. .,Venetian Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
34
|
Yang J, Wu W, Wen J, Ye H, Luo H, Bai P, Tang M, Wang F, Zheng L, Yang S, Li W, Peng A, Yang L, Wan L, Chen L. Liposomal honokiol induced lysosomal degradation of Hsp90 client proteins and protective autophagy in both gefitinib-sensitive and gefitinib-resistant NSCLC cells. Biomaterials 2017; 141:188-198. [DOI: 10.1016/j.biomaterials.2017.07.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 06/30/2017] [Accepted: 07/02/2017] [Indexed: 12/19/2022]
|
35
|
Manni S, Carrino M, Manzoni M, Gianesin K, Nunes SC, Costacurta M, Tubi LQ, Macaccaro P, Taiana E, Cabrelle A, Barilà G, Martines A, Zambello R, Bonaldi L, Trentin L, Neri A, Semenzato G, Piazza F. Inactivation of CK1α in multiple myeloma empowers drug cytotoxicity by affecting AKT and β-catenin survival signaling pathways. Oncotarget 2017; 8:14604-14619. [PMID: 28099937 PMCID: PMC5362429 DOI: 10.18632/oncotarget.14654] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/07/2017] [Indexed: 11/25/2022] Open
Abstract
Recent evidence indicates that protein kinase CK1α may support the growth of multiple myeloma (MM) plasma cells. Here, by analyzing a large cohort of MM cases, we found that high CK1α mRNA levels are virtually associated with all MM patients. Moreover, we provided functional evidence that CK1α activity is essential for malignant plasma cell survival even in the protective niche generated by co-cultures with bone marrow stromal cells. We demonstrated that CK1α inactivation, while toxic for myeloma cells, is dispensable for the survival of healthy B lymphocytes and stromal cells. Disruption of CK1α function in myeloma cells resulted in decreased Mdm2, increased p53 and p21 and reduced expression of β-catenin and AKT. These effects were mediated partially by p53 and caspase activity. Finally, we discovered that CK1α inactivation enhanced the cytotoxic effect of both bortezomib and lenalidomide. Overall, our study supports a role for CK1α as a potential therapeutic target in MM in combination with proteasome inhibitors and/or immunomodulatory drugs.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Marilena Carrino
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Martina Manzoni
- Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy.,Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy
| | - Ketty Gianesin
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Sara Canovas Nunes
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Matteo Costacurta
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Laura Quotti Tubi
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Paolo Macaccaro
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Elisa Taiana
- Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy.,Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy
| | - Anna Cabrelle
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Gregorio Barilà
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Annalisa Martines
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS- Padova, Italy
| | - Renato Zambello
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Laura Bonaldi
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS- Padova, Italy
| | - Livio Trentin
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Antonino Neri
- Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy.,Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milano, Italy
| | - Gianpietro Semenzato
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Francesco Piazza
- Department of Medicine, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
36
|
Inhibition of protein kinase CK2 sensitizes non-small cell lung cancer cells to cisplatin via upregulation of PML. Mol Cell Biochem 2017; 436:87-97. [PMID: 28744813 DOI: 10.1007/s11010-017-3081-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/30/2017] [Indexed: 12/24/2022]
Abstract
Non-small cell lung carcinoma (NSCLC), a malignancy of lungs, is very aggressive and usually ends up with a dismal prognosis. Cisplatin (CDDP)-based systemic chemotherapy is the main pharmaceutical approach for treating NSCLC, but its effect is discounted by some hitherto unknown reasons. Thus, this study is dedicated to improving the efficacy of CDDP. Our results show that combining use of CDDP with CK2 siRNA or inhibitor is more efficient in suppressing cancer cell growth and promoting apoptosis than use of CDDP alone. The underlying mechanism is that CDDP has two pathways to go: one is that it directly induces apoptosis and the other is that it activates CK2, which suppresses proapoptosis gene promyelocytic leukemia (PML). In conclusion, inhibiting CK2 can enhance sensitivity of CDDP to NSCLC cancer cells through PML.
