1
|
Wang C, Du M, Jiang Z, Cong R, Wang W, Zhang T, Chen J, Zhang G, Li L. PI3K-AKT-mediated phosphorylation of Thr260 in CgCaspase-3/6/7 regulates heat-induced activation in oysters. Commun Biol 2024; 7:1459. [PMID: 39511363 PMCID: PMC11543851 DOI: 10.1038/s42003-024-07184-4] [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: 03/11/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024] Open
Abstract
Cysteine-aspartic proteases (caspases) are critical drivers of apoptosis, exhibiting expansion and domain shuffling in mollusks. However, the functions and regulatory mechanisms of these caspases remain unclear. In this study, we identified a group of Caspase-3/6/7 in Bivalvia and Gastropoda with a long inter-subunit linker (IL) that inhibits cleavage activation. Within this region, we found that conserved phosphorylation at Thr260 in oysters, mediated by the PI3K-AKT pathway, suppresses heat-induced activation. This mechanism is involved in divergent temperature adaptation between two allopatric congeneric oyster species, the relatively cold-adapted Crassostrea gigas and warm-adapted Crassostrea angulata. Our study elucidates the role of these effector caspase members and their long IL in bivalves, revealing that the PI3K-AKT pathway phosphorylates Thr260 on CgCASP3/6/7's linker to inhibit heat-induced activation. These findings provide insights into the evolution and function of apoptotic regulatory mechanisms in bivalves.
Collapse
Affiliation(s)
- Chaogang Wang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture(CAS), Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
| | - Mingyang Du
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhuxiang Jiang
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Rihao Cong
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Wei Wang
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Taiping Zhang
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jincheng Chen
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Guofan Zhang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture(CAS), Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
- Shandong Technology Innovation Center of Oyster Seed Industry, Qingdao, China
| | - Li Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture(CAS), Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, China.
- Shandong Technology Innovation Center of Oyster Seed Industry, Qingdao, China.
| |
Collapse
|
2
|
Ghorbani N, Yaghubi R, Davoodi J, Pahlavan S. How does caspases regulation play role in cell decisions? apoptosis and beyond. Mol Cell Biochem 2024; 479:1599-1613. [PMID: 37976000 DOI: 10.1007/s11010-023-04870-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/05/2023] [Indexed: 11/19/2023]
Abstract
Caspases are a family of cysteine proteases, and the key factors behind the cellular events which occur during apoptosis and inflammation. However, increasing evidence shows the non-conventional pro-survival action of apoptotic caspases in crucial processes. These cellular events include cell proliferation, differentiation, and migration, which may appear in the form of metastasis, and chemotherapy resistance in cancerous situations. Therefore, there should be a precise and strict control of caspases activity, perhaps through maintaining the threshold below the required levels for apoptosis. Thus, understanding the regulators of caspase activities that render apoptotic caspases as non-apoptotic is of paramount importance both mechanistically and clinically. Furthermore, the functions of apoptotic caspases are affected by numerous post-translational modifications. In the present mini-review, we highlight the various mechanisms that directly impact caspases with respect to their anti- or non-apoptotic functions. In this regard, post-translational modifications (PTMs), isoforms, subcellular localization, transient activity, substrate availability, substrate selection, and interaction-mediated regulations are discussed.
Collapse
Affiliation(s)
- Negar Ghorbani
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Roham Yaghubi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Jamshid Davoodi
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
3
|
Ghorbani N, Shiri M, Alian M, Yaghubi R, Shafaghi M, Hojjat H, Pahlavan S, Davoodi J. A Non-Apoptotic Pattern of Caspase-9/Caspase-3 Activation During Differentiation of Human Embryonic Stem Cells into Cardiomyocytes. Adv Biol (Weinh) 2024; 8:e2400026. [PMID: 38640919 DOI: 10.1002/adbi.202400026] [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: 01/15/2024] [Revised: 03/22/2024] [Indexed: 04/21/2024]
Abstract
In vitro studies have demonstrated that the differentiation of embryonic stem cells (ESCs) into cardiomyocytes requires activation of caspases through the mitochondrial pathway. These studies have relied on synthetic substrates for activity measurements, which can be misleading due to potential none-specific hydrolysis of these substrates by proteases other than caspases. Hence, caspase-9 and caspase-3 activation are investigated during the differentiation of human ESCs (hESCs) by directly assessing caspase-9 and -3 cleavage. Western blot reveals the presence of the cleaved caspase-9 prior to and during the differentiation of human ESCs (hESCs) into cardiomyocytes at early stages, which diminishes as the differentiation progresses, without cleavage and activation of endogenous procaspase-3. Activation of exogenous procaspase-3 by endogenous caspase-9 and subsequent cleavage of chromogenic caspase-3 substrate i.e. DEVD-pNA during the course of differentiation confirmes that endogenous caspase-9 has the potency to recognize and activate procaspase-3, but for reasons that are unknown to us fails to do so. These observations suggest the existence of distinct mechanisms of caspase regulation in differentiation as compared to apoptosis. Bioinformatics analysis suggests the presence of caspase-9 regulators, which may influence proteolytic function under specific conditions.
Collapse
Affiliation(s)
- Negar Ghorbani
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, 1417614335, Iran
| | - Mahshad Shiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Maedeh Alian
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, 1417614335, Iran
| | - Roham Yaghubi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, 1417614411, Iran
| | - Mojtaba Shafaghi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Hamidreza Hojjat
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, 1417614335, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Jamshid Davoodi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, 1417614335, Iran
| |
Collapse
|
4
|
Bancet A, Frem R, Jeanneret F, Mularoni A, Bazelle P, Roelants C, Delcros JG, Guichou JF, Pillet C, Coste I, Renno T, Battail C, Cochet C, Lomberget T, Filhol O, Krimm I. Cancer selective cell death induction by a bivalent CK2 inhibitor targeting the ATP site and the allosteric αD pocket. iScience 2024; 27:108903. [PMID: 38318383 PMCID: PMC10838953 DOI: 10.1016/j.isci.2024.108903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/25/2023] [Accepted: 01/09/2024] [Indexed: 02/07/2024] Open
Abstract
Although the involvement of protein kinase CK2 in cancer is well-documented, there is a need for selective CK2 inhibitors suitable for investigating CK2 specific roles in cancer-related biological pathways and further exploring its therapeutic potential. Here, we report the discovery of AB668, an outstanding selective inhibitor that binds CK2 through a bivalent mode, interacting both at the ATP site and an allosteric αD pocket unique to CK2. Using caspase activation assay, live-cell imaging, and transcriptomic analysis, we have compared the effects of this bivalent inhibitor to representative ATP-competitive inhibitors, CX-4945, and SGC-CK2-1. Our results show that in contrast to CX-4945 or SGC-CK2-1, AB668, by targeting the CK2 αD pocket, has a distinct mechanism of action regarding its anti-cancer activity, inducing apoptotic cell death in several cancer cell lines and stimulating distinct biological pathways in renal cell carcinoma.
Collapse
Affiliation(s)
- Alexandre Bancet
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Small Molecules for Biological Targets », 69373 Lyon, France
- Kairos Discovery SAS, 36 Rue Jeanne d’Arc, 69003 Lyon, France
| | - Rita Frem
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Targeting Non-canonical Protein Functions in Cancer », 69373 Lyon, France
| | - Florian Jeanneret
- Université Grenoble Alpes, IRIG, Laboratoire Biosciences et Bioingénierie pour la Santé, UA 13 INSERM-CEA-UGA, 38000 Grenoble, France
| | - Angélique Mularoni
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Small Molecules for Biological Targets », 69373 Lyon, France
| | - Pauline Bazelle
- Université Grenoble Alpes, IRIG, Laboratoire Biosciences et Bioingénierie pour la Santé, UA 13 INSERM-CEA-UGA, 38000 Grenoble, France
| | - Caroline Roelants
- University Grenoble Alpes, INSERM 1292, CEA, UMR Biosanté, 38000 Grenoble, France
| | - Jean-Guy Delcros
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Small Molecules for Biological Targets », 69373 Lyon, France
| | - Jean-François Guichou
- Centre de Biologie Structurale, CNRS, INSERM, University Montpellier, 34090 Montpellier, France
| | - Catherine Pillet
- University Grenoble Alpes, INSERM 1292, CEA, UMR Biosanté, 38000 Grenoble, France
| | - Isabelle Coste
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Targeting Non-canonical Protein Functions in Cancer », 69373 Lyon, France
| | - Toufic Renno
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Targeting Non-canonical Protein Functions in Cancer », 69373 Lyon, France
| | - Christophe Battail
- Université Grenoble Alpes, IRIG, Laboratoire Biosciences et Bioingénierie pour la Santé, UA 13 INSERM-CEA-UGA, 38000 Grenoble, France
| | - Claude Cochet
- University Grenoble Alpes, INSERM 1292, CEA, UMR Biosanté, 38000 Grenoble, France
| | - Thierry Lomberget
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), COSSBA Team, Faculté de Pharmacie-ISPB, 8 Avenue Rockefeller, 69373 Lyon Cedex 08, France
| | - Odile Filhol
- University Grenoble Alpes, INSERM 1292, CEA, UMR Biosanté, 38000 Grenoble, France
| | - Isabelle Krimm
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Small Molecules for Biological Targets », 69373 Lyon, France
| |
Collapse
|
5
|
Montenarh M, Götz C. Protein Kinase CK2α', More than a Backup of CK2α. Cells 2023; 12:2834. [PMID: 38132153 PMCID: PMC10741536 DOI: 10.3390/cells12242834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
The serine/threonine protein kinase CK2 is implicated in the regulation of fundamental processes in eukaryotic cells. CK2 consists of two catalytic α or α' isoforms and two regulatory CK2β subunits. These three proteins exist in a free form, bound to other cellular proteins, as tetrameric holoenzymes composed of CK2α2/β2, CK2αα'/β2, or CK2α'2/β2 as well as in higher molecular forms of the tetramers. The catalytic domains of CK2α and CK2α' share a 90% identity. As CK2α contains a unique C-terminal sequence. Both proteins function as protein kinases. These properties raised the question of whether both isoforms are just backups of each other or whether they are regulated differently and may then function in an isoform-specific manner. The present review provides observations that the regulation of both CK2α isoforms is partly different concerning the subcellular localization, post-translational modifications, and aggregation. Up to now, there are only a few isoform-specific cellular binding partners. The expression of both CK2α isoforms seems to vary in different cell lines, in tissues, in the cell cycle, and with differentiation. There are different reports about the expression and the functions of the CK2α isoforms in tumor cells and tissues. In many cases, a cell-type-specific expression and function is known, which raises the question about cell-specific regulators of both isoforms. Another future challenge is the identification or design of CK2α'-specific inhibitors.
Collapse
Affiliation(s)
- Mathias Montenarh
- Medical Biochemistry and Molecular Biology, Saarland University, Building 44, 66421 Homburg, Germany;
| | | |
Collapse
|
6
|
Firnau MB, Brieger A. CK2 and the Hallmarks of Cancer. Biomedicines 2022; 10:1987. [PMID: 36009534 PMCID: PMC9405757 DOI: 10.3390/biomedicines10081987] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer is a leading cause of death worldwide. Casein kinase 2 (CK2) is commonly dysregulated in cancer, impacting diverse molecular pathways. CK2 is a highly conserved serine/threonine kinase, constitutively active and ubiquitously expressed in eukaryotes. With over 500 known substrates and being estimated to be responsible for up to 10% of the human phosphoproteome, it is of significant importance. A broad spectrum of diverse types of cancer cells has been already shown to rely on disturbed CK2 levels for their survival. The hallmarks of cancer provide a rationale for understanding cancer's common traits. They constitute the maintenance of proliferative signaling, evasion of growth suppressors, resisting cell death, enabling of replicative immortality, induction of angiogenesis, the activation of invasion and metastasis, as well as avoidance of immune destruction and dysregulation of cellular energetics. In this work, we have compiled evidence from the literature suggesting that CK2 modulates all hallmarks of cancer, thereby promoting oncogenesis and operating as a cancer driver by creating a cellular environment favorable to neoplasia.
