1
|
Hou Z, Liu H. Mapping the Protein Kinome: Current Strategy and Future Direction. Cells 2023; 12:cells12060925. [PMID: 36980266 PMCID: PMC10047437 DOI: 10.3390/cells12060925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/23/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
The kinome includes over 500 different protein kinases, which form an integrated kinase network that regulates cellular phosphorylation signals. The kinome plays a central role in almost every cellular process and has strong linkages with many diseases. Thus, the evaluation of the cellular kinome in the physiological environment is essential to understand biological processes, disease development, and to target therapy. Currently, a number of strategies for kinome analysis have been developed, which are based on monitoring the phosphorylation of kinases or substrates. They have enabled researchers to tackle increasingly complex biological problems and pathological processes, and have promoted the development of kinase inhibitors. Additionally, with the increasing interest in how kinases participate in biological processes at spatial scales, it has become urgent to develop tools to estimate spatial kinome activity. With multidisciplinary efforts, a growing number of novel approaches have the potential to be applied to spatial kinome analysis. In this paper, we review the widely used methods used for kinome analysis and the challenges encountered in their applications. Meanwhile, potential approaches that may be of benefit to spatial kinome study are explored.
Collapse
Affiliation(s)
- Zhanwu Hou
- Center for Mitochondrial Biology and Medicine, Douglas C. Wallace Institute for Mitochondrial and Epigenetic Information Sciences, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Huadong Liu
- School of Health and Life Science, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| |
Collapse
|
2
|
Miao W, Yin J, Porter DF, Jiang X, Khavari PA, Wang Y. Targeted Proteomic Approaches for Proteome-Wide Characterizations of the AMP-Binding Capacities of Kinases. J Proteome Res 2022; 21:2063-2070. [PMID: 35820187 PMCID: PMC9357193 DOI: 10.1021/acs.jproteome.2c00225] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Kinases play important roles in cell signaling, and adenosine monophosphate (AMP) is known to modulate cellular energy homeostasis through AMP-activated protein kinase (AMPK). Here, we explored novel AMP-binding kinases by employing a desthiobiotin-conjugated AMP acyl-phosphate probe to enrich efficiently AMP-binding proteins. Together with a parallel-reaction monitoring-based targeted proteomic approach, we uncovered 195 candidate AMP-binding kinases. We also enriched desthiobiotin-labeled peptides from adenine nucleotide-binding sites of kinases and analyzed them using LC-MS/MS in the multiple-reaction monitoring mode, which resulted in the identification of 44 peptides derived from 43 kinases displaying comparable or better binding affinities toward AMP relative to adenosine triphosphate (ATP). Moreover, our proteomic data revealed a potential involvement of AMP in the MAPK pathway through binding directly to the relevant kinases, especially MEK2 and MEK3. Together, we revealed the AMP-binding capacities of a large number of kinases, and our work built a strong foundation for understanding how AMP functions as a second messenger to modulate cell signaling.
Collapse
Affiliation(s)
- Weili Miao
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, California 94305, United States
| | | | - Douglas F Porter
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, California 94305, United States
| | | | - Paul A Khavari
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, California 94305, United States
| | | |
Collapse
|
3
|
East MP, Johnson GL. Adaptive chromatin remodeling and transcriptional changes of the functional kinome in tumor cells in response to targeted kinase inhibition. J Biol Chem 2021; 298:101525. [PMID: 34958800 PMCID: PMC8888345 DOI: 10.1016/j.jbc.2021.101525] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/06/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022] Open
Abstract
Pharmacological inhibition of protein kinases induces adaptive reprogramming of tumor cell regulatory networks by altering expression of genes that regulate signaling, including protein kinases. Adaptive responses are dependent on transcriptional changes resulting from remodeling of enhancer and promoter landscapes. Enhancer and promoter remodeling in response to targeted kinase inhibition is controlled by changes in open chromatin state and by activity of specific transcription factors, such as c-MYC. This review focuses on the dynamic plasticity of protein kinase expression of the tumor cell kinome and the resulting adaptive resistance to targeted kinase inhibition. Plasticity of the functional kinome has been shown in patient window trials where triple-negative and human epidermal growth factor receptor 2–positive breast cancer patient tumors were characterized by RNAseq after biopsies before and after 1 week of therapy. The expressed kinome changed dramatically during drug treatment, and these changes in kinase expression were shown in cell lines and xenografts in mice to be correlated with adaptive tumor cell drug resistance. The dynamic transcriptional nature of the kinome also differs for inhibitors targeting different kinase signaling pathways (e.g., BRAF-MEK-ERK versus PI3K-AKT) that are commonly activated in cancers. Heterogeneity arising from differences in gene regulation and mutations represents a challenge to therapeutic durability and prevention of clinical drug resistance with drug-tolerant tumor cell populations developing and persisting through treatment. We conclude that understanding the heterogeneity of kinase expression at baseline and in response to therapy is imperative for development of combinations and timing intervals of therapies making interventions durable.
