1
|
Ranjbaran R, Abbasi M, Rafiei Dehbidi G, Seyyedi N, Behzad-Behbahani A, Sharifzadeh S. Phosflow assessment of PDGFRA phosphorylation state: A guide for tyrosine kinase inhibitor targeted therapy in hypereosinophilia patients. Cytometry A 2021; 99:784-792. [PMID: 33386673 DOI: 10.1002/cyto.a.24302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/14/2020] [Accepted: 12/28/2020] [Indexed: 11/06/2022]
Abstract
Clonal eosinophilia is a hematologic disorder caused by translocation in growth factor receptor (GFR) genes. Despite the identified molecular mechanisms underlying clonal hypereosinophilia, the distinction between clonal and reactive eosinophilia has remained challenging due to the diversity of partner genes for translocated GFRs. This study aimed to examine the feasibility of phosphoflow cytometry in the diagnosis of clonal hypereosinophilia through evaluating the level of platelet-derived growth factor receptor alpha (PDGFRA) phosphorylation and its correlation with PDGFRA genetic aberration. Blood samples were collected from 45 hypereosinophilia patients and 10 healthy controls. Using phosphoflow cytometry method, the phosphorylation state of PDGFRA was assessed. The specificity of phosflow results was confirmed by western blotting and eventually compared with qRT-PCR expression analysis of 3'-region of PDGFRA. To detect the genetic aberration of PDGFRA, 5'-rapid amplification of cDNA ends (5'-RACE) was performed. Phosflow analysis illustrated that 9 of 45 hypereosinophilic patients had higher level of PDGFRA phosphorylation while sequence analysis of 5'-RACE-PCR fragments confirmed that in seven cases of them, there was a PDGFRA-FIP1L1 fusion. We also verified that two of nine patients with hyperposphorylated PDGFRA hold ETV6-PDGFRA and STRN-PDGFRA rearrangements. Importantly, nine cases also had significantly higher levels of PDGFRA mRNA expression when compared with healthy controls, and cases with no PDGFRA rearrangement. These findings highlight a robust correlation between hyperphosphorylation state of PDGFRA and aberrant PDGFRA gene fusions. This implicates phosflow as an efficient and reliable technique raising an intriguing possibility that it could replace other genomic and cDNA-amplification-based diagnostic approaches with limited effectiveness.
Collapse
Affiliation(s)
- Reza Ranjbaran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojdeh Abbasi
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.,Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Gholamreza Rafiei Dehbidi
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Noorossadat Seyyedi
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Behzad-Behbahani
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sedigheh Sharifzadeh
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
2
|
Yang YW, Marrufo A, Chase J, Woodard GA, Jablons DM, Lemjabbar-Alaoui H. Ponatinib is a potential therapeutic approach for malignant pleural mesothelioma. Exp Lung Res 2020; 47:9-25. [PMID: 33107354 DOI: 10.1080/01902148.2020.1836691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Malignant pleural mesothelioma (MPM) is a rare and deadly malignancy. Current MPM therapies remain inadequate, and outcomes are often disappointing. New meaningful therapeutic approaches are urgently needed. Accumulating evidence indicates that the cAbl pathway promotes various tumor-stimulating processes in MPM. In this study, we sought to determine ponatinib's potential utility, a clinically approved and potent cAbl inhibitor, in MPM treatment. MATERIAL AND METHODS Four MPM lines (MSTO211H, H28, H2452, H2052) were treated with ponatinib in vitro, and their growth was assessed. Scratch wound assay was used to investigate the ponatinib effect on cell migration. The expression levels of pAbl and its downstream effectors pCrkL, pAKT, and pSTAT5 were characterized. The in vivo ponatinib effect was evaluated in human MPM cells derived tumor model. RESULTS In all four MPM lines, significant expression levels of phosphorylated cAbl/Arg and pCrkl were observed. Differentially but strongly, ponatinib inhibited the in vitro cell growth and migration of all four MPM line. Western blot analysis showed that the activation of Abl signaling was blocked in the ponatinib-treated MMP lines. In keeping, the cellular levels of pAbl and its downstream effector pCrkL, pAKT, and pSTAT5 were markedly decrease following ponatinib treatment. Moreover, ponatinib treatment amplified the levels of γH2AX in cells denoting increased double-strand DNA breaks levels. Notably, ponatinib treatment reduced in vivo tumor growth and reduced pCrkl and pSTAT5 levels in tumor samples. CONCLUSION Ponatinib may offer a new therapeutic strategy for MPM patients based on cAbl signaling pathway inhibition.