Collapse
Affiliation(s)
| | - Angela Brieger
- Department of Internal Medicine I, Biomedical Research Laboratory, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
7
|
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
|
8
|
Gyenis L, Menyhart D, Cruise ES, Jurcic K, Roffey SE, Chai DB, Trifoi F, Fess SR, Desormeaux PJ, Núñez de Villavicencio Díaz T, Rabalski AJ, Zukowski SA, Turowec JP, Pittock P, Lajoie G, Litchfield DW. Chemical Genetic Validation of CSNK2 Substrates Using an Inhibitor-Resistant Mutant in Combination with Triple SILAC Quantitative Phosphoproteomics. Front Mol Biosci 2022; 9:909711. [PMID: 35755813 PMCID: PMC9225150 DOI: 10.3389/fmolb.2022.909711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022] Open
Abstract
Casein Kinase 2 (CSNK2) is an extremely pleiotropic, ubiquitously expressed protein kinase involved in the regulation of numerous key biological processes. Mapping the CSNK2-dependent phosphoproteome is necessary for better characterization of its fundamental role in cellular signalling. While ATP-competitive inhibitors have enabled the identification of many putative kinase substrates, compounds targeting the highly conserved ATP-binding pocket often exhibit off-target effects limiting their utility for definitive kinase-substrate assignment. To overcome this limitation, we devised a strategy combining chemical genetics and quantitative phosphoproteomics to identify and validate CSNK2 substrates. We engineered U2OS cells expressing exogenous wild type CSNK2A1 (WT) or a triple mutant (TM, V66A/H160D/I174A) with substitutions at residues important for inhibitor binding. These cells were treated with CX-4945, a clinical-stage inhibitor of CSNK2, and analyzed using large-scale triple SILAC (Stable Isotope Labelling of Amino Acids in Cell Culture) quantitative phosphoproteomics. In contrast to wild-type CSNK2A1, CSNK2A1-TM retained activity in the presence of CX-4945 enabling identification and validation of several CSNK2 substrates on the basis of their increased phosphorylation in cells expressing CSNK2A1-TM. Based on high conservation within the kinase family, we expect that this strategy can be broadly adapted for identification of other kinase-substrate relationships.
Collapse
Affiliation(s)
- Laszlo Gyenis
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Daniel Menyhart
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Edward S Cruise
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Kristina Jurcic
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Scott E Roffey
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Darren B Chai
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Flaviu Trifoi
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Sam R Fess
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Paul J Desormeaux
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | | | - Adam J Rabalski
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Stephanie A Zukowski
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Jacob P Turowec
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Paula Pittock
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Gilles Lajoie
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - David W Litchfield
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada.,Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| |
Collapse
|
9
|
Wińska P, Widło Ł, Senkara E, Koronkiewicz M, Cieśla JM, Krzyśko A, Skierka K, Cieśla J. Inhibition of Protein Kinase CK2 Affects Thymidylate Synthesis Cycle Enzyme Level and Distribution in Human Cancer Cells. Front Mol Biosci 2022; 9:847829. [PMID: 35281258 PMCID: PMC8914513 DOI: 10.3389/fmolb.2022.847829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Thymidylate synthase (TS), dihydrofolate reductase (DHFR), and serine hydroxymethyltransferase (SHMT) constitute the thymidylate synthesis cycle providing thymidylate for DNA synthesis and repair. Our previous studies indicated that TS and DHFR are the substrates of protein kinase CK2. This work has been aimed at the elucidation of the effect of CK2 activity on cell cycle progression, thymidylate synthesis enzyme expression and localization, and the role of CK2-mediated TS phosphorylation in in vitro di- and trimolecular complex formation. The results were obtained by means of western blot, confocal microscopy, flow cytometry, quantitative polymerase chain reaction (QPCR), quartz crystal microbalance with dissipation monitoring (QCM-D), and microthermophoresis (MST). Our research indicates that CK2 inhibition does not change the levels of the transcripts; however, it affects the protein levels of DHFR and TS in both tested cell lines, i.e., A549 and CCRF-CEM, and the level of SHMT1 in CCRF-CEM cells. Moreover, we show that CK2-mediated phosphorylation of TS enables the protein (pTS) interaction with SHMT1 and leads to the stability of the tri-complex containing SHMT1, DHFR, and pTS. Our results suggest an important regulatory role of CK2-mediated phosphorylation for inter- and intracellular protein level of enzymes involved in the thymidylate biosynthesis cycle.
Collapse
Affiliation(s)
- Patrycja Wińska
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
- *Correspondence: Patrycja Wińska, ; Joanna Cieśla,
| | - Łukasz Widło
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Elżbieta Senkara
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | | | - Jarosław M. Cieśla
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Alicja Krzyśko
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Katarzyna Skierka
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Joanna Cieśla
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
- *Correspondence: Patrycja Wińska, ; Joanna Cieśla,
| |
Collapse
|
10
|
Olguín HC. The Gentle Side of the UPS: Ubiquitin-Proteasome System and the Regulation of the Myogenic Program. Front Cell Dev Biol 2022; 9:821839. [PMID: 35127730 PMCID: PMC8811165 DOI: 10.3389/fcell.2021.821839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, the ubiquitin-proteasome system (UPS) has emerged as an important regulator of stem cell function. Here we review recent findings indicating that UPS also plays critical roles in the biology of satellite cells, the muscle stem cell responsible for its maintenance and regeneration. While we focus our attention on the control of key transcriptional regulators of satellite cell function, we briefly discuss early studies suggesting the UPS participates more broadly in the regulation of satellite cell stemness and regenerative capacity.
Collapse
|
11
|
Lindenblatt D, Applegate V, Nickelsen A, Klußmann M, Neundorf I, Götz C, Jose J, Niefind K. Molecular Plasticity of Crystalline CK2α' Leads to KN2, a Bivalent Inhibitor of Protein Kinase CK2 with Extraordinary Selectivity. J Med Chem 2021; 65:1302-1312. [PMID: 34323071 DOI: 10.1021/acs.jmedchem.1c00063] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
CK2α and CK2α' are paralogous catalytic subunits of CK2, which belongs to the eukaryotic protein kinases. CK2 promotes tumorigenesis and the spread of pathogenic viruses like SARS-CoV-2 and is thus an attractive drug target. Efforts to develop selective CK2 inhibitors binding offside the ATP site had disclosed the αD pocket in CK2α; its occupation requires large conformational adaptations of the helix αD. As shown here, the αD pocket is accessible also in CK2α', where the necessary structural plasticity can be triggered with suitable ligands even in the crystalline state. A CK2α' structure with an ATP site and an αD pocket ligand guided the design of the bivalent CK2 inhibitor KN2. It binds to CK2 with low nanomolar affinity, is cell-permeable, and suppresses the intracellular phosphorylation of typical CK2 substrates. Kinase profiling revealed a high selectivity of KN2 for CK2 and emphasizes the selectivity-promoting potential of the αD pocket.
Collapse
Affiliation(s)
- Dirk Lindenblatt
- Department für Chemie, Institut für Biochemie, Universität zu Köln, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Violetta Applegate
- Department für Chemie, Institut für Biochemie, Universität zu Köln, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Anna Nickelsen
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, PharmaCampus, Corrensstr. 48, D-48149 Münster, Germany
| | - Merlin Klußmann
- Department für Chemie, Institut für Biochemie, Universität zu Köln, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Ines Neundorf
- Department für Chemie, Institut für Biochemie, Universität zu Köln, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Claudia Götz
- Medizinische Biochemie und Molekularbiologie, Universität des Saarlandes, Kirrberger Str., Geb. 44, D-66421 Homburg/Saar, Germany
| | - Joachim Jose
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, PharmaCampus, Corrensstr. 48, D-48149 Münster, Germany
| | - Karsten Niefind
- Department für Chemie, Institut für Biochemie, Universität zu Köln, Zülpicher Str. 47, D-50674 Köln, Germany
| |
Collapse
|
12
|
Chojnacki K, Wińska P, Karatsai O, Koronkiewicz M, Milner-Krawczyk M, Wielechowska M, Rędowicz MJ, Bretner M, Borowiecki P. Synthesis of Novel Acyl Derivatives of 3-(4,5,6,7-Tetrabromo-1 H-benzimidazol-1-yl)propan-1-ols-Intracellular TBBi-Based CK2 Inhibitors with Proapoptotic Properties. Int J Mol Sci 2021; 22:6261. [PMID: 34200807 PMCID: PMC8230474 DOI: 10.3390/ijms22126261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 12/09/2022] Open
Abstract
Protein kinase CK2 has been considered as an attractive drug target for anti-cancer therapy. The synthesis of N-hydroxypropyl TBBi and 2MeTBBi derivatives as well as their respective esters was carried out by using chemoenzymatic methods. Concomitantly with kinetic studies toward recombinant CK2, the influence of the obtained compounds on the viability of two human breast carcinoma cell lines (MCF-7 and MDA-MB-231) was evaluated using MTT assay. Additionally, an intracellular inhibition of CK2 as well as an induction of apoptosis in the examined cells after the treatment with the most active compounds were studied by Western blot analysis, phase-contrast microscopy and flow cytometry method. The results of the MTT test revealed potent cytotoxic activities for most of the newly synthesized compounds (EC50 4.90 to 32.77 µM), corresponding to their solubility in biological media. We concluded that derivatives with the methyl group decrease the viability of both cell lines more efficiently than their non-methylated analogs. Furthermore, inhibition of CK2 in breast cancer cells treated with the tested compounds at the concentrations equal to their EC50 values correlates well with their lipophilicity since derivatives with higher values of logP are more potent intracellular inhibitors of CK2 with better proapoptotic properties than their parental hydroxyl compounds.