Collapse
|
37
|
The mammalian STE20-like kinase 1 (MST1) is a substrate for the apoptosis inhibiting protein kinase CK2. Cell Signal 2017; 36:163-175. [PMID: 28487119 DOI: 10.1016/j.cellsig.2017.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 04/28/2017] [Accepted: 05/05/2017] [Indexed: 10/19/2022]
Abstract
Apoptosis and the response to cell stress are evolutionary highly conserved mechanisms. Both processes require strict regulation, which is often performed by protein kinases. The mammalian Sterile 20-like kinase 1 (MST1) is a pro-apoptotic protein kinase, which is activated and cleaved by caspases upon the induction of cell stress. Being a phosphoprotein itself, the activity of MST1 is regulated by phosphorylation. Protein kinase CK2 is an anti-apoptotic protein kinase which seems to be involved in the regulation of many different cellular processes including apoptosis. There is increasing evidence that the cleavage of many substrates by caspases is regulated by phosphorylation in the close vicinity of the caspase cleavage sites. One of these kinases, implicated in the phosphorylation of caspase substrates, is protein kinase CK2. Here, we report that serine 320 of the MST1 protein is a novel phosphorylation site for the anti-apoptotic protein kinase CK2. Although serine 320 is in close vicinity to the caspase 3 cleavage site, caspase 3 cleavage of MST1 is not affected by CK2 phosphorylation. Using biochemical approaches, we were able to show that MST1 co-localizes with the CK2 subunits in the pancreatic β-cell line INS-1 and that full-length MST1 and the activated N-terminal fragment of MST1 both interacted with the CK2 subunits in vitro and in vivo. MST1 is a basophilic kinase whereas CK2 is an acidophilic kinase. Thus, binding of these two kinases in the cytosol and in the nucleus opens the door to the phosphorylation of a variety of new substrates.
Collapse
|
38
|
Chua MMJ, Ortega CE, Sheikh A, Lee M, Abdul-Rassoul H, Hartshorn KL, Dominguez I. CK2 in Cancer: Cellular and Biochemical Mechanisms and Potential Therapeutic Target. Pharmaceuticals (Basel) 2017; 10:E18. [PMID: 28134850 PMCID: PMC5374422 DOI: 10.3390/ph10010018] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/23/2017] [Accepted: 01/23/2017] [Indexed: 01/09/2023] Open
Abstract
CK2 genes are overexpressed in many human cancers, and most often overexpression is associated with worse prognosis. Site-specific expression in mice leads to cancer development (e.g., breast, lymphoma) indicating the oncogenic nature of CK2. CK2 is involved in many key aspects of cancer including inhibition of apoptosis, modulation of signaling pathways, DNA damage response, and cell cycle regulation. A number of CK2 inhibitors are now available and have been shown to have activity against various cancers in vitro and in pre-clinical models. Some of these inhibitors are now undergoing exploration in clinical trials as well. In this review, we will examine some of the major cancers in which CK2 inhibition has promise based on in vitro and pre-clinical studies, the proposed cellular and signaling mechanisms of anti-cancer activity by CK2 inhibitors, and the current or recent clinical trials using CK2 inhibitors.
Collapse
Affiliation(s)
- Melissa M J Chua
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| | - Charina E Ortega
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| | - Ayesha Sheikh
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| | - Migi Lee
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| | - Hussein Abdul-Rassoul
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| | - Kevan L Hartshorn
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| | - Isabel Dominguez
- Department of Medicine, School of Medicine, Boston University, Boston, MA 02118, USA.