Collapse
Affiliation(s)
- Yi-Wei Yang
- Department of Surgery, Thoracic Oncology Program, University of California, San Francisco, San Francisco, California, USA
| | - Angelica Marrufo
- Department of Surgery, Thoracic Oncology Program, University of California, San Francisco, San Francisco, California, USA
| | - Jillian Chase
- Department of Surgery, Thoracic Oncology Program, University of California, San Francisco, San Francisco, California, USA
| | - Gavitt A Woodard
- Department of Surgery, Thoracic Oncology Program, University of California, San Francisco, San Francisco, California, USA
| | - David M Jablons
- Department of Surgery, Thoracic Oncology Program, University of California, San Francisco, San Francisco, California, USA
| | - Hassan Lemjabbar-Alaoui
- Department of Surgery, Thoracic Oncology Program, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
3
|
Nath A, Wang J, Stephanie Huang R. Pharmacogenetics and Pharmacogenomics of Targeted Therapeutics in Chronic Myeloid Leukemia. Mol Diagn Ther 2018; 21:621-631. [PMID: 28698977 DOI: 10.1007/s40291-017-0292-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The advent of targeted therapeutics has greatly improved outcomes of chronic myeloid leukemia (CML) patients. Despite increased efficacy and better clinical responses over cytotoxic chemotherapies, many patients receiving targeted drugs exhibit a poor initial response, develop drug resistance, or undergo relapse after initial success. This inter-individual variation in response has heightened the interest in studying pharmacogenetics and pharmacogenomics (PGx) of cancer drugs. In this review, we discuss the influence of various germline and somatic factors on targeted drug response in CML. Specifically, we examine the role of genetic variants in drug metabolism genes, i.e. CYP3A family genes, and drug transporters, i.e. ABC and SLC family genes. Additionally, we focus on acquired somatic variations in BCR-ABL1, and the potential role played by additional downstream signaling pathways, in conferring resistance to targeted drugs in CML. This review highlights the importance of PGx of targeted therapeutics and its potential application to improving treatment decisions and patient outcomes.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cytochrome P-450 CYP3A/genetics
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Fusion Proteins, bcr-abl/genetics
- Glucuronosyltransferase/genetics
- Humans
- Inactivation, Metabolic/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Organic Cation Transporter 1/genetics
- Pharmacogenetics
- Protein Kinase Inhibitors/therapeutic use
Collapse
Affiliation(s)
- Aritro Nath
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jacqueline Wang
- Biological Sciences Collegiate Division, The University of Chicago, Chicago, IL, USA
| | - R Stephanie Huang
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
4
|
Hua JY, Zhou XH, Ouyang ST, Wu YB. [Effect of bortezomib in inducing apoptosis of imatinib-resistant K562 cells and the mechanism]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:1136-1139. [PMID: 28801299 PMCID: PMC6765724 DOI: 10.3969/j.issn.1673-4254.2017.08.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To investigate the effect of bortezomib in inducing apoptosis in imatinib-resistant K562 (K562R) cells and its possible mechanism. METHODS K562 cells were cultured in gradient concentrations of imatinib for several months to generate imatinib-resistant K562 cells. The viability of K562R cells treated with bortezomib was measured using CCK-8 cell proliferation assay, and the cell apoptosis was analyzed by flow cytometry with annexin V/PI dual staining. Western blotting was used to detect the protein expressions of Mcl-1,Bcl-2 and Bcr/Abl. RESULTS K562R cell line was successfully established, which showed 31.8 folds of imatinib resistance compared with the na?ve cells. Bortezomib treatment produced dose- and time-dependent inhibitory effect on the proliferation of both K562 cells and K562R cells and dose-dependently induced apoptosis in K562R cells. Combination of bortezomib with imatinib significantly enhanced the apoptosis of the cells. Western blotting showed that bortezomib treatment dose-dependently decreased the protein levels of both Mcl-1and Bcr/Abl in K562R cells without affecting bcl-2 protein expression. CONCLUSION Bortezomib can inhibit the proliferation of K562R cells and induce cell apoptosis possibly by down-regulating Mcl-1 and Bcr/Abl expression and enhancing Mcl-1 cleavage.