Collapse
Affiliation(s)
- Konrad Chojnacki
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Patrycja Wińska
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Olena Karatsai
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (O.K.); (M.J.R.)
| | - Mirosława Koronkiewicz
- Department of Drug Biotechnology and Bioinformatics, National Medicines Institute, 00-725 Warsaw, Poland;
| | - Małgorzata Milner-Krawczyk
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Monika Wielechowska
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (O.K.); (M.J.R.)
| | - Maria Bretner
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| | - Paweł Borowiecki
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.C.); (M.M.-K.); (M.W.); (M.B.); (P.B.)
| |
Collapse
|
13
|
Szumilak M, Wiktorowska-Owczarek A, Stanczak A. Hybrid Drugs-A Strategy for Overcoming Anticancer Drug Resistance? Molecules 2021; 26:2601. [PMID: 33946916 PMCID: PMC8124695 DOI: 10.3390/molecules26092601] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
Despite enormous progress in the treatment of many malignancies, the development of cancer resistance is still an important reason for cancer chemotherapy failure. Increasing knowledge of cancers' molecular complexity and mechanisms of their resistance to anticancer drugs, as well as extensive clinical experience, indicate that an effective fight against cancer requires a multidimensional approach. Multi-target chemotherapy may be achieved using drugs combination, co-delivery of medicines, or designing hybrid drugs. Hybrid drugs simultaneously targeting many points of signaling networks and various structures within a cancer cell have been extensively explored in recent years. The single hybrid agent can modulate multiple targets involved in cancer cell proliferation, possesses a simpler pharmacokinetic profile to reduce the possibility of drug interactions occurrence, and facilitates the process of drug development. Moreover, a single medication is expected to enhance patient compliance due to a less complicated treatment regimen, as well as a diminished number of adverse reactions and toxicity in comparison to a combination of drugs. As a consequence, many efforts have been made to design hybrid molecules of different chemical structures and functions as a means to circumvent drug resistance. The enormous number of studies in this field encouraged us to review the available literature and present selected research results highlighting the possible role of hybrid drugs in overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Marta Szumilak
- Department of Hospital Pharmacy, Faculty of Pharmacy, Medical University of Lodz, 1 Muszynskiego Street, 90-151 Lodz, Poland
| | - Anna Wiktorowska-Owczarek
- Department of Pharmacology and Toxicology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland;
| | - Andrzej Stanczak
- Department of Community Pharmacy, Faculty of Pharmacy, Medical University of Lodz, 1 Muszynskiego Street, 90-151 Lodz, Poland;
| |
Collapse
|
14
|
Yang F, Liu Q, Chen Y, Ye H, Wang H, Zeng S. Integrative Proteomic and Phosphoproteomic Analyses of Granulosa Cells During Follicular Atresia in Porcine. Front Cell Dev Biol 2021; 8:624985. [PMID: 33520998 PMCID: PMC7843964 DOI: 10.3389/fcell.2020.624985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/09/2020] [Indexed: 12/16/2022] Open
Abstract
Ovarian follicular atresia is a natural physiological process; however, the mechanism is not fully understood. In this study, quantitative proteomic and phosphoproteomic analyses of granulosa cells (GCs) in healthy (H), slightly atretic (SA), and atretic follicles (A) of porcine were performed by TMT labeling, enrichment of phosphopeptides, and liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis. In total, 6,201 proteins were quantified, and 4,723 phosphorylation sites of 1,760 proteins were quantified. In total, 24 (11 up, 13 down) and 50 (29 up, 21 down) proteins with a fold change (FC) > 5 were identified in H/SA and H/A, respectively. In addition, there were 20 (H/SA, up) and 39 (H/A, up) phosphosites with an FC > 7 that could serve as potential biomarkers for distinguishing different quality categories of follicles. Western blotting and immunofluorescence confirmed the reliability of the proteomic analysis. Some key proteins (e.g., MIF, beta catenin, integrin β2), phosphosites (e.g., S76 of caspase6, S22 and S636 of lamin A/C), pathways (e.g., apoptosis, regulation of actin cytoskeleton pathway), transcription factors (e.g., STAT5A, FOXO1, and BCLAF1), and kinases (e.g., PBK, CDK5, CDK12, and AKT3) involved in the atresia process were revealed via further analysis of the differentially expressed proteins (DEPs) and phosphorylated proteins (DEPPs). Further study showed that mutant caspase6 Ser76 to Ala increased the ratios of cleaved caspase6/caspase6 and cleaved caspase3/caspase3 and dephosphorylation of caspase6 at Ser76 increased cell apoptotic rate, a new potential pathway of follicular atresia. Collectively, the proteomic and phosphoproteomic profiling and functional research in the current study comprehensively analyzed the dynamic changes in protein expression and phosphorylation during follicular atresia and provided some new explanations regarding the regulation of this process.
Collapse
Affiliation(s)
- Feng Yang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qiang Liu
- Beijing Advanced Innovation Center for Genomics, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yanhong Chen
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Huizhen Ye
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Han Wang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shenming Zeng
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
15
|
Hashemolhosseini S. The role of protein kinase CK2 in skeletal muscle: Myogenesis, neuromuscular junctions, and rhabdomyosarcoma. Neurosci Lett 2020; 729:135001. [DOI: 10.1016/j.neulet.2020.135001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 01/08/2023]
|
16
|
Connolly P, Garcia-Carpio I, Villunger A. Cell-Cycle Cross Talk with Caspases and Their Substrates. Cold Spring Harb Perspect Biol 2020; 12:a036475. [PMID: 31727679 PMCID: PMC7263087 DOI: 10.1101/cshperspect.a036475] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Caspases play central roles in mediating both cell death and inflammation. It has more recently become evident that caspases also drive other biological processes. Most prominently, caspases have been shown to be involved in differentiation. Several stem and progenitor cell types rely on caspases to initiate and execute their differentiation processes. These range from neural and glial cells, to skeletal myoblasts and osteoblasts, and several cell types of the hematopoietic system. Beyond differentiation, caspases have also been shown to play roles in other "noncanonical" processes, including cell proliferation, arrest, and senescence, thereby contributing to the mechanisms that regulate tissue homeostasis at multiple levels. Remarkably, caspases directly influence the course of the cell cycle in both a positive and negative manner. Caspases both cleave elements of the cell-cycle machinery and are themselves substrates of cell-cycle kinases. Here we aim to summarize the breadth of interactions between caspases and cell-cycle regulators. We also highlight recent developments in this area.
Collapse
Affiliation(s)
- Patrick Connolly
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Irmina Garcia-Carpio
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Andreas Villunger
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna 1090, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| |
Collapse
|
17
|
Tan Y, Wu Q, Zhou F. Targeting acute myeloid leukemia stem cells: Current therapies in development and potential strategies with new dimensions. Crit Rev Oncol Hematol 2020; 152:102993. [PMID: 32502928 DOI: 10.1016/j.critrevonc.2020.102993] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/15/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
High relapse rate of acute myeloid leukemia (AML) is still a crucial problem despite considerable advances in anti-cancer therapies. One crucial cause of relapse is the existence of leukemia stem cells (LSCs) with self-renewal ability, which contribute to repeated treatment resistance and recurrence. Treatments targeting LSCs, especially in combination with existing chemotherapy regimens or hematopoietic stem cell transplantation might help achieve a higher complete remission rate and improve overall survival. Many novel agents of different therapeutic strategies that aim to modulate LSCs self-renewal, proliferation, apoptosis, and differentiation are under investigation. In this review, we summarize the latest advances of different therapies in development based on the biological characteristics of LSCs, with particular attention on natural products, synthetic compounds, antibody therapies, and adoptive cell therapies that promote the LSC eradication. We also explore the causes of AML recurrence and proposed potential strategies with new dimensions for targeting LSCs in the future.
Collapse
Affiliation(s)
- Yuxin Tan
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, People's Republic of China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, People's Republic of China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, People's Republic of China.
| |
Collapse
|
18
|
Richter A, Sender S, Lenz A, Schwarz R, Hinz B, Knuebel G, Sekora A, Murua Escobar H, Junghanss C, Roolf C. Influence of Casein kinase II inhibitor CX-4945 on BCL6-mediated apoptotic signaling in B-ALL in vitro and in vivo. BMC Cancer 2020; 20:184. [PMID: 32131762 PMCID: PMC7057698 DOI: 10.1186/s12885-020-6650-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/17/2020] [Indexed: 12/18/2022] Open
Abstract
Background Casein kinase II (CK2) is involved in multiple tumor-relevant signaling pathways affecting proliferation and apoptosis. CK2 is frequently upregulated in acute B-lymphoblastic leukemia (B-ALL) and can be targeted by the ATP-competitive CK2 inhibitor CX-4945. While reduced proliferation of tumor entities including B-ALL after CX-4945 incubation has been shown in vitro and in vivo, the detailed way of action is unknown. Here, we investigated the influence on the PI3K/AKT and apoptosis cascades in vivo and in vitro for further clarification. Methods A B-ALL xenograft model in NSG mice was used to perform in vivo longitudinal bioluminescence imaging during six day CX-4945 treatment. CX-4945 serum levels were determined at various time points. Flow cytometry of bone marrow and spleen cells was performed to analyze CX-4945-induced effects on tumor cell proliferation and distribution in B-ALL engrafted mice. ALL cells were enriched and characterized by targeted RNA sequencing. In vitro, B-ALL cell lines SEM, RS4;11 and NALM-6 were incubated with CX-4945 and gene expression of apoptosis regulators BCL6 and BACH2 was determined. Results In B-ALL-engrafted mice, overall tumor cell proliferation and distribution was not significantly influenced by CK2 inhibition. CX-4945 was detectable in serum during therapy and serum levels declined rapidly after cessation of CX-4945. While overall proliferation was not affected, early bone marrow and spleen blast frequencies seemed reduced after CK2 inhibition. Gene expression analyses revealed reduced expression of anti-apoptotic oncogene BCL6 in bone marrow blasts of CX-4945-treated animals. Further, BCL6 protein expression decreased in B-ALL cell lines exposed to CX-4945 in vitro. Surprisingly, levels of BCL6 opponent and tumor suppressor BACH2 also declined after prolonged incubation. Simultaneously, increased phosphorylation of direct CK2 target and tumor initiator AKT was detected at respective time points, even in initially pAKT-negative cell line NALM-6. Conclusions The CK2 inhibitor CX-4945 has limited clinical effects in an in vivo B-ALL xenograft model when applied as a single drug over a six day period. However, gene expression in B-ALL cells was altered and suggested effects on apoptosis via downregulation of BCL6. Unexpectedly, the BCL6 opponent BACH2 was also reduced. Interactions and regulation loops have to be further evaluated.
Collapse
Affiliation(s)
- Anna Richter
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Strasse 6, 18057, Rostock, Germany
| | - Sina Sender
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Strasse 6, 18057, Rostock, Germany
| | - Annemarie Lenz
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Strasse 6, 18057, Rostock, Germany
| | - Rico Schwarz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057, Rostock, Germany
| | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057, Rostock, Germany
| | - Gudrun Knuebel
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Strasse 6, 18057, Rostock, Germany
| | - Anett Sekora
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Strasse 6, 18057, Rostock, Germany
| | - Hugo Murua Escobar
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Strasse 6, 18057, Rostock, Germany
| | - Christian Junghanss
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Strasse 6, 18057, Rostock, Germany.
| | - Catrin Roolf
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Strasse 6, 18057, Rostock, Germany
| |
Collapse
|
19
|
Signaling Pathways, Chemical and Biological Modulators of Nucleotide Excision Repair: The Faithful Shield against UV Genotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4654206. [PMID: 31485292 PMCID: PMC6702832 DOI: 10.1155/2019/4654206] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/10/2019] [Indexed: 12/28/2022]
Abstract
The continuous exposure of the human body's cells to radiation and genotoxic stresses leads to the accumulation of DNA lesions. Fortunately, our body has several effective repair mechanisms, among which is nucleotide excision repair (NER), to counteract these lesions. NER includes both global genome repair (GG-NER) and transcription-coupled repair (TC-NER). Deficiencies in the NER pathway underlie the development of several DNA repair diseases, such as xeroderma pigmentosum (XP), Cockayne syndrome (CS), and trichothiodystrophy (TTD). Deficiencies in GG-NER and TC-NER render individuals to become prone to cancer and neurological disorders, respectively. Therefore, NER regulation is of interest in fine-tuning these risks. Distinct signaling cascades including the NFE2L2 (NRF2), AHR, PI3K/AKT1, MAPK, and CSNK2A1 pathways can modulate NER function. In addition, several chemical and biological compounds have proven success in regulating NER's activity. These modulators, particularly the positive ones, could therefore provide potential treatments for genetic DNA repair-based diseases. Negative modulators, nonetheless, can help sensitize cells to killing by genotoxic chemicals. In this review, we will summarize and discuss the major upstream signaling pathways and molecules that could modulate the NER's activity.