| |
Collapse
|
39
|
Lumley EC, Osborn AR, Scott JE, Scholl AG, Mercado V, McMahan YT, Coffman ZG, Brewster JL. Moderate endoplasmic reticulum stress activates a PERK and p38-dependent apoptosis. Cell Stress Chaperones 2017; 22:43-54. [PMID: 27761878 PMCID: PMC5225058 DOI: 10.1007/s12192-016-0740-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 10/05/2016] [Accepted: 10/07/2016] [Indexed: 02/07/2023] Open
Abstract
The endoplasmic reticulum (ER) has the ability to signal organelle dysfunction via a complex signaling network known as the unfolded protein response (UPR). In this work, hamster fibroblast cells exhibiting moderate levels of ER stress were compared to those exhibiting severe ER stress. Inhibition of N-linked glycosylation was accomplished via a temperature-sensitive mutation in the Dad1 subunit of the oligosaccharyltransferase (OST) complex or by direct inhibition with tunicamycin (Tm). Temperature shift (TS) treatment generated weak activation of ER stress signaling when compared to doses of Tm that are typically used in ER stress studies (500-1000 nM). A dose-response analysis of key ER stress signaling mediators, inositol-requiring enzyme 1 (IRE1) and protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), revealed 20-40 nM of Tm to generate activation intensity similar to TS treatment. In parental BHK21 cells, moderate (20-40 nM) and high doses (200-1000 nM) of Tm were compared to identify physiological and signaling-based differences in stress response. Inhibition of ER Ca2+ release via ITPR activity with 2-aminoethoxydiphenyl borate (2-APB) or Xestospongin C (XeC) was sufficient to protect against apoptosis induced by moderate but not higher doses of Tm. Analysis of kinase activation over a range of Tm exposures revealed the p38 stress-activated protein kinase (SAPK) to display increasing activation with Tm dosage. Interestingly, Tm induced the extracellular regulated kinases (Erk1/2) only at moderate doses of Tm. Inhibition of ER transmembrane stress sensors (IRE1, PERK) or cytosolic signaling mediators (p38, Jnk1, Erk1/2) was used to evaluate pathways involved in apoptosis activation during ER stress. Inhibition of either PERK or p38 was sufficient to reduce cell death and apoptosis induced by moderate, but not high, doses of Tm. During ER stress, cells exhibited a rapid decline in anti-apoptotic Mcl-1 and survivin proteins. Inhibition of PERK was sufficient to block this affect. This work reveals moderate doses of ER stress to generate patterns of stress signaling that are distinct from higher doses and that apoptosis activation at moderate levels of stress are dependent upon PERK and p38 signaling. Studies exploring ER stress signaling should recognize that this signaling acts as a rheostat rather than a simple switch, behaving distinctively in a dose-dependent manner.
Collapse
Affiliation(s)
- Emily C Lumley
- Natural Science Division, Pepperdine University, 24255 Pacific Coast Highway, Malibu,, CA, 90263, USA
| | - Acadia R Osborn
- Natural Science Division, Pepperdine University, 24255 Pacific Coast Highway, Malibu,, CA, 90263, USA
| | - Jessica E Scott
- Natural Science Division, Pepperdine University, 24255 Pacific Coast Highway, Malibu,, CA, 90263, USA
| | - Amanda G Scholl
- Natural Science Division, Pepperdine University, 24255 Pacific Coast Highway, Malibu,, CA, 90263, USA
| | - Vicki Mercado
- Natural Science Division, Pepperdine University, 24255 Pacific Coast Highway, Malibu,, CA, 90263, USA
| | - Young T McMahan
- Natural Science Division, Pepperdine University, 24255 Pacific Coast Highway, Malibu,, CA, 90263, USA
| | - Zachary G Coffman
- Natural Science Division, Pepperdine University, 24255 Pacific Coast Highway, Malibu,, CA, 90263, USA
| | - Jay L Brewster
- Natural Science Division, Pepperdine University, 24255 Pacific Coast Highway, Malibu,, CA, 90263, USA.
| |
Collapse
|
40
|
Synergistic cytotoxic effects of bortezomib and CK2 inhibitor CX-4945 in acute lymphoblastic leukemia: turning off the prosurvival ER chaperone BIP/Grp78 and turning on the pro-apoptotic NF-κB. Oncotarget 2016; 7:1323-40. [PMID: 26593250 PMCID: PMC4811463 DOI: 10.18632/oncotarget.6361] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 11/15/2015] [Indexed: 01/22/2023] Open
Abstract
The proteasome inhibitor bortezomib is a new targeted treatment option for refractory or relapsed acute lymphoblastic leukemia (ALL) patients. However, a limited efficacy of bortezomib alone has been reported. A terminal pro-apoptotic endoplasmic reticulum (ER) stress/unfolded protein response (UPR) is one of the several mechanisms of bortezomib-induced apoptosis. Recently, it has been documented that UPR disruption could be considered a selective anti-leukemia therapy. CX-4945, a potent casein kinase (CK) 2 inhibitor, has been found to induce apoptotic cell death in T-ALL preclinical models, via perturbation of ER/UPR pathway. In this study, we analyzed in T- and B-ALL preclinical settings, the molecular mechanisms of synergistic apoptotic effects observed after bortezomib/CX-4945 combined treatment. We demonstrated that, adding CX-4945 after bortezomib treatment, prevented leukemic cells from engaging a functional UPR in order to buffer the bortezomib-mediated proteotoxic stress in ER lumen. We documented that the combined treatment decreased pro-survival ER chaperon BIP/Grp78 expression, via reduction of chaperoning activity of Hsp90. Bortezomib/CX-4945 treatment inhibited NF-κB signaling in T-ALL cell lines and primary cells from T-ALL patients, but, intriguingly, in B-ALL cells the drug combination activated NF-κB p65 pro-apoptotic functions. In fact in B-cells, the combined treatment induced p65-HDAC1 association with consequent repression of the anti-apoptotic target genes, Bcl-xL and XIAP. Exposure to NEMO (IKKγ)-binding domain inhibitor peptide reduced the cytotoxic effects of bortezomib/CX-4945 treatment. Overall, our findings demonstrated that CK2 inhibition could be useful in combination with bortezomib as a novel therapeutic strategy in both T- and B-ALL.