Collapse
Affiliation(s)
- Michael P East
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Gary L Johnson
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.
| |
Collapse
|
4
|
Miao W, Yang YY, Wang Y. Quantitative Proteomic Analysis Revealed Broad Roles of N6-Methyladenosine in Heat Shock Response. J Proteome Res 2021; 20:3611-3620. [PMID: 34043365 DOI: 10.1021/acs.jproteome.1c00191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As optimum temperature is essential for all living organisms, heat shock represents a challenging problem for their survival. Therefore, cellular response to heat shock is among the most extensively investigated stress response pathways; however, how the human proteome responds to heat shock has not been comprehensively investigated. In this study, we employed stable isotope labeling by amino acids in cell culture (SILAC), together with liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis, to fulfill an in-depth analysis of the alterations in the human proteome in M14 human melanoma cells in response to heat shock stress. We found that, after heat shock, 284 and 278 out of the 4319 quantified proteins were with substantially diminished and elevated expressions, respectively. We also examined the alterations in human kinome after heat shock by using our recently developed targeted proteomic method relying on parallel-reaction monitoring. Our results showed that the expression levels of 11 and 22 kinase proteins were increased and decreased, respectively, by at least 1.5-fold upon heat shock. By interrogating publicly available RNA-seq and m6A sequencing data, we observed that the elevated expression of more than 30 proteins, including CHEK1 and CCND3 kinases, could occur via an m6A-mediated mechanism. Furthermore, our results from single-base elongation and ligation-based quantitative polymerase chain reaction (qPCR) amplification (SELECT) and luciferase reporter assays revealed that heat shock gave rise to elevated m6A levels at A280 and A286 sites in the 5'-untranslated region of HSPH1 mRNA, thereby leading to increased translation of HSPH1 protein. Together, our discovery and targeted proteomic methods revealed the reprogramming of human proteome and kinome upon heat shock stress and provided insights into cellular responses toward heat shock stress.
Collapse
Affiliation(s)
- Weili Miao
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Yen-Yu Yang
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| |
Collapse
|
5
|
Miao W, Bade D, Wang Y. Targeted Proteomic Analysis Revealed Kinome Reprogramming during Acquisition of Radioresistance in Breast Cancer Cells. J Proteome Res 2021; 20:2830-2838. [PMID: 33739118 DOI: 10.1021/acs.jproteome.1c00075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Radiotherapy constitutes a major therapeutic modality for early management of breast cancer. Despite the high efficacy in treating breast cancer (BC), radiation resistance and tumor recurrence are major hurdles in breast cancer radiotherapy. Herein, stable isotope labeling by amino acids in cell culture (SILAC) was employed, along with the parallel-reaction monitoring (PRM)-based targeted quantitative proteomic method, to examine the differences in kinase protein expression in MCF-7 and MDA-MB-231 breast cancer cells and their corresponding radioresistant C6 and C5 clones. We quantified the relative protein expression levels of 300 and 281 kinases in C5/MDA-MB-231 and C6/MCF-7 pairs of breast cancer cells, respectively. We also showed that TAF9, which was one of the differentially expressed kinases, enhances radiation resistance in breast cancer cells. Moreover, a correlation analysis of gene expression suggested TAF9's role in upregulating the expression of genes involved with radioresistance. Overall, our study uncovered a large number of differentially expressed kinases accompanied with the acquisition of radioresistance and revealed a role of TAF9 in promoting radioresistance in breast cancer.