Collapse
|
20
|
Hoque A, Williamson NA, Ameen SS, Ciccotosto GD, Hossain MI, Oakhill JS, Ng DCH, Ang CS, Cheng HC. Quantitative proteomic analyses of dynamic signalling events in cortical neurons undergoing excitotoxic cell death. Cell Death Dis 2019; 10:213. [PMID: 30824683 PMCID: PMC6397184 DOI: 10.1038/s41419-019-1445-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/20/2019] [Accepted: 02/01/2019] [Indexed: 12/23/2022]
Abstract
Excitotoxicity, caused by overstimulation or dysregulation of ionotropic glutamate receptors (iGluRs), is a pathological process directing neuronal death in many neurological disorders. The aberrantly stimulated iGluRs direct massive influx of calcium ions into the affected neurons, leading to changes in expression and phosphorylation of specific proteins to modulate their functions and direct their participation in the signalling pathways that induce excitotoxic neuronal death. To define these pathways, we used quantitative proteomic approaches to identify these neuronal proteins (referred to as the changed proteins) and determine how their expression and/or phosphorylation dynamically changed in association with excitotoxic cell death. Our data, available in ProteomeXchange with identifier PXD008353, identified over 100 changed proteins exhibiting significant alterations in abundance and/or phosphorylation levels at different time points (5–240 min) in neurons after glutamate overstimulation. Bioinformatic analyses predicted that many of them are components of signalling networks directing defective neuronal morphology and functions. Among them, the well-known neuronal survival regulators including mitogen-activated protein kinases Erk1/2, glycogen synthase kinase 3 (GSK3) and microtubule-associated protein (Tau), were selected for validation by biochemical approaches, which confirmed the findings of the proteomic analysis. Bioinformatic analysis predicted Protein Kinase B (Akt), c-Jun kinase (JNK), cyclin-dependent protein kinase 5 (Cdk5), MAP kinase kinase (MEK), Casein kinase 2 (CK2), Rho-activated protein kinase (Rock) and Serum/glucocorticoid-regulated kinase 1 (SGK1) as the potential upstream kinases phosphorylating some of the changed proteins. Further biochemical investigation confirmed the predictions of sustained changes of the activation states of neuronal Akt and CK2 in excitotoxicity. Thus, future investigation to define the signalling pathways directing the dynamic alterations in abundance and phosphorylation of the identified changed neuronal proteins will help elucidate the molecular mechanism of neuronal death in excitotoxicity.
Collapse
Affiliation(s)
- Ashfaqul Hoque
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia.,Cell Signalling Research Laboratories, University of Melbourne, Parkville, VIC, 3010, Australia.,Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.,Metabolic Signalling Laboratory, St. Vincent's Institute for Medical Research, University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Nicholas A Williamson
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - S Sadia Ameen
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia.,Cell Signalling Research Laboratories, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Giuseppe D Ciccotosto
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC, 3010, Australia
| | - M Iqbal Hossain
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia.,Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St. Vincent's Institute for Medical Research, University of Melbourne, Fitzroy, VIC, 3065, Australia.,Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, 3000, Australia
| | - Dominic C H Ng
- School of Biomedical Sciences, University of Queensland, St. Lucia, QLD, Australia
| | - Ching-Seng Ang
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Heung-Chin Cheng
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia. .,Cell Signalling Research Laboratories, University of Melbourne, Parkville, VIC, 3010, Australia. .,Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
21
|
Putinski C, Abdul-Ghani M, Brunette S, Burgon PG, Megeney LA. Caspase Cleavage of Gelsolin Is an Inductive Cue for Pathologic Cardiac Hypertrophy. J Am Heart Assoc 2018; 7:e010404. [PMID: 30486716 PMCID: PMC6405540 DOI: 10.1161/jaha.118.010404] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Cardiac hypertrophy is an adaptive remodeling event that may improve or diminish contractile performance of the heart. Physiologic and pathologic hypertrophy yield distinct outcomes, yet both are dependent on caspase‐directed proteolysis. This suggests that each form of myocardial growth may derive from a specific caspase cleavage event(s). We examined whether caspase 3 cleavage of the actin capping/severing protein gelsolin is essential for the development of pathologic hypertrophy. Methods and Results Caspase targeting of gelsolin was established through protein analysis of hypertrophic cardiomyocytes and mass spectrometry mapping of cleavage sites. Pathologic agonists induced late‐stage caspase‐mediated cleavage of gelsolin. The requirement of caspase‐mediated gelsolin cleavage for hypertrophy induction was evaluated in primary cardiomyocytes by cell size analysis, monitoring of prohypertrophy markers, and measurement of hypertrophy‐related transcription activity. The in vivo impact of caspase‐mediated cleavage was investigated by echo‐guided intramyocardial injection of adenoviral‐expressed gelsolin. Expression of the N‐terminal gelsolin caspase cleavage fragment was necessary and sufficient to cause pathologic remodeling in isolated cardiomyocytes and the intact heart, whereas expression of a noncleavable form prevents cardiac remodeling. Alterations in myocardium structure and function were determined by echocardiography and end‐stage cardiomyocyte cell size analysis. Gelsolin secretion was also monitored for its impact on naïve cells using competitive antibody trapping, demonstrating that hypertrophic agonist stimulation of cardiomyocytes leads to gelsolin secretion, which induces hypertrophy in naïve cells. Conclusions These results suggest that cell autonomous caspase cleavage of gelsolin is essential for pathologic hypertrophy and that cardiomyocyte secretion of gelsolin may accelerate this negative remodeling response.
Collapse
Affiliation(s)
- Charis Putinski
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada.,2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada
| | - Mohammad Abdul-Ghani
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada.,2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada
| | - Steve Brunette
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada
| | - Patrick G Burgon
- 2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada.,3 Department of Medicine University of Ottawa Ontario Canada.,4 University of Ottawa Heart Institute Ottawa Ontario Canada
| | - Lynn A Megeney
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada.,2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada.,3 Department of Medicine University of Ottawa Ontario Canada
| |
Collapse
|
22
|
Thiazole- and selenazole-comprising high-affinity inhibitors possess bright microsecond-scale photoluminescence in complex with protein kinase CK2. Bioorg Med Chem 2018; 26:5062-5068. [PMID: 30217463 DOI: 10.1016/j.bmc.2018.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/30/2018] [Accepted: 09/03/2018] [Indexed: 12/14/2022]
Abstract
A previously disclosed protein kinase (PK) CK2-selective inhibitor 4-(2-amino-1,3-thiazol-5-yl)benzoic acid (ATB) and its selenium-containing counterpart (ASB) revealed remarkable room temperature phosphorescence when bound to the ATP pocket of the protein kinase CK2. Conjugation of these fragments with a mimic of CK2 substrate peptide resulted in bisubstrate inhibitors with increased affinity towards the kinase. Attachment of the fluorescent acceptor dye 5-TAMRA to the conjugates led to significant enhancement of intensity of long-lifetime (microsecond-scale) photoluminescence of both sulfur- and selenium-containing compounds. The developed photoluminescent probes make possible selective determination of the concentration of CK2 in cell lysates and characterization of CK2 inhibitors by means of time-gated measurement of photoluminescence.
Collapse
|
23
|
Sharma S, Čermáková K, De Rijck J, Demeulemeester J, Fábry M, El Ashkar S, Van Belle S, Lepšík M, Tesina P, Duchoslav V, Novák P, Hubálek M, Srb P, Christ F, Řezáčová P, Hodges HC, Debyser Z, Veverka V. Affinity switching of the LEDGF/p75 IBD interactome is governed by kinase-dependent phosphorylation. Proc Natl Acad Sci U S A 2018; 115:E7053-E7062. [PMID: 29997176 PMCID: PMC6065015 DOI: 10.1073/pnas.1803909115] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lens epithelium-derived growth factor/p75 (LEDGF/p75, or PSIP1) is a transcriptional coactivator that tethers other proteins to gene bodies. The chromatin tethering function of LEDGF/p75 is hijacked by HIV integrase to ensure viral integration at sites of active transcription. LEDGF/p75 is also important for the development of mixed-lineage leukemia (MLL), where it tethers the MLL1 fusion complex at aberrant MLL targets, inducing malignant transformation. However, little is known about how the LEDGF/p75 protein interaction network is regulated. Here, we obtained solution structures of the complete interfaces between the LEDGF/p75 integrase binding domain (IBD) and its cellular binding partners and validated another binding partner, Mediator subunit 1 (MED1). We reveal that structurally conserved IBD-binding motifs (IBMs) on known LEDGF/p75 binding partners can be regulated by phosphorylation, permitting switching between low- and high-affinity states. Finally, we show that elimination of IBM phosphorylation sites on MLL1 disrupts the oncogenic potential of primary MLL1-rearranged leukemic cells. Our results demonstrate that kinase-dependent phosphorylation of MLL1 represents a previously unknown oncogenic dependency that may be harnessed in the treatment of MLL-rearranged leukemia.
Collapse
Affiliation(s)
| | - Kateřina Čermáková
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague 6, Czech Republic
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - Jan De Rijck
- Molecular Virology and Gene Therapy, KU Leuven, 3000 Leuven, Belgium;
| | | | - Milan Fábry
- Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague 4, Czech Republic
| | - Sara El Ashkar
- Molecular Virology and Gene Therapy, KU Leuven, 3000 Leuven, Belgium
| | - Siska Van Belle
- Molecular Virology and Gene Therapy, KU Leuven, 3000 Leuven, Belgium
| | - Martin Lepšík
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague 6, Czech Republic
| | - Petr Tesina
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague 6, Czech Republic
- Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague 4, Czech Republic
| | - Vojtěch Duchoslav
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague 6, Czech Republic
| | - Petr Novák
- Institute of Microbiology of the Czech Academy of Sciences, 142 20 Prague 4, Czech Republic
| | - Martin Hubálek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague 6, Czech Republic
| | - Pavel Srb
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague 6, Czech Republic
| | - Frauke Christ
- Molecular Virology and Gene Therapy, KU Leuven, 3000 Leuven, Belgium
| | - Pavlína Řezáčová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague 6, Czech Republic
- Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague 4, Czech Republic
| | - H Courtney Hodges
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
- Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Zeger Debyser
- Molecular Virology and Gene Therapy, KU Leuven, 3000 Leuven, Belgium;
| | - Václav Veverka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague 6, Czech Republic;
- Department of Cell Biology, Faculty of Science, Charles University, 116 36 Prague 1, Czech Republic
| |
Collapse
|
24
|
N6-Furfuryladenine is protective in Huntington's disease models by signaling huntingtin phosphorylation. Proc Natl Acad Sci U S A 2018; 115:E7081-E7090. [PMID: 29987005 DOI: 10.1073/pnas.1801772115] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The huntingtin N17 domain is a modulator of mutant huntingtin toxicity and is hypophosphorylated in Huntington's disease (HD). We conducted high-content analysis to find compounds that could restore N17 phosphorylation. One lead compound from this screen was N6-furfuryladenine (N6FFA). N6FFA was protective in HD model neurons, and N6FFA treatment of an HD mouse model corrects HD phenotypes and eliminates cortical mutant huntingtin inclusions. We show that N6FFA restores N17 phosphorylation levels by being salvaged to a triphosphate form by adenine phosphoribosyltransferase (APRT) and used as a phosphate donor by casein kinase 2 (CK2). N6FFA is a naturally occurring product of oxidative DNA damage. Phosphorylated huntingtin functionally redistributes and colocalizes with CK2, APRT, and N6FFA DNA adducts at sites of induced DNA damage. We present a model in which this natural product compound is salvaged to provide a triphosphate substrate to signal huntingtin phosphorylation via CK2 during low-ATP stress under conditions of DNA damage, with protective effects in HD model systems.