Collapse
|
41
|
Mandato E, Manni S, Zaffino F, Semenzato G, Piazza F. Targeting CK2-driven non-oncogene addiction in B-cell tumors. Oncogene 2016; 35:6045-6052. [PMID: 27041560 DOI: 10.1038/onc.2016.86] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 02/15/2016] [Accepted: 02/15/2016] [Indexed: 12/14/2022]
Abstract
Genetic mutations of oncogenes often underlie deranged cell growth and altered differentiation pathways leading to malignant transformation of B-lymphocytes. However, addiction to oncogenes is not the only drive to lymphoid tumor pathogenesis. Dependence on non-oncogenes, which act by propelling basic mechanisms of cell proliferation and survival, has also been recognized in the pathobiology of lymphoid leukemias, lymphomas and multiple myeloma. Among the growing number of molecules that may uphold non-oncogene addiction, a key place is increasingly being recognized to the serine-threonine kinase CK2. This enzyme is overexpressed and overactive in B-acute lymphoblastic leukemia, multiple myeloma, chronic lymphocytic leukemia and non-Hodgkin lymphomas, such as mantle cell, follicular, Burkitt's and diffuse large B-cell lymphomas. In these tumors, CK2 may serve the activity of oncogenes, similar to BCR-ABL and c-MYC, control the activation of critical signaling cascades, such as NF-κB (nuclear factor-κB), STAT3 (signal transducer and activator of transcription 3) and PTEN/PI3K/AKT (phosphatase and tensin homolog protein/phosphoinositide 3-kinase/AKR thymoma), and sustain multiple cellular stress-elicited pathways, such as the proteotoxic stress, unfolded protein and DNA-damage responses. CK2 has also been shown to have an essential role in tuning signals derived from the stromal tumor microenvironment. Not surprisingly, targeting CK2 in lymphoid tumor cell lines or mouse xenograft models can boost the cytotoxic effects of both conventional chemotherapeutics and novel agents, similar to heat-shock protein 90, proteasome and tyrosine kinases inhibitors. In this review, we summarize the evidence indicating how CK2 embodies most of the features of a cancer growth-promoting non-oncogene, focusing on lymphoid tumors. We further discuss the preclinical data of the use of small ATP-competitive CK2 inhibitors, which hold the promise to be additional options in novel drug combinations for the therapy of lymphoid and plasmacellular malignancies.