Collapse
Affiliation(s)
- Jia-Ye Hua
- Department of Hematology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China. E-mail:
| | | | | | | |
Collapse
|
5
|
Juen L, Brachet-Botineau M, Parmenon C, Bourgeais J, Hérault O, Gouilleux F, Viaud-Massuard MC, Prié G. New Inhibitor Targeting Signal Transducer and Activator of Transcription 5 (STAT5) Signaling in Myeloid Leukemias. J Med Chem 2017; 60:6119-6136. [PMID: 28654259 DOI: 10.1021/acs.jmedchem.7b00369] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Signal transducers and activators of transcription 5 (STAT5s) are crucial effectors of tyrosine kinase oncogenes in myeloid leukemias. Inhibition of STAT5 would contribute to reducing the survival of leukemic cells and also tackling their chemoresistance. In a first screening experiment, we identified hit 13 as able to inhibit STAT5 phosphorylation and leukemic cell growth. The synthesis of 18 analogues of 13 allowed us to identify one compound, 17f, as having the most potent antileukemic effect. 17f inhibited the growth of acute and chronic myeloid leukemia cells and the phosphorylation and transcriptional activity of STAT5. Importantly, 17f had minimal effects on bone marrow stromal cells that play vital functions in the microenvironment of hematopoietic and leukemic cells. We also demonstrated that 17f inhibits STAT5 but not STAT3, AKT, or Erk1/2 phosphorylation. These results suggest that 17f might be a new lead molecule targeting STAT5 signaling in myeloid leukemias.
Collapse
Affiliation(s)
- Ludovic Juen
- Equipe IMT "Innovation Moléculaire et Thérapeutique", GICC UMR 7292 CNRS, Université de Tours, Labex SYNORG, Faculté de Pharmacie, 31 avenue Monge, 37200 Tours, France
| | - Marie Brachet-Botineau
- Equipe LNOx "Niche leucémique & métabolisme oxidatif", GICC UMR 7292 CNRS, Université de Tours, Faculté de Médecine, Bâtiment Dutrochet, 10bis boulevard Tonnellé, 37032 Tours, France.,CHRU de Tours, Service d'Hématologie Biologique, 2 boulevard Tonnellé, 37044 Tours, France
| | - Cécile Parmenon
- Equipe IMT "Innovation Moléculaire et Thérapeutique", GICC UMR 7292 CNRS, Université de Tours, Labex SYNORG, Faculté de Pharmacie, 31 avenue Monge, 37200 Tours, France
| | - Jérôme Bourgeais
- Equipe LNOx "Niche leucémique & métabolisme oxidatif", GICC UMR 7292 CNRS, Université de Tours, Faculté de Médecine, Bâtiment Dutrochet, 10bis boulevard Tonnellé, 37032 Tours, France.,CHRU de Tours, Service d'Hématologie Biologique, 2 boulevard Tonnellé, 37044 Tours, France
| | - Olivier Hérault
- Equipe LNOx "Niche leucémique & métabolisme oxidatif", GICC UMR 7292 CNRS, Université de Tours, Faculté de Médecine, Bâtiment Dutrochet, 10bis boulevard Tonnellé, 37032 Tours, France.,CHRU de Tours, Service d'Hématologie Biologique, 2 boulevard Tonnellé, 37044 Tours, France
| | - Fabrice Gouilleux
- Equipe LNOx "Niche leucémique & métabolisme oxidatif", GICC UMR 7292 CNRS, Université de Tours, Faculté de Médecine, Bâtiment Dutrochet, 10bis boulevard Tonnellé, 37032 Tours, France