Collapse
|
25
|
Schnitzler A, Gratz A, Bollacke A, Weyrich M, Kuckländer U, Wünsch B, Götz C, Niefind K, Jose J. A π-Halogen Bond of Dibenzofuranones with the Gatekeeper Phe113 in Human Protein Kinase CK2 Leads to Potent Tight Binding Inhibitors. Pharmaceuticals (Basel) 2018; 11:ph11010023. [PMID: 29462988 PMCID: PMC5874719 DOI: 10.3390/ph11010023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 01/05/2023] Open
Abstract
Human protein kinase CK2 is an emerging target for neoplastic diseases. Potent lead structures for human CK2 inhibitors are derived from dibenzofuranones. Two new derivatives, 7,9-dichloro-1,2-dihydro-8-hydroxy-4-[(4-methoxyphenylamino)-methylene]dibenzo[b,d]furan-3(2H)-one (4a) and (E)-1,3-dichloro-6-[(4-methoxyphenylimino)-methyl]dibenzo[b,d]furan-2,7-diol (5) were tested for inhibition of CK2 and induction of apoptosis in LNCaP cells. Both turned out to be tight binding inhibitors, with IC50 values of 7 nM (4a) and 5 nM (5) and an apparent Ki value of 0.4 nM for both. Compounds 4a and 5 reduced cellular CK2 activity, indicating cell permeability. Cell viability was substantially impaired in LNCaP cells, as well as apoptosis was induced, which was not appearing in non-neoplastic ARPE-19 cells. Co-crystallization of 4a and 5 revealed an unexpected π-halogen bond of the chloro substituent at C9 with the gatekeeper amino acid Phe113, leading to an inverted binding mode in comparison to parent compound 4b, with the Cl at C6 instead, which was co-crystallized as a control. This indicates that the position of the chloro substituent on ring A of the dibenzofuran scaffold is responsible for an inversion of the binding mode that enhances potency.
Collapse
Affiliation(s)
- Alexander Schnitzler
- Institut für Biochemie, Department für Chemie, Universität zu Köln, Zülpicher Straße 47, D-50674 Köln, Germany.
| | - Andreas Gratz
- Institut für Pharmazeutische und Medizinische Chemie, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany.
| | - Andre Bollacke
- Institut für Pharmazeutische und Medizinische Chemie, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany.
| | - Michael Weyrich
- Medizinische Biochemie und Molekularbiologie, Universität des Saarlandes, Kirrberger Str., Geb. 44, D-66421 Homburg, Germany.
| | - Uwe Kuckländer
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany.
| | - Claudia Götz
- Medizinische Biochemie und Molekularbiologie, Universität des Saarlandes, Kirrberger Str., Geb. 44, D-66421 Homburg, Germany.
| | - Karsten Niefind
- Institut für Biochemie, Department für Chemie, Universität zu Köln, Zülpicher Straße 47, D-50674 Köln, Germany.
| | - Joachim Jose
- Institut für Pharmazeutische und Medizinische Chemie, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany.
| |
Collapse
|
26
|
Kumar S, Cieplak P. Effect of phosphorylation and single nucleotide polymorphisms on caspase substrates processing. Apoptosis 2018; 23:194-200. [DOI: 10.1007/s10495-018-1442-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
27
|
Thomas ME, Grinshpon R, Swartz P, Clark AC. Modifications to a common phosphorylation network provide individualized control in caspases. J Biol Chem 2018; 293:5447-5461. [PMID: 29414778 DOI: 10.1074/jbc.ra117.000728] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/24/2018] [Indexed: 11/06/2022] Open
Abstract
Caspase-3 activation and function have been well-defined during programmed cell death, but caspase activity, at low levels, is also required for developmental processes such as lymphoid proliferation and erythroid differentiation. Post-translational modification of caspase-3 is one method used by cells to fine-tune activity below the threshold required for apoptosis, but the allosteric mechanism that reduces activity is unknown. Phosphorylation of caspase-3 at a conserved allosteric site by p38-MAPK (mitogen-activated protein kinase) promotes survival in human neutrophils, and the modification of the loop is thought to be a key regulator in many developmental processes. We utilized phylogenetic, structural, and biophysical studies to define the interaction networks that facilitate the allosteric mechanism in caspase-3. We show that, within the modified loop, Ser150 evolved with the apoptotic caspases, whereas Thr152 is a more recent evolutionary event in mammalian caspase-3. Substitutions at Ser150 result in a pH-dependent decrease in dimer stability, and localized changes in the modified loop propagate to the active site of the same protomer through a connecting surface helix. Likewise, a cluster of hydrophobic amino acids connects the conserved loop to the active site of the second protomer. The presence of Thr152 in the conserved loop introduces a "kill switch" in mammalian caspase-3, whereas the more ancient Ser150 reduces without abolishing enzyme activity. These data reveal how evolutionary changes in a conserved allosteric site result in a common pathway for lowering activity during development or a more recent cluster-specific switch to abolish activity.
Collapse
Affiliation(s)
- Melvin E Thomas
- From the Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina 27608 and
| | - Robert Grinshpon
- From the Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina 27608 and
| | - Paul Swartz
- From the Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina 27608 and
| | - A Clay Clark
- the Department of Biology, University of Texas, Arlington, Texas 76019
| |
Collapse
|
28
|
Rabalski AJ, Gyenis L, Litchfield DW. Molecular Pathways: Emergence of Protein Kinase CK2 (CSNK2) as a Potential Target to Inhibit Survival and DNA Damage Response and Repair Pathways in Cancer Cells. Clin Cancer Res 2018; 22:2840-7. [PMID: 27306791 DOI: 10.1158/1078-0432.ccr-15-1314] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/04/2016] [Indexed: 11/16/2022]
Abstract
Protein kinase CK2 (designated CSNK2) is a constitutively active protein kinase with a vast repertoire of putative substrates that has been implicated in several human cancers, including cancer of the breast, lung, colon, and prostate, as well as hematologic malignancies. On the basis of these observations, CSNK2 has emerged as a candidate for targeted therapy, with two CSNK2 inhibitors in ongoing clinical trials. CX-4945 is a bioavailable small-molecule ATP-competitive inhibitor targeting its active site, and CIGB-300 is a cell-permeable cyclic peptide that prevents phosphorylation of the E7 protein of HPV16 by CSNK2. In preclinical models, either of these inhibitors exhibit antitumor efficacy. Furthermore, in combinations with chemotherapeutics such as cisplatin or gemcitabine, either CX-4945 or CIGB-300 promote synergistic induction of apoptosis. While CSNK2 is a regulatory participant in many processes related to cancer, its potential to modulate caspase action may be particularly pertinent to its emergence as a therapeutic target. Because the substrate recognition motifs for CSNK2 and caspases are remarkably similar, CSNK2 can block the cleavage of many caspase substrates through the phosphorylation of sites adjacent to cleavage sites. Phosphoproteomic strategies have also revealed previously underappreciated roles for CSNK2 in the phosphorylation of several key constituents of DNA damage and DNA repair pathways. Going forward, applications of proteomic strategies to interrogate responses to CSNK2 inhibitors are expected to reveal signatures for CSNK2 inhibition and molecular insights to guide new strategies to interfere with its potential to inhibit caspase action or enhance the susceptibility of cancer cells to DNA damage. Clin Cancer Res; 22(12); 2840-7. ©2016 AACR.
Collapse
Affiliation(s)
- Adam J Rabalski
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Laszlo Gyenis
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - David W Litchfield
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada. Department of Oncology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
29
|
CK2β regulates thrombopoiesis and Ca2+-triggered platelet activation in arterial thrombosis. Blood 2017; 130:2774-2785. [DOI: 10.1182/blood-2017-05-784413] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/12/2017] [Indexed: 02/06/2023] Open
Abstract
Key Points
CK2β is critically required for thrombopoiesis by regulating tubulin polymerization, MK fragmentation, and proplatelet formation. CK2β facilitates inositol triphosphate–mediated increase of cytosolic Ca2+ and is essential for platelet activation in arterial thrombosis in vivo.
Collapse
|
30
|
Burgon PG, Megeney LA. Caspase signaling, a conserved inductive cue for metazoan cell differentiation. Semin Cell Dev Biol 2017; 82:96-104. [PMID: 29129746 DOI: 10.1016/j.semcdb.2017.11.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/16/2022]
Abstract
Caspase signaling pathways were originally discovered as conveyors of programmed cell death, yet a compendium of research over the past two decades have demonstrated that these same conduits have a plethora of physiologic functions. Arguably the most extensive non-death activity that has been attributed to this protease clade is the capacity to induce cell differentiation. Caspase control of differentiation is conserved across diverse metazoan organisms from flies to humans, suggesting an ancient origin for this form of cell fate control. Here we discuss the mechanisms by which caspase enzymes manage differentiation, the targeted substrates that may be common across cell lineages, and the countervailing signals that may be essential for these proteases to 'execute' this non-death cell fate.
Collapse
Affiliation(s)
- Patrick G Burgon
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada; Department of Medicine, Division of Cardiology, University of Ottawa, Ottawa, Ontario, Canada.
| | - Lynn A Megeney
- Regenerative Medicine Program, Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada; Department of Medicine, Division of Cardiology, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
31
|
Serrano BP, Szydlo HS, Alfandari D, Hardy JA. Active site-adjacent phosphorylation at Tyr-397 by c-Abl kinase inactivates caspase-9. J Biol Chem 2017; 292:21352-21365. [PMID: 29066624 DOI: 10.1074/jbc.m117.811976] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/11/2017] [Indexed: 12/11/2022] Open
Abstract
Caspase-9 (casp-9) is an initiator caspase and plays a central role in activating apoptotic cell death. Control of all caspases is tightly regulated by a series of phosphorylation events enacted by several different kinases. Caspase-9 is the most heavily phosphorylated of all caspases, with phosphorylation of at least 11 distinct residues in all three caspase-9 domains by nine kinases. Caspase-9 phosphorylation by the non-receptor tyrosine kinase c-Abl at Tyr-153 reportedly leads to caspase-9 activation. All other phosphorylation events on caspases have been shown to block proteolytic function by a number of mechanisms, so we sought to unravel the molecular mechanism of the putative caspase-9 activation by phosphorylation. Surprisingly, we observed no evidence for Tyr-153 phosphorylation of caspase-9 in vitro or in cells, suggesting that Tyr-153 is not phosphorylated by c-Abl. Instead, we identified a new site for c-Abl-mediated phosphorylation, Tyr-397. This residue is adjacent to the caspase-9 active site but, as a member of the second shell, not a residue that directly contacts substrate. Our results further indicate that Tyr-397 is the dominant site of c-Abl phosphorylation both in vitro and upon c-Abl activation in cells. Of note, phosphorylation at this site inhibits caspase-9 activity, and the bulk of the added phosphate moiety appeared to directly block substrate binding. c-Abl plays both proapoptotic and prosurvival roles, and our findings suggest that c-Abl's effects on caspase-9 activity promote the prosurvival mode.