Collapse
Affiliation(s)
- E Mandato
- Department of Medicine, Hematology Branch, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - S Manni
- Department of Medicine, Hematology Branch, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - F Zaffino
- Department of Medicine, Hematology Branch, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - G Semenzato
- Department of Medicine, Hematology Branch, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - F Piazza
- Department of Medicine, Hematology Branch, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
42
|
Pizzi M, Piazza F, Agostinelli C, Fuligni F, Benvenuti P, Mandato E, Casellato A, Rugge M, Semenzato G, Pileri SA. Protein kinase CK2 is widely expressed in follicular, Burkitt and diffuse large B-cell lymphomas and propels malignant B-cell growth. Oncotarget 2016; 6:6544-52. [PMID: 25788269 PMCID: PMC4466633 DOI: 10.18632/oncotarget.3446] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 01/28/2015] [Indexed: 12/02/2022] Open
Abstract
Serine-threonine kinase CK2 is highly expressed and pivotal for survival and proliferation in multiple myeloma, chronic lymphocytic leukemia and mantle cell lymphoma. Here, we investigated the expression of α catalytic and β regulatory CK2 subunits by immunohistochemistry in 57 follicular (FL), 18 Burkitt (BL), 52 diffuse large B-cell (DLBCL) non-Hodgkin lymphomas (NHL) and in normal reactive follicles. In silico evaluation of available Gene Expression Profile (GEP) data sets from patients and Western blot (WB) analysis in NHL cell-lines were also performed. Moreover, the novel, clinical-grade, ATP-competitive CK2-inhibitor CX-4945 (Silmitasertib) was assayed on lymphoma cells. CK2 was detected in 98.4% of cases with a trend towards a stronger CK2α immunostain in BL compared to FL and DLBCL. No significant differences were observed between Germinal Center B (GCB) and non-GCB DLBCL types. GEP data and WB confirmed elevated CK2 mRNA and protein levels as well as active phosphorylation of specific targets in NHL cells. CX-4945 caused a dose-dependent growth-arresting effect on GCB, non-GCB DLBCL and BL cell-lines and it efficiently shut off phosphorylation of NF-κB RelA and CDC37 on CK2 target sites. Thus, CK2 is highly expressed and could represent a suitable therapeutic target in BL, FL and DLBCL NHL.
Collapse
Affiliation(s)
- Marco Pizzi
- Department of Medicine, Surgical Pathology and Cytopathology Unit, DIMED University of Padua, Padua, Italy
| | - Francesco Piazza
- Department of Medicine, Hematology and Clinical Immunology Branch, DIMED University of Padua, Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Claudio Agostinelli
- Department of Experimental, Hematopathology and Hematology Sections, Diagnostic and Specialty Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Fabio Fuligni
- Department of Experimental, Hematopathology and Hematology Sections, Diagnostic and Specialty Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Pietro Benvenuti
- Department of Medicine, Surgical Pathology and Cytopathology Unit, DIMED University of Padua, Padua, Italy
| | - Elisa Mandato
- Department of Medicine, Hematology and Clinical Immunology Branch, DIMED University of Padua, Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Alessandro Casellato
- Department of Medicine, Hematology and Clinical Immunology Branch, DIMED University of Padua, Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Massimo Rugge
- Department of Medicine, Surgical Pathology and Cytopathology Unit, DIMED University of Padua, Padua, Italy
| | - Gianpietro Semenzato
- Department of Medicine, Hematology and Clinical Immunology Branch, DIMED University of Padua, Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Stefano A Pileri
- Department of Experimental, Hematopathology and Hematology Sections, Diagnostic and Specialty Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| |
Collapse
|
43
|
Rossi M, Tagliaferri P, Tassone P. MicroRNAs in multiple myeloma and related bone disease. ANNALS OF TRANSLATIONAL MEDICINE 2016; 3:334. [PMID: 26734644 DOI: 10.3978/j.issn.2305-5839.2015.12.13] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
MicroRNAs (miRNAs) are short non coding RNAs aberrantly expressed in solid and hematopoietic malignancies where they play a pivotal function as post-transcriptional regulators of gene expression. Recent reports have unveiled a central role of miRNAs in multiple myeloma onset and progression and preclinical findings are progressively disclosing their potential therapeutic value as drugs or targets. In this review, we provide the basic insights of miRNA biology and function, showing how these molecules are extensively dysregulated in malignant plasma cells (PC) and related microenvironment, thus favoring clone survival and proliferation. We here describe how these critical activities have recently been evaluated to design miRNA-based therapies against multiple myeloma cells and its surrounding microenvironment.