| | - Marie-Claude Viaud-Massuard
- Equipe IMT "Innovation Moléculaire et Thérapeutique", GICC UMR 7292 CNRS, Université de Tours, Labex SYNORG, Faculté de Pharmacie, 31 avenue Monge, 37200 Tours, France
| | - Gildas Prié
- Equipe IMT "Innovation Moléculaire et Thérapeutique", GICC UMR 7292 CNRS, Université de Tours, Labex SYNORG, Faculté de Pharmacie, 31 avenue Monge, 37200 Tours, France
| |
Collapse
|
6
|
Brumbaugh K, Liao WC, Houchins JP, Cooper J, Stoesz S. Phosphosite-Specific Antibodies: A Brief Update on Generation and Applications. Methods Mol Biol 2017; 1554:1-40. [PMID: 28185181 DOI: 10.1007/978-1-4939-6759-9_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Phosphate addition is a posttranslational modification of proteins, and this modification can affect the activity and other properties of intracellular proteins. Different animal species can be used to generate phosphosite-specific antibodies as either polyclonals or monoclonals, and each approach offers its own benefits and disadvantages. The validation of phosphosite-specific antibodies requires multiple techniques and tactics to demonstrate their specificity. These antibodies can be used in arrays, flow cytometry, and imaging platforms. The specificity of phosphosite-specific antibodies is vital for their use in proteomics and profiling of disease.
Collapse
Affiliation(s)
- Kathy Brumbaugh
- Bio-Techne, Inc., 614 McKinley Place NE, Minneapolis, MN, 55413, USA.
| | - Wen-Chie Liao
- Bio-Techne, Inc., 614 McKinley Place NE, Minneapolis, MN, 55413, USA
| | - J P Houchins
- Bio-Techne, Inc., 614 McKinley Place NE, Minneapolis, MN, 55413, USA
| | - Jeff Cooper
- Bio-Techne, Inc., 614 McKinley Place NE, Minneapolis, MN, 55413, USA
| | - Steve Stoesz
- Bio-Techne, Inc., 614 McKinley Place NE, Minneapolis, MN, 55413, USA
| |
Collapse
|
7
|
Arock M, Mahon FX, Valent P. Characterization and targeting of neoplastic stem cells in Ph + chronic myeloid leukemia. Int J Hematol Oncol 2015. [DOI: 10.2217/ijh.15.16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm characterized by the presence of an oncogenic fusion gene, BCR–ABL1. This fusion gene produces a cytoplasmic protein with tyrosine kinase activity that acts as a main driver of oncogenesis and abnormal proliferation of myeloid cells in CML. Targeted therapy with BCR–ABL1 tyrosine kinase inhibitors (TKIs) such as imatinib is followed by long-term responses in most patients. However, despite continuous treatment, relapses occur, suggesting the presence of TKI-resistant neoplastic stem cells in these patients. Here, we discuss potential mechanisms and signaling molecules involved in the prosurvival and self-renewal capacity of CML neoplastic stem cells as well as antigens expressed by these cells. Several of these signaling molecules and cell surface antigens may serve as potential targets of therapy and their use may overcome TKI resistance in CML in the future.