Collapse
Affiliation(s)
| | - Hannah S Szydlo
- Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003
| | - Dominique Alfandari
- Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003
| | | |
Collapse
|
32
|
Buontempo F, McCubrey JA, Orsini E, Ruzzene M, Cappellini A, Lonetti A, Evangelisti C, Chiarini F, Evangelisti C, Barata JT, Martelli AM. Therapeutic targeting of CK2 in acute and chronic leukemias. Leukemia 2017; 32:1-10. [PMID: 28951560 PMCID: PMC5770594 DOI: 10.1038/leu.2017.301] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/06/2017] [Accepted: 09/08/2017] [Indexed: 12/22/2022]
Abstract
CK2 is a ubiquitously expressed, constitutively active Ser/Thr protein kinase, which is considered the most pleiotropic protein kinase in the human kinome. Such a pleiotropy explains the involvement of CK2 in many cellular events. However, its predominant roles are stimulation of cell growth and prevention of apoptosis. High levels of CK2 messenger RNA and protein are associated with CK2 pathological functions in human cancers. Over the last decade, basic and translational studies have provided evidence of CK2 as a pivotal molecule driving the growth of different blood malignancies. CK2 overexpression has been demonstrated in nearly all the types of hematological cancers, including acute and chronic leukemias, where CK2 is a key regulator of signaling networks critical for cell proliferation, survival and drug resistance. The findings that emerged from these studies suggest that CK2 could be a valuable therapeutic target in leukemias and supported the initiation of clinical trials using CK2 antagonists. In this review, we summarize the recent advances on the understanding of the signaling pathways involved in CK2 inhibition-mediated effects with a particular emphasis on the combinatorial use of CK2 inhibitors as novel therapeutic strategies for treating both acute and chronic leukemia patients.
Collapse
Affiliation(s)
- F Buontempo
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - J A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - E Orsini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - M Ruzzene
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - A Cappellini
- Department of Human, Social and Health Sciences, University of Cassino, Cassino, Italy
| | - A Lonetti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - C Evangelisti
- Institute of Molecular Genetics, National Research Council, Bologna, Italy.,Cell and Molecular Biology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - F Chiarini
- Institute of Molecular Genetics, National Research Council, Bologna, Italy.,Cell and Molecular Biology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - C Evangelisti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - J T Barata
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - A M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
33
|
Rahnel H, Viht K, Lavogina D, Mazina O, Haljasorg T, Enkvist E, Uri A. A Selective Biligand Inhibitor of CK2 Increases Caspase-3 Activity in Cancer Cells and Inhibits Platelet Aggregation. ChemMedChem 2017; 12:1723-1736. [PMID: 28837260 DOI: 10.1002/cmdc.201700457] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Indexed: 11/08/2022]
Abstract
Cancer cells express high levels of CK2, and its inhibition leads to apoptosis. CK2 has therefore emerged as a new drug target for cancer therapy. A biligand inhibitor ARC-772 was constructed by conjugating 4-(2-amino-1,3-thiazol-5-yl)benzoic acid and a carboxylate-rich peptoid. ARC-772 was found to bind CK2 with a Kd value of 0.3 nm and showed remarkable CK2 inhibitory selectivity in a panel of 140 protein kinases (Gini coefficient: 0.75 at c=100 nm). ARC-775, the acetoxymethyl ester prodrug of ARC-772, was efficiently taken up by cells. Once internalized, the inhibitor is activated by cellular esterase activity. In HeLa cancer cells ARC-775 was found to activate caspase-3 (an apoptosis marker) at sub-micromolar concentrations (EC50 =0.3 μm), a 20-fold lower extracellular concentration than CX-4945, the only CK2 inhibitor under clinical trials. At micromolar concentrations, ARC-775 was also found to inhibit ADP-induced aggregation of human platelets. The overall results of this study demonstrate that oligo-anionic biligand inhibitors have good potential for drug development.
Collapse
Affiliation(s)
- Hedi Rahnel
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411, Tartu, Estonia
| | - Kaido Viht
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411, Tartu, Estonia
| | - Darja Lavogina
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411, Tartu, Estonia
| | - Olga Mazina
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411, Tartu, Estonia
| | - Tõiv Haljasorg
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411, Tartu, Estonia
| | - Erki Enkvist
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411, Tartu, Estonia
| | - Asko Uri
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411, Tartu, Estonia
| |
Collapse
|
34
|
Padilla-Benavides T, Nasipak BT, Paskavitz AL, Haokip DT, Schnabl JM, Nickerson JA, Imbalzano AN. Casein kinase 2-mediated phosphorylation of Brahma-related gene 1 controls myoblast proliferation and contributes to SWI/SNF complex composition. J Biol Chem 2017; 292:18592-18607. [PMID: 28939766 PMCID: PMC5682968 DOI: 10.1074/jbc.m117.799676] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/06/2017] [Indexed: 01/01/2023] Open
Abstract
Transcriptional regulation is modulated in part by chromatin-remodeling enzymes that control gene accessibility by altering chromatin compaction or nucleosome positioning. Brahma-related gene 1 (Brg1), a catalytic subunit of the mammalian SWI/SNF chromatin-remodeling enzymes, is required for both myoblast proliferation and differentiation, and the control of Brg1 phosphorylation by calcineurin, PKCβ1, and p38 regulates the transition to differentiation. However, we hypothesized that Brg1 activity might be regulated by additional kinases. Here, we report that Brg1 is also a target of casein kinase 2 (CK2), a serine/threonine kinase, in proliferating myoblasts. We found that CK2 interacts with Brg1, and mutation of putative phosphorylation sites to non-phosphorylatable (Ser to Ala, SA) or phosphomimetic residues (Ser to Glu, SE) reduced Brg1 phosphorylation by CK2. Although BRG1-deleted myoblasts that ectopically express the SA-Brg1 mutant proliferated similarly to the parental cells or cells ectopically expressing wild-type (WT) Brg1, ectopic expression of the SE-Brg1 mutant reduced proliferation and increased cell death, similar to observations from cells lacking Brg1. Moreover, pharmacological inhibition of CK2 increased myoblast proliferation. Furthermore, the Pax7 promoter, which controls expression of a key transcription factor required for myoblast proliferation, was in an inaccessible chromatin state in the SE-Brg1 mutant, suggesting that hyperphosphorylated Brg1 cannot remodel chromatin. WT-, SA-, and SE-Brg1 exhibited distinct differences in interacting with and affecting expression of the SWI/SNF subunits Baf155 and Baf170 and displayed differential sub-nuclear localization. Our results indicate that CK2-mediated phosphorylation of Brg1 regulates myoblast proliferation and provides insight into one mechanism by which composition of the mammalian SWI/SNF enzyme complex is regulated.
Collapse
Affiliation(s)
- Teresita Padilla-Benavides
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - Brian T Nasipak
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - Amanda L Paskavitz
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - Dominic T Haokip
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - Jake M Schnabl
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - Jeffrey A Nickerson
- the Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Anthony N Imbalzano
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| |
Collapse
|
35
|
Abdul-Ghani M, Suen C, Jiang B, Deng Y, Weldrick JJ, Putinski C, Brunette S, Fernando P, Lee TT, Flynn P, Leenen FHH, Burgon PG, Stewart DJ, Megeney LA. Cardiotrophin 1 stimulates beneficial myogenic and vascular remodeling of the heart. Cell Res 2017; 27:1195-1215. [PMID: 28785017 PMCID: PMC5630684 DOI: 10.1038/cr.2017.87] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/06/2017] [Accepted: 06/21/2017] [Indexed: 12/12/2022] Open
Abstract
The post-natal heart adapts to stress and overload through hypertrophic growth, a process that may be pathologic or beneficial (physiologic hypertrophy). Physiologic hypertrophy improves cardiac performance in both healthy and diseased individuals, yet the mechanisms that propagate this favorable adaptation remain poorly defined. We identify the cytokine cardiotrophin 1 (CT1) as a factor capable of recapitulating the key features of physiologic growth of the heart including transient and reversible hypertrophy of the myocardium, and stimulation of cardiomyocyte-derived angiogenic signals leading to increased vascularity. The capacity of CT1 to induce physiologic hypertrophy originates from a CK2-mediated restraining of caspase activation, preventing the transition to unrestrained pathologic growth. Exogenous CT1 protein delivery attenuated pathology and restored contractile function in a severe model of right heart failure, suggesting a novel treatment option for this intractable cardiac disease.
Collapse
Affiliation(s)
- Mohammad Abdul-Ghani
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Colin Suen
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Baohua Jiang
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada
| | - Yupu Deng
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada
| | - Jonathan J Weldrick
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Charis Putinski
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Steve Brunette
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada
| | - Pasan Fernando
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Biology, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Tom T Lee
- Fate Therapeutics Inc., 3535 General Atomics Court Suite 200, San Diego, CA 92121, USA
| | - Peter Flynn
- Fate Therapeutics Inc., 3535 General Atomics Court Suite 200, San Diego, CA 92121, USA
| | - Frans H H Leenen
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Patrick G Burgon
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Duncan J Stewart
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Lynn A Megeney
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
36
|
Okumu DO, East MP, Levine M, Herring LE, Zhang R, Gilbert TSK, Litchfield DW, Zhang Y, Graves LM. BIRC6 mediates imatinib resistance independently of Mcl-1. PLoS One 2017; 12:e0177871. [PMID: 28520795 PMCID: PMC5433768 DOI: 10.1371/journal.pone.0177871] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 05/04/2017] [Indexed: 12/13/2022] Open
Abstract
Baculoviral IAP repeat containing 6 (BIRC6) is a member of the inhibitors of apoptosis proteins (IAPs), a family of functionally and structurally related proteins that inhibit apoptosis. BIRC6 has been implicated in drug resistance in several different human cancers, however mechanisms regulating BIRC6 have not been extensively explored. Our phosphoproteomic analysis of an imatinib-resistant chronic myelogenous leukemia (CML) cell line (MYL-R) identified increased amounts of a BIRC6 peptide phosphorylated at S480, S482, and S486 compared to imatinib-sensitive CML cells (MYL). Thus we investigated the role of BIRC6 in mediating imatinib resistance and compared it to the well-characterized anti-apoptotic protein, Mcl-1. Both BIRC6 and Mcl-1 were elevated in MYL-R compared to MYL cells. Lentiviral shRNA knockdown of BIRC6 in MYL-R cells increased imatinib-stimulated caspase activation and resulted in a ~20-25-fold increase in imatinib sensitivity, without affecting Mcl-1. Treating MYL-R cells with CDK9 inhibitors decreased BIRC6 mRNA, but not BIRC6 protein levels. By contrast, while CDK9 inhibitors reduced Mcl-1 mRNA and protein, they did not affect imatinib sensitivity. Since the Src family kinase Lyn is highly expressed and active in MYL-R cells, we tested the effects of Lyn inhibition on BIRC6 and Mcl-1. RNAi-mediated knockdown or inhibition of Lyn (dasatinib/ponatinib) reduced BIRC6 protein stability and increased caspase activation. Inhibition of Lyn also increased formation of an N-terminal BIRC6 fragment in parallel with reduced amount of the BIRC6 phosphopeptide, suggesting that Lyn may regulate BIRC6 phosphorylation and stability. In summary, our data show that BIRC6 stability is dependent on Lyn, and that BIRC6 mediates imatinib sensitivity independently of Mcl-1 or CDK9. Hence, BIRC6 may be a novel target for the treatment of drug-resistant CML where Mcl-1 or CDK9 inhibitors have failed.