Collapse
Affiliation(s)
- Marco Rossi
- 1 Department of Experimental and Clinical Medicine, Magna Graecia University, Campus Salvatore Venuta, Catanzaro, Italy ; 2 Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Pierosandro Tagliaferri
- 1 Department of Experimental and Clinical Medicine, Magna Graecia University, Campus Salvatore Venuta, Catanzaro, Italy ; 2 Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Pierfrancesco Tassone
- 1 Department of Experimental and Clinical Medicine, Magna Graecia University, Campus Salvatore Venuta, Catanzaro, Italy ; 2 Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| |
Collapse
|
44
|
Bhardwaj M, Paul S, Jakhar R, Kang SC. Potential role of vitexin in alleviating heat stress-induced cytotoxicity: Regulatory effect of Hsp90 on ER stress-mediated autophagy. Life Sci 2015; 142:36-48. [DOI: 10.1016/j.lfs.2015.10.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/25/2015] [Accepted: 10/10/2015] [Indexed: 12/19/2022]
|
45
|
Combined inhibition of Hsp90 and the proteasome affects NSCLC proteostasis and attenuates cell migration. Anticancer Drugs 2015; 25:998-1006. [PMID: 25153785 DOI: 10.1097/cad.0000000000000140] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Lung cancer remains the most common cause of cancer-related death worldwide. This malignancy is a complex disease, and it is important to identify potential biological targets, the blockade of which would affect multiple downstream signaling cascades. A growing number of reports recognize novel therapeutic targets in the protein homeostasis network responsible for generating and protecting the protein fold. The heat shock protein 90 (Hsp90) is an essential molecular chaperon involved in the posttranslational folding and stability of proteins. It is required for conformational maturation of multiple oncogenic kinases that drive signal transduction and proliferation of cancer cells. However, in the case of unfolded protein accumulation endoplasmic reticulum (ER) stress is induced and several response pathways such as proteasome functions are activated. The ubiquitin-proteasome system orchestrates the turnover of innumerable cellular proteins. Here, we suggest that the therapeutic efficacy of Hsp90 inhibition may be augmented by coadministering proteasome inhibitor on human non-small-cell lung cancer (NSCLC) cell lines. Indeed, we showed that coadministration of the Hsp90 inhibitor 17-demethoxygeldanamycin (17-DMAG) and proteasome inhibitor (velcade) induced ER stress evidenced by increased unfolded protein response markers. The consequences were evident in multiple aspects of the NSCLC phenotype: reduced viability and cell count, increased apoptotic cell death, and most profoundly, synergistically decreased cell motility. Our findings provide proof-of-concept that targeting ER homeostasis is therapeutically beneficial in NSCLC cell lines.
Collapse
|
46
|
Wang Y, McAlpine SR. N-terminal and C-terminal modulation of Hsp90 produce dissimilar phenotypes. Chem Commun (Camb) 2015; 51:1410-3. [DOI: 10.1039/c4cc07284g] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Classic oncogenic heat shock protein 90 (Hsp90) inhibitors target the N-terminus of the protein, triggering a survival mechanism in cancer cells referred to as the heat shock response (HSR).
Collapse
Affiliation(s)
- Y. Wang
- Department of chemistry
- Sydney
- Australia
| | | |
Collapse
|
47
|
Abstract
The term 'casein kinase' has been widely used for decades to denote protein kinases sharing the ability to readily phosphorylate casein in vitro. These fall into three main classes: two of them, later renamed as protein kinases CK1 (casein kinase 1, also known as CKI) and CK2 (also known as CKII), are pleiotropic members of the kinome functionally unrelated to casein, whereas G-CK, or genuine casein kinase, responsible for the phosphorylation of casein in the Golgi apparatus of the lactating mammary gland, has only been identified recently with Fam20C [family with sequence similarity 20C; also known as DMP-4 (dentin matrix protein-4)], a member of the four-jointed family of atypical protein kinases, being responsible for the phosphorylation of many secreted proteins. In hindsight, therefore, the term 'casein kinase' is misleading in every instance; in the case of CK1 and CK2, it is because casein is not a physiological substrate, and in the case of G-CK/Fam20C/DMP-4, it is because casein is just one out of a plethora of its targets, and a rather marginal one at that. Strikingly, casein kinases altogether, albeit representing a minimal proportion of the whole kinome, appear to be responsible for the generation of up to 40-50% of non-redundant phosphosites currently retrieved in human phosphopeptides database. In the present review, a short historical explanation will be provided accounting for the usage of the same misnomer to denote three unrelated classes of protein kinases, together with an update of our current knowledge of these pleiotropic enzymes, sharing the same misnomer while playing very distinct biological roles.