Collapse
Affiliation(s)
- Michel Arock
- Molecular & Cellular Oncology, LBPA CNRS UMR8113, Ecole Normale Supérieure de Cachan, Cachan, France
- Laboratory of Hematology, Pitié-Salpêtrière Hospital, Paris, France
| | - François-Xavier Mahon
- Laboratory of Hematology, CHU de Bordeaux, Bordeaux, France
- Laboratoire Hématopoïèse Leucémique et Cible Thérapeutique INSERM U1035, Université de Bordeaux, Bordeaux, France
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
8
|
Redaelli S, Perini P, Ceccon M, Piazza R, Rigolio R, Mauri M, Boschelli F, Giannoudis A, Gambacorti-Passerini C. In vitro and in vivo identification of ABCB1 as an efflux transporter of bosutinib. J Hematol Oncol 2015; 8:81. [PMID: 26149173 PMCID: PMC4491863 DOI: 10.1186/s13045-015-0179-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 06/24/2015] [Indexed: 11/13/2022] Open
Abstract
Background Bosutinib is a recently approved ABL inhibitor. In spite of the well-documented effectiveness of BCR-ABL inhibitors in treating chronic myeloid leukemia, development of resistance is a continuous clinical challenge. Transporters that facilitate drug uptake and efflux have been proposed as one potential source of resistance to tyrosine kinase inhibitor treatment. Our aim was to determine which carriers are responsible for bosutinib transport. Methods K562S cells overexpressing the drug transporters ABCB1, ABCG2, and SLC22A1 were generated, characterized and used in proliferation assay and intracellular uptake and retention assay (IUR). In vivo experiments were performed in nude mice injected with K562S, K562DOX cells (overexpressing ABCB1), and K562DOX silenced for ABCB1 (K562DOX/sh P-GP). Results The IUR assay using C-14 bosutinib showed that only ABCB1 was responsible for active bosutinib transport. K562DOX cells showed the lowest intracellular level of bosutinib, while K562DOX cells treated with the ABCB1 inhibitor verapamil showed intracellular bosutinib levels comparable with parental K562S. Proliferation assays demonstrated that K562DOX are resistant to bosutinib treatment while verapamil is able to restore the sensitivity to the drug. Nude mice injected with K562DOX and treated with bosutinib showed very limited response and quickly relapsed after stopping treatment while K562S as well as K562DOX/sh P-GP remained tumor-free. Conclusions Our data suggest that the analysis of ABCB1 expression levels might help determine treatment options for patients exhibiting resistance to bosutinib. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0179-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sara Redaelli
- Department of Health sciences, University of Milano-Bicocca, S.Gerardo Hospital, Monza, Italy.
| | - Pietro Perini
- Department of Health sciences, University of Milano-Bicocca, S.Gerardo Hospital, Monza, Italy.
| | - Monica Ceccon
- Department of Health sciences, University of Milano-Bicocca, S.Gerardo Hospital, Monza, Italy.
| | - Rocco Piazza
- Department of Health sciences, University of Milano-Bicocca, S.Gerardo Hospital, Monza, Italy.
| | - Roberta Rigolio
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, Monza, Italy.
| | - Mario Mauri
- Department of Health sciences, University of Milano-Bicocca, S.Gerardo Hospital, Monza, Italy.
| | - Frank Boschelli
- Department of Oncology, Pfizer Research, Pearl River, New City, NY, USA.
| | - Athina Giannoudis
- Institute of Translational Medicine, Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK.
| | | |
Collapse
|
9
|
Niu S, Wang Z, Ge D, Zhang G, Li Y. Prediction of functional phosphorylation sites by incorporating evolutionary information. Protein Cell 2012; 3:675-90. [PMID: 22802047 DOI: 10.1007/s13238-012-2048-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 06/27/2012] [Indexed: 01/05/2023] Open
Abstract
Protein phosphorylation is a ubiquitous protein post-translational modification, which plays an important role in cellular signaling systems underlying various physiological and pathological processes. Current in silico methods mainly focused on the prediction of phosphorylation sites, but rare methods considered whether a phosphorylation site is functional or not. Since functional phosphorylation sites are more valuable for further experimental research and a proportion of phosphorylation sites have no direct functional effects, the prediction of functional phosphorylation sites is quite necessary for this research area. Previous studies have shown that functional phosphorylation sites are more conserved than non-functional phosphorylation sites in evolution. Thus, in our method, we developed a web server by integrating existing phosphorylation site prediction methods, as well as both absolute and relative evolutionary conservation scores to predict the most likely functional phosphorylation sites. Using our method, we predicted the most likely functional sites of the human, rat and mouse proteomes and built a database for the predicted sites. By the analysis of overall prediction results, we demonstrated that protein phosphorylation plays an important role in all the enriched KEGG pathways. By the analysis of protein-specific prediction results, we demonstrated the usefulness of our method for individual protein studies. Our method would help to characterize the most likely functional phosphorylation sites for further studies in this research area.