Collapse
Affiliation(s)
- Denis O. Okumu
- Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Michael P. East
- Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Merlin Levine
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Laura E. Herring
- Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- UNC Michael Hooker Proteomics Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Raymond Zhang
- Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Thomas S. K. Gilbert
- Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- UNC Michael Hooker Proteomics Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - David W. Litchfield
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Yanping Zhang
- Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Lee M. Graves
- Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
- UNC Michael Hooker Proteomics Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
37
|
The Impacts of Chrysanthemum indicum Extract on Oxidative Stress and Inflammatory Responses in Adjuvant-Induced Arthritic Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:3285394. [PMID: 28491105 PMCID: PMC5402234 DOI: 10.1155/2017/3285394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/24/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023]
Abstract
Chrysanthemum indicum has been used as a therapeutic agent against inflammation, hypertension, and respiratory conditions for many years. This research's aim has been to examine the antioxidant impacts that Chrysanthemum indicum extract (CIE) has on the oxidative stress and inflammatory responses in adjuvant-induced arthritic (AA) rats. 40 rats were categorised into 4 groups according to a completely randomized approach: Group I involved normal control rats (CTRL) that received a basal diet; Group II involved arthritic control rats (CTRL-AA) that received the same diet; Group III involved rats that received a basal diet and 30 mg/kg CIE; and Group IV involved arthritic rats with the same diet as Group III rats (CIE-AA). After injection with complete Freund's adjuvant, body weight, arthritis score, and the serum levels of TNF-α, IL-1β, IL-6, myeloperoxidase (MPO), malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GSH-PX) were assessed. The results demonstrated that CIE delayed the onset time of arthritis and decreased the clinical arthritis severity score (P < 0.05). Observations of CIE-AA and CTRL-AA rats demonstrated that CIE alleviates oxidative stress and inflammatory responses in CIE-AA group. In conclusion, CIE alleviated oxidative stress and inflammatory responses, thereby highlighting its potential use as a candidate for clinical treatments of rheumatoid arthritis.
Collapse
|
38
|
Evolution of caspase-mediated cell death and differentiation: twins separated at birth. Cell Death Differ 2017; 24:1359-1368. [PMID: 28338655 PMCID: PMC5520454 DOI: 10.1038/cdd.2017.37] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/16/2017] [Accepted: 02/20/2017] [Indexed: 12/28/2022] Open
Abstract
The phenotypic and biochemical similarities between caspase-mediated apoptosis and cellular differentiation are striking. They include such diverse phenomenon as mitochondrial membrane perturbations, cytoskeletal rearrangements and DNA fragmentation. The parallels between the two disparate processes suggest some common ancestry and highlight the paradoxical nature of the death-centric view of caspases. That is, what is the driving selective pressure that sustains death-inducing proteins throughout eukaryotic evolution? Plausibly, caspase function may be rooted in a primordial non-death function, such as cell differentiation, and was co-opted for its role in programmed cell death. This review will delve into the links between caspase-mediated apoptosis and cell differentiation and examine the distinguishing features of these events. More critically, we chronicle the evolutionary origins of caspases and propose that caspases may have held an ancient role in mediating the fidelity of cell division/differentiation through its effects on proteostasis and protein quality control.
Collapse
|
39
|
Nuñez de Villavicencio-Diaz T, Rabalski AJ, Litchfield DW. Protein Kinase CK2: Intricate Relationships within Regulatory Cellular Networks. Pharmaceuticals (Basel) 2017; 10:ph10010027. [PMID: 28273877 PMCID: PMC5374431 DOI: 10.3390/ph10010027] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 02/25/2017] [Accepted: 03/02/2017] [Indexed: 01/20/2023] Open
Abstract
Protein kinase CK2 is a small family of protein kinases that has been implicated in an expanding array of biological processes. While it is widely accepted that CK2 is a regulatory participant in a multitude of fundamental cellular processes, CK2 is often considered to be a constitutively active enzyme which raises questions about how it can be a regulatory participant in intricately controlled cellular processes. To resolve this apparent paradox, we have performed a systematic analysis of the published literature using text mining as well as mining of proteomic databases together with computational assembly of networks that involve CK2. These analyses reinforce the notion that CK2 is involved in a broad variety of biological processes and also reveal an extensive interplay between CK2 phosphorylation and other post-translational modifications. The interplay between CK2 and other post-translational modifications suggests that CK2 does have intricate roles in orchestrating cellular events. In this respect, phosphorylation of specific substrates by CK2 could be regulated by other post-translational modifications and CK2 could also have roles in modulating other post-translational modifications. Collectively, these observations suggest that the actions of CK2 are precisely coordinated with other constituents of regulatory cellular networks.
Collapse
Affiliation(s)
| | - Adam J Rabalski
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| | - David W Litchfield
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
- Department of Oncology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| |
Collapse
|
40
|
Larsen BD, Sørensen CS. The caspase-activated DNase: apoptosis and beyond. FEBS J 2016; 284:1160-1170. [PMID: 27865056 DOI: 10.1111/febs.13970] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/04/2016] [Accepted: 11/17/2016] [Indexed: 01/13/2023]
Abstract
Organismal development and function requires multiple and accurate signal transduction pathways to ensure that proper balance between cell proliferation, differentiation, inactivation, and death is achieved. Cell death via apoptotic caspase signal transduction is extensively characterized and integral to this balance. Importantly, the view of apoptotic signal transduction has expanded over the previous decades. Subapoptotic caspase signaling has surfaced as mechanism that can promote the adoption of a range of cellular fates. An emerging mechanism of subapoptotic caspase signaling is the activation of the caspase-activated DNase (CAD) through controlled cleavage of the inhibitor of CAD (ICAD). CAD-induced DNA breaks incite a DNA damage response, frequently invoking p53 signaling, that transduces a change in cell fate. Cell differentiation and senescence are fates demonstrated to arise from CAD-induced DNA breaks. Furthermore, an apparent consequence of CAD activity is also emerging, as a potential source of oncogenic mutations. This review will discuss the mechanisms underlying CAD-induced DNA breaks and highlight how CAD activity promotes diverse cell fates.
Collapse
Affiliation(s)
- Brian D Larsen
- Biotech Research and Innovation Centre, University of Copenhagen, Denmark
| | - Claus S Sørensen
- Biotech Research and Innovation Centre, University of Copenhagen, Denmark
| |
Collapse
|
41
|
Eron SJ, Raghupathi K, Hardy JA. Dual Site Phosphorylation of Caspase-7 by PAK2 Blocks Apoptotic Activity by Two Distinct Mechanisms. Structure 2016; 25:27-39. [PMID: 27889207 DOI: 10.1016/j.str.2016.11.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/20/2016] [Accepted: 10/27/2016] [Indexed: 01/23/2023]
Abstract
Caspases, the cysteine proteases that execute apoptosis, are tightly regulated via phosphorylation by a series of kinases. Although all apoptotic caspases work in concert to promote apoptosis, different kinases regulate individual caspases. Several sites of caspase-7 phosphorylation have been reported, but without knowing the molecular details, it has been impossible to exploit or control these complex interactions, which normally prevent unwanted proliferation. During dysregulation, PAK2 kinase plays an alternative anti-apoptotic role, phosphorylating caspase-7 and promoting unfettered cell growth and chemotherapeutic resistance. PAK2 phosphorylates caspase-7 at two sites, inhibiting activity using two different molecular mechanisms, before and during apoptosis. Phosphorylation of caspase-7 S30 allosterically obstructs its interaction with caspase-9, preventing intersubunit linker processing, slowing or preventing caspase-7 activation. S239 phosphorylation renders active caspase-7 incapable of binding substrate, blocking later events in apoptosis. Each of these mechanisms is novel, representing new opportunities for synergistic control of caspases and their counterpart kinases.
Collapse
Affiliation(s)
- Scott J Eron
- Department of Chemistry, University of Massachusetts Amherst, 104 LGRT, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Kishore Raghupathi
- Department of Chemistry, University of Massachusetts Amherst, 104 LGRT, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Jeanne A Hardy
- Department of Chemistry, University of Massachusetts Amherst, 104 LGRT, 710 North Pleasant Street, Amherst, MA 01003, USA.
| |
Collapse
|
42
|
Luissint AC, Parkos CA, Nusrat A. Inflammation and the Intestinal Barrier: Leukocyte-Epithelial Cell Interactions, Cell Junction Remodeling, and Mucosal Repair. Gastroenterology 2016; 151:616-32. [PMID: 27436072 PMCID: PMC5317033 DOI: 10.1053/j.gastro.2016.07.008] [Citation(s) in RCA: 390] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/13/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023]
Abstract
The intestinal tract is lined by a single layer of columnar epithelial cells that forms a dynamic, permeable barrier allowing for selective absorption of nutrients, while restricting access to pathogens and food-borne antigens. Precise regulation of epithelial barrier function is therefore required for maintaining mucosal homeostasis and depends, in part, on barrier-forming elements within the epithelium and a balance between pro- and anti-inflammatory factors in the mucosa. Pathologic states, such as inflammatory bowel disease, are associated with a leaky epithelial barrier, resulting in excessive exposure to microbial antigens, recruitment of leukocytes, release of soluble mediators, and ultimately mucosal damage. An inflammatory microenvironment affects epithelial barrier properties and mucosal homeostasis by altering the structure and function of epithelial intercellular junctions through direct and indirect mechanisms. We review our current understanding of complex interactions between the intestinal epithelium and immune cells, with a focus on pathologic mucosal inflammation and mechanisms of epithelial repair. We discuss leukocyte-epithelial interactions, as well as inflammatory mediators that affect the epithelial barrier and mucosal repair. Increased knowledge of communication networks between the epithelium and immune system will lead to tissue-specific strategies for treating pathologic intestinal inflammation.
Collapse
Affiliation(s)
- Anny-Claude Luissint
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|
43
|
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: 25] [Impact Index Per Article: 2.8] [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
|
44
|
Abstract
Compensatory growth and regeneration of skeletal muscle is dependent on the resident stem cell population, satellite cells (SCs). Self-renewal and maintenance of the SC niche is coordinated by the paired-box transcription factor Pax7, and yet continued expression of this protein inhibits the myoblast differentiation program. As such, the reduction or removal of Pax7 may denote a key prerequisite for SCs to abandon self-renewal and acquire differentiation competence. Here, we identify caspase 3 cleavage inactivation of Pax7 as a crucial step for terminating the self-renewal process. Inhibition of caspase 3 results in elevated Pax7 protein and SC self-renewal, whereas caspase activation leads to Pax7 cleavage and initiation of the myogenic differentiation program. Moreover, in vivo inhibition of caspase 3 activity leads to a profound disruption in skeletal muscle regeneration with an accumulation of SCs within the niche. We have also noted that casein kinase 2 (CK2)-directed phosphorylation of Pax7 attenuates caspase-directed cleavage. Together, these results demonstrate that SC fate is dependent on opposing posttranslational modifications of the Pax7 protein.