Collapse
|
48
|
CSNK1α1 mediates malignant plasma cell survival. Leukemia 2014; 29:474-82. [PMID: 24962017 DOI: 10.1038/leu.2014.202] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 05/12/2014] [Accepted: 05/19/2014] [Indexed: 12/18/2022]
Abstract
Here we report that targeting casein kinase 1-α1 (CSNK1α1) is a potential novel treatment strategy in multiple myeloma (MM) therapy distinct from proteasome inhibition. CSNK1α1 is expressed in all the tested MM cell lines and patient MM cells, and is not altered during bortezomib-triggered cytotoxicity. Inhibition of CSNK1α1 kinase activity in MM cells with targeted therapy D4476 or small hairpin RNAs triggers cell G0/G1-phase arrest, prolonged G2/M phase and apoptosis. D4476 also induced cytotoxicity in bortezomib-resistant MM cells and enhanced bortezomib-triggered cytotoxicity. CSNK1α1 signaling pathways include CDKN1B, P53 and FADD; gene signatures involved included interferon-α, tumor necrosis factor-α and LIN9. In addition, reduction of Csnk1α1 prevents cMYC/KRAS12V transformation of BaF3 cells independent of interleukin-3. Impartially, reducing Csnk1α1 prevented development of cMYC/KRAS12V-induced plasmacytomas in mice, suggesting that CSNK1α1 may be involved in MM initiation and progression. Our data suggest that targeting CSNK1α1, alone or combined with bortezomib, is a potential novel therapeutic strategy in MM. Moreover, inhibition of CSNK1α1 may prevent the progression of monoclonal gammopathy of undetermined significance to MM.
Collapse
|
49
|
Bone marrow stromal cell-fueled multiple myeloma growth and osteoclastogenesis are sustained by protein kinase CK2. Leukemia 2014; 28:2094-7. [PMID: 24897506 DOI: 10.1038/leu.2014.178] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
50
|
Jiang Q, Li F, Shi K, Wu P, An J, Yang Y, Xu C. Involvement of p38 in signal switching from autophagy to apoptosis via the PERK/eIF2α/ATF4 axis in selenite-treated NB4 cells. Cell Death Dis 2014; 5:e1270. [PMID: 24874742 PMCID: PMC4047911 DOI: 10.1038/cddis.2014.200] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 04/04/2014] [Accepted: 04/08/2014] [Indexed: 02/07/2023]
Abstract
Selenite has emerged as an optional chemotherapeutic agent for hematological malignancies. Autophagy and apoptosis are both engaged in selenite-induced cell death. In a previous report, we have identified heat shock protein 90 (Hsp90) as a critical modulator of the balance between autophagy and apoptosis in selenite-treated leukemia cells. However, the mechanisms by which selenite mediates the crosstalk between autophagy and apoptosis remain largely unknown. Herein, we demonstrate that the endoplasmic reticulum (ER) stress-related PERK/eIF2α/ATF4 pathway and p38 are core modules for the selenite-induced switch to apoptosis from autophagy. We found that selenite activated PERK and eIF2α/ATF4 downstream to promote apoptosis. During this progression, p38 was dissociated from PERK-inhibiting Hsp90 and became autophosphorylated. Then, activated p38 further enhanced the docking of activating transcription factor 4 (ATF4) onto the CHOP (CCAAT/enhancer-binding protein homologous protein) promoter via eIF2α to enhance apoptosis. We also found that activated p38 suppressed the phosphorylation of eIF4E that directed ATF4 to bind to the MAP1LC3B (microtubule-associated protein 1 light chain 3B) promoter. Because of the deactivation of eIF4E, the association of ATF4 with the MAP1LC3B promoter was inhibited, and autophagy was compromised. Intriguingly, p53 played important roles in mediating the p38-mediated regulation of eIF2α and eIF4E. When activated by p38, p53 induced the phosphorylation of eIF2α and the dephosphorylation of eIF4E, particularly in the nucleus where the ATF4 transcription factor was modulated, ultimately resulting in differential expression of CHOP and LC3. Moreover, selenite exhibited potent antitumor effects in vivo. In an NB4 cell xenograft model, selenite induced apoptosis and hampered autophagy. In addition, related signaling proteins demonstrated similar changes to those observed in vitro. These data suggest that selenite may be a candidate drug for leukemia therapy.
Collapse
Affiliation(s)
- Q Jiang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - F Li
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - K Shi
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - P Wu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - J An
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Y Yang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - C Xu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|