Collapse
Affiliation(s)
- Shen Niu
- Key Laboratory of Systems Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | | | | | | | | |
Collapse
|
10
|
Brumbaugh K, Johnson W, Liao WC, Lin MS, Houchins JP, Cooper J, Stoesz S, Campos-Gonzalez R. Overview of the generation, validation, and application of phosphosite-specific antibodies. Methods Mol Biol 2011; 717:3-43. [PMID: 21370022 DOI: 10.1007/978-1-61779-024-9_1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein phosphorylation is a universal key posttranslational modification that affects the activity and other properties of intracellular proteins. Phosphosite-specific antibodies can be produced as polyclonals or monoclonals in different animal species, and each approach offers its own benefits and disadvantages. The validation of phosphosite-specific antibodies requires multiple techniques and tactics to demonstrate their specificity. These antibodies can be used in arrays, flow cytometry, and imaging platforms. The specificity of phosphosite-specific antibodies is key for their use in proteomics and profiling of disease.
Collapse
|
11
|
Sylvester JE, Kron SJ. A bead-based activity screen for small-molecule inhibitors of signal transduction in chronic myelogenous leukemia cells. Mol Cancer Ther 2010; 9:1469-81. [PMID: 20423990 PMCID: PMC2868067 DOI: 10.1158/1535-7163.mct-10-0157] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chronic myelogenous leukemia is characterized by the presence of the chimeric BCR-ABL gene, which is expressed as the constitutively active Bcr-Abl kinase. Although kinase activity is directly responsible for the clinical phenotype, current diagnostic and prognostic methods focus on a genetic classification system in which molecularly distinct subcategories are used to predict patient responses to small-molecule inhibitors of the Bcr-Abl kinase. Point mutations in the kinase domain are a central factor regulating inhibitor resistance; however, compensatory signaling caused by the activation of unrelated kinases can influence inhibitor efficacy. Kinase activity profiling can be used as a complementary approach to genetic screening and allows direct screening of small-molecule inhibitors. We developed a quantitative assay to monitor tyrosine kinase activities and inhibitor sensitivities in a model of chronic myelogenous leukemia using peptide reporters covalently immobilized on Luminex beads. Kinase activity is quantified by nonlinear regression from well-specific internal standard curves. Using optimized synthetic substrates and peptides derived from native substrates as probes, we measured kinase inhibition in cell lysates by the signal transduction inhibitors imatinib and dasatinib. Taking advantage of a convenient 96-well plate format, this assay also allows a straightforward and quantitative analysis of the differential effects of ATP and inhibitors on kinase activity. This method for analyzing a focused signaling network benefits from rigorous statistical analysis and short processing times, thereby offering a powerful tool for drug discovery and clinical testing.