Collapse
|
45
|
Filhol O, Giacosa S, Wallez Y, Cochet C. Protein kinase CK2 in breast cancer: the CK2β regulatory subunit takes center stage in epithelial plasticity. Cell Mol Life Sci 2015; 72:3305-22. [PMID: 25990538 PMCID: PMC11113558 DOI: 10.1007/s00018-015-1929-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 05/06/2015] [Accepted: 05/11/2015] [Indexed: 12/11/2022]
Abstract
Structurally, protein kinase CK2 consists of two catalytic subunits (α and α') and two regulatory subunits (β), which play a critical role in targeting specific CK2 substrates. Compelling evidence shows the complexity of the CK2 cellular signaling network and supports the view that this enzyme is a key component of regulatory protein kinase networks that are involved in several aspects of cancer. CK2 both activates and suppresses the expression of a number of essential oncogenes and tumor suppressors, and its expression and activity are upregulated in blood tumors and virtually all solid tumors. The prognostic significance of CK2α expression in association with various clinicopathological parameters highlighted this kinase as an adverse prognostic marker in breast cancer. In addition, several recent studies reported its implication in the regulation of the epithelial-to-mesenchymal transition (EMT), an early step in cancer invasion and metastasis. In this review, we briefly overview the contribution of CK2 to several aspects of cancer and discuss how in mammary epithelial cells, the expression of its CK2β regulatory subunit plays a critical role in maintaining an epithelial phenotype through CK2-mediated control of key EMT-related transcription factors. Importantly, decreased CK2β expression in breast tumors is correlated with inefficient phosphorylation and nuclear translocation of Snail1 and Foxc2, ultimately leading to EMT induction. This review highlights the pivotal role played by CK2β in the mammary epithelial phenotype and discusses how a modest alteration in its expression may be sufficient to induce dramatic effects facilitating the early steps in tumor cell dissemination through the coordinated regulation of two key transcription factors.
Collapse
Affiliation(s)
- Odile Filhol
- Institut National de la Santé et de la Recherche Médicale, U1036, Grenoble, France
- Institute of Life Sciences Research and Technologies, Biology of Cancer and Infection, Commissariat à l’Energie Atomique, Grenoble, France
- Unité Mixte de Recherche-S1036, University of Grenoble Alpes, Grenoble, France
| | - Sofia Giacosa
- Institut National de la Santé et de la Recherche Médicale, U1036, Grenoble, France
- Institute of Life Sciences Research and Technologies, Biology of Cancer and Infection, Commissariat à l’Energie Atomique, Grenoble, France
- Unité Mixte de Recherche-S1036, University of Grenoble Alpes, Grenoble, France
| | - Yann Wallez
- Institut National de la Santé et de la Recherche Médicale, U1036, Grenoble, France
- Institute of Life Sciences Research and Technologies, Biology of Cancer and Infection, Commissariat à l’Energie Atomique, Grenoble, France
- Unité Mixte de Recherche-S1036, University of Grenoble Alpes, Grenoble, France
| | - Claude Cochet
- Institut National de la Santé et de la Recherche Médicale, U1036, Grenoble, France
- Institute of Life Sciences Research and Technologies, Biology of Cancer and Infection, Commissariat à l’Energie Atomique, Grenoble, France
- Unité Mixte de Recherche-S1036, University of Grenoble Alpes, Grenoble, France
| |
Collapse
|
46
|
Qaiser F, Trembley JH, Kren BT, Wu JJ, Naveed AK, Ahmed K. Protein kinase CK2 inhibition induces cell death via early impact on mitochondrial function. J Cell Biochem 2015; 115:2103-15. [PMID: 25043911 DOI: 10.1002/jcb.24887] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 07/11/2014] [Indexed: 01/03/2023]
Abstract
CK2 (official acronym for casein kinase 2 or II) is a potent suppressor of apoptosis in response to diverse apoptotic stimuli-thus its molecular downregulation or activity inhibition results in potent induction of cell death. CK2 downregulation is known to impact mitochondrial apoptotic circuitry but the underlying mechanism(s) remain unclear. Utilizing prostate cancer cell lines subjected to CK2-specific inhibitors which cause loss of cell viability, we have found that CK2 inhibition in cells causes rapid early decrease in mitochondrial membrane potential (Δψm). Cells treated with the CK2 inhibitors TBB (4,5,6,7-tetrabromobenzotriazole) or TBCA (tetrabromocinnamic acid) demonstrate changes in Δψm which become apparent within 2 h, that is, significantly prior to evidence of activation of other mitochondrial apoptotic signals whose temporal expression ensues subsequent to loss of Δψm. Further, we have demonstrated the presence of CK2 in purified mitochondria and it appears that the effect on Δψm evoked by inhibition of CK2 may involve mitochondrial localized CK2. Results also suggest that alterations in Ca(2+) signaling may be involved in the CK2 mediated regulation of Δψm and mitochondrial permeability. Thus, we propose that a key mechanism of CK2 impact on mitochondrial apoptotic circuitry and cell death involves early loss of Δψm which may be a primary trigger for apoptotic signaling and cell death resulting from CK2 inhibition.
Collapse
Affiliation(s)
- Fatima Qaiser
- Cellular and Molecular Biochemistry Research Laboratory (151), Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota, 55417; Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, 55455; Department of Biochemistry and Molecular Biology, Army Medical College, National University of Sciences and Technology, Islamabad, Pakistan
| | | | | | | | | | | |
Collapse
|
47
|
So J, Pasculescu A, Dai AY, Williton K, James A, Nguyen V, Creixell P, Schoof EM, Sinclair J, Barrios-Rodiles M, Gu J, Krizus A, Williams R, Olhovsky M, Dennis JW, Wrana JL, Linding R, Jorgensen C, Pawson T, Colwill K. Integrative analysis of kinase networks in TRAIL-induced apoptosis provides a source of potential targets for combination therapy. Sci Signal 2015; 8:rs3. [PMID: 25852190 DOI: 10.1126/scisignal.2005700] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an endogenous secreted peptide and, in preclinical studies, preferentially induces apoptosis in tumor cells rather than in normal cells. The acquisition of resistance in cells exposed to TRAIL or its mimics limits their clinical efficacy. Because kinases are intimately involved in the regulation of apoptosis, we systematically characterized kinases involved in TRAIL signaling. Using RNA interference (RNAi) loss-of-function and cDNA overexpression screens, we identified 169 protein kinases that influenced the dynamics of TRAIL-induced apoptosis in the colon adenocarcinoma cell line DLD-1. We classified the kinases as sensitizers or resistors or modulators, depending on the effect that knockdown and overexpression had on TRAIL-induced apoptosis. Two of these kinases that were classified as resistors were PX domain-containing serine/threonine kinase (PXK) and AP2-associated kinase 1 (AAK1), which promote receptor endocytosis and may enable cells to resist TRAIL-induced apoptosis by enhancing endocytosis of the TRAIL receptors. We assembled protein interaction maps using mass spectrometry-based protein interaction analysis and quantitative phosphoproteomics. With these protein interaction maps, we modeled information flow through the networks and identified apoptosis-modifying kinases that are highly connected to regulated substrates downstream of TRAIL. The results of this analysis provide a resource of potential targets for the development of TRAIL combination therapies to selectively kill cancer cells.
Collapse
Affiliation(s)
- Jonathan So
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada. Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Adrian Pasculescu
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Anna Y Dai
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Kelly Williton
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Andrew James
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Vivian Nguyen
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Pau Creixell
- Cellular Signal Integration Group (C-SIG), Technical University of Denmark (DTU), DK-2800 Lyngby, Denmark
| | - Erwin M Schoof
- Cellular Signal Integration Group (C-SIG), Technical University of Denmark (DTU), DK-2800 Lyngby, Denmark
| | - John Sinclair
- Cell Communication Team, The Institute of Cancer Research, London SW3 6JB, UK
| | - Miriam Barrios-Rodiles
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Jun Gu
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Aldis Krizus
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Ryan Williams
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Marina Olhovsky
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - James W Dennis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada. Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jeffrey L Wrana
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada. Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Rune Linding
- Cellular Signal Integration Group (C-SIG), Technical University of Denmark (DTU), DK-2800 Lyngby, Denmark. Biotech Research and Innovation Centre (BRIC), University of Copenhagen (UCPH), DK-2200 Copenhagen, Denmark.
| | - Claus Jorgensen
- Cell Communication Team, The Institute of Cancer Research, London SW3 6JB, UK.
| | - Tony Pawson
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada. Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada. Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada.
| |
Collapse
|
48
|
Pin1: Intimate involvement with the regulatory protein kinase networks in the global phosphorylation landscape. Biochim Biophys Acta Gen Subj 2015; 1850:2077-86. [PMID: 25766872 DOI: 10.1016/j.bbagen.2015.02.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 02/25/2015] [Accepted: 02/27/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Protein phosphorylation is a universal regulatory mechanism that involves an extensive network of protein kinases. The discovery of the phosphorylation-dependent peptidyl-prolyl isomerase Pin1 added an additional layer of complexity to these regulatory networks. SCOPE OF REVIEW We have evaluated interactions between Pin1 and the regulatory kinome and proline-dependent phosphoproteome taking into consideration findings from targeted studies as well as data that has emerged from systematic phosphoproteomic workflows and from curated protein interaction databases. MAJOR CONCLUSIONS The relationship between Pin1 and the regulatory protein kinase networks is not restricted simply to the recognition of proteins that are substrates for proline-directed kinases. In this respect, Pin1 itself is phosphorylated in cells by protein kinases that modulate its functional properties. Furthermore, the phosphorylation-dependent targets of Pin1 include a number of protein kinases as well as other enzymes such as phosphatases and regulatory subunits of kinases that modulate the actions of protein kinases. GENERAL SIGNIFICANCE As a result of its interactions with numerous protein kinases and their substrates, as well as itself being a target for phosphorylation, Pin1 has an intricate relationship with the regulatory protein kinase and phosphoproteomic networks that orchestrate complex cellular processes and respond to environmental cues. This article is part of a Special Issue entitled Proline-directed Foldases: Cell Signaling Catalysts and Drug Targets.
Collapse
|
49
|
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
|
50
|
Ko CC, Chen YJ, Chen CT, Liu YC, Cheng FC, Hsu KC, Chow LP. Chemical proteomics identifies heterogeneous nuclear ribonucleoprotein (hnRNP) A1 as the molecular target of quercetin in its anti-cancer effects in PC-3 cells. J Biol Chem 2014; 289:22078-89. [PMID: 24962584 DOI: 10.1074/jbc.m114.553248] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Quercetin, a flavonoid abundantly present in plants, is widely used as a phytotherapy in prostatitis and prostate cancer. Although quercetin has been reported to have a number of therapeutic effects, the cellular target(s) responsible for its anti-cancer action has not yet been clearly elucidated. Here, employing affinity chromatography and mass spectrometry, we identified heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) as a direct target of quercetin. A specific interaction between quercetin and hnRNPA1 was validated by immunoblotting and in vitro binding experiments. We found that quercetin bound the C-terminal region of hnRNPA1, impairing the ability of hnRNPA1 to shuttle between the nucleus and cytoplasm and ultimately resulting in its cytoplasmic retention. In addition, hnRNPA1 was recruited to stress granules after treatment of cells with quercetin for up to 48 h, and the levels of cIAP1 (cellular inhibitor of apoptosis), an internal ribosome entry site translation-dependent protein, were reduced by hnRNPA1 regulation. This is the first report that anti-cancer effects of quercetin are mediated, in part, by impairing functions of hnRNPA1, insights that were obtained using a chemical proteomics strategy.
Collapse
Affiliation(s)
- Chia-Chen Ko
- From the Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan and
| | - Yun-Ju Chen
- From the Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan and
| | - Chih-Ta Chen
- From the Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan and
| | - Yu-Chih Liu
- From the Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan and
| | - Fong-Chi Cheng
- Eurofins Panlabs Taiwan Ltd., 158 Li-Teh Road, Peitou, Taipei 112, Taiwan
| | - Kai-Chao Hsu
- From the Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan and
| | - Lu-Ping Chow
- From the Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan and
| |
Collapse
|