Collapse
MESH Headings
- Antineoplastic Agents/analysis
- Antineoplastic Agents/isolation & purification
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Calibration
- Dose-Response Relationship, Drug
- Drug Screening Assays, Antitumor/methods
- Drug Screening Assays, Antitumor/standards
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/metabolism
- Humans
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Microspheres
- Models, Biological
- Phosphorylation/drug effects
- Protein Kinase Inhibitors/analysis
- Protein Kinase Inhibitors/isolation & purification
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Pyridines/pharmacology
- Pyridines/therapeutic use
- Pyridones/pharmacology
- Pyridones/therapeutic use
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
- Signal Transduction/drug effects
- Small Molecule Libraries/analysis
- Time Factors
Collapse
Affiliation(s)
- Juliesta E. Sylvester
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois 60637
- Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois 60637
| | - Stephen J. Kron
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, Illinois 60637
- Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
12
|
Khorashad JS, Wagner S, Greener L, Marin D, Reid A, Milojkovic D, Patel H, Willimott S, Rezvani K, Gerrard G, Loaiza S, Davis J, Goldman J, Melo J, Apperley J, Foroni L. The level of BCR-ABL1 kinase activity before treatment does not identify chronic myeloid leukemia patients who fail to achieve a complete cytogenetic response on imatinib. Haematologica 2009; 94:861-4. [PMID: 19377081 DOI: 10.3324/haematol.2008.003715] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Imatinib is currently the first line therapy for newly diagnosed patients with chronic myeloid leukemia. However, 20-25% of patients do not achieve durable complete cytogenetic responses. The mechanism underlying this primary resistance is unknown, but variations in BCR-ABL1 kinase activity may play a role and can be investigated by measuring the autophosphorylation levels of BCR-ABL1 or of a surrogate target such as Crkl. In this study we used flow cytometry to investigate the in vitro inhibition of Crkl phosphorylation by imatinib in CD34(+) cells in diagnostic samples from two groups of patients distinguished by their cytogenetic response. No difference in inhibition of Crkl phosphorylation was observed in the two groups. The observation that increasing the dose of imatinib in vivo did not increase the level of cytogenetic response in some non-responders suggests that in at least a proportion of patients imatinib resistance may be due to activation of BCR-ABL1-independent pathway.
Collapse
|
13
|
Rainey MD, Charlton ME, Stanton RV, Kastan MB. Transient inhibition of ATM kinase is sufficient to enhance cellular sensitivity to ionizing radiation. Cancer Res 2008; 68:7466-74. [PMID: 18794134 PMCID: PMC2559948 DOI: 10.1158/0008-5472.can-08-0763] [Citation(s) in RCA: 186] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In response to DNA damage, the ATM protein kinase activates signal transduction pathways essential for coordinating cell cycle progression with DNA repair. In the human disease ataxia-telangiectasia, mutation of the ATM gene results in multiple cellular defects, including enhanced sensitivity to ionizing radiation (IR). This phenotype highlights ATM as a potential target for novel inhibitors that could be used to enhance tumor cell sensitivity to radiotherapy. A targeted compound library was screened for potential inhibitors of the ATM kinase, and CP466722 was identified. The compound is nontoxic and does not inhibit phosphatidylinositol 3-kinase (PI3K) or PI3K-like protein kinase family members in cells. CP466722 inhibited cellular ATM-dependent phosphorylation events and disruption of ATM function resulted in characteristic cell cycle checkpoint defects. Inhibition of cellular ATM kinase activity was rapidly and completely reversed by removing CP466722. Interestingly, clonogenic survival assays showed that transient inhibition of ATM is sufficient to sensitize cells to IR and suggests that therapeutic radiosensitization may only require ATM inhibition for short periods of time. The ability of CP466722 to rapidly and reversibly regulate ATM activity provides a new tool to ask questions about ATM function that could not easily be addressed using genetic models or RNA interference technologies.
Collapse
Affiliation(s)
- Michael D. Rainey
- Department of Oncology, St. Jude Children’s Research Hospital, 332 North Lauderdale Street, Memphis, TN, 38105. USA
| | - Maura E. Charlton
- Pfizer Research Technology Center, Pfizer Global Research and Development, 620 Memorial Drive, Cambridge, MA, 02139. USA
| | - Robert V. Stanton
- Pfizer Research Technology Center, Pfizer Global Research and Development, 620 Memorial Drive, Cambridge, MA, 02139. USA
| | - Michael B. Kastan
- Department of Oncology, St. Jude Children’s Research Hospital, 332 North Lauderdale Street, Memphis, TN, 38105. USA
| |
Collapse
|