Collapse
|
6
|
Balakirouchenane D, Guégan S, Csajka C, Jouinot A, Heidelberger V, Puszkiel A, Zehou O, Khoudour N, Courlet P, Kramkimel N, Lheure C, Franck N, Huillard O, Arrondeau J, Vidal M, Goldwasser F, Maubec E, Dupin N, Aractingi S, Guidi M, Blanchet B. Population Pharmacokinetics/Pharmacodynamics of Dabrafenib Plus Trametinib in Patients with BRAF-Mutated Metastatic Melanoma. Cancers (Basel) 2020; 12:cancers12040931. [PMID: 32283865 PMCID: PMC7226106 DOI: 10.3390/cancers12040931] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/30/2020] [Accepted: 04/05/2020] [Indexed: 12/15/2022] Open
Abstract
Patients treated with dabrafenib/trametinib (DAB/TRA) exhibit a large interindividual variability in clinical outcomes. The aims of this study were to characterize the pharmacokinetics of DAB, hydroxy-dabrafenib (OHD), and TRA in BRAF-mutated patients and to investigate the exposure–response relationship for toxicity and efficacy in metastatic melanoma (MM) patients. Univariate Fisher and Wilcoxon models including drug systemic exposure (area under the plasma concentration curve, AUC) were used to identify prognostic factors for the onset of dose-limiting toxicities (DLT), and Cox models for overall (OS) and progression-free survival (PFS). Seventy-three BRAF-mutated patients were included in pharmacokinetic (n = 424, NONMEM) and 52 in pharmacokinetic/pharmacodynamic analyses. Age and sex were identified as determinants of DAB and OHD clearances (p < 0.01). MM patients experiencing DLT were overexposed to DAB compared to patients without DLT (AUC: 9624 vs. 7485 ng∙h/mL, respectively, p < 0.01). Eastern Cooperative Oncology Group Performance Status (ECOG PS) ≥ 2 and plasma ratio AUCOHD/AUCDAB ≥ 1 were independently associated with shorter OS (HR: 6.58 (1.29–33.56); p = 0.023 and 10.61 (2.34–48.15), p = 0.022, respectively). A number of metastatic sites ≥3 and cerebral metastases were associated with shorter PFS (HR = 3.25 (1.11–9.50); p = 0.032 and HR = 1.23 (1.35–10.39), p = 0.011; respectively). TRA plasma exposure was neither associated with toxicity nor efficacy. Our results suggest that early drug monitoring could be helpful to prevent the onset of DLT in MM patients, especially in fragile patients such as the elderly. Regarding efficacy, the clinical benefit to monitor plasma ratio AUCOHD/AUCDAB deserves more investigation in a larger cohort of MM patients.
Collapse
Affiliation(s)
- David Balakirouchenane
- Department of Pharmacokinetics and Pharmacochemistry, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France; (D.B.); (A.P.); (N.K.); (M.V.)
- UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, University of Paris, PRES Sorbonne Paris Cité, CARPEM, 75006 Paris, France
| | - Sarah Guégan
- Department of Dermatology, Cochin Hospital AP-HP, 75014 Paris, France; (S.G.); (N.K.); (C.L.); (N.F.); (N.D.); (S.A.)
- Cochin Institute, INSERM U1016, University of Paris, 75014 Paris, France;
| | - Chantal Csajka
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (C.C.); (M.G.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Anne Jouinot
- Cochin Institute, INSERM U1016, University of Paris, 75014 Paris, France;
| | - Valentine Heidelberger
- Department of Dermatology, Avicenne Hospital AP-HP, 93000 Bobigny, France; (V.H.); (E.M.)
| | - Alicja Puszkiel
- Department of Pharmacokinetics and Pharmacochemistry, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France; (D.B.); (A.P.); (N.K.); (M.V.)
| | - Ouidad Zehou
- Department of Dermatology, Henri Mondor Hospital AP-HP, 94010 Créteil, France;
| | - Nihel Khoudour
- Department of Pharmacokinetics and Pharmacochemistry, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France; (D.B.); (A.P.); (N.K.); (M.V.)
| | - Perrine Courlet
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland;
| | - Nora Kramkimel
- Department of Dermatology, Cochin Hospital AP-HP, 75014 Paris, France; (S.G.); (N.K.); (C.L.); (N.F.); (N.D.); (S.A.)
| | - Coralie Lheure
- Department of Dermatology, Cochin Hospital AP-HP, 75014 Paris, France; (S.G.); (N.K.); (C.L.); (N.F.); (N.D.); (S.A.)
| | - Nathalie Franck
- Department of Dermatology, Cochin Hospital AP-HP, 75014 Paris, France; (S.G.); (N.K.); (C.L.); (N.F.); (N.D.); (S.A.)
| | - Olivier Huillard
- Department of Medical Oncology, Cochin Hospital AP-HP, 75014 Paris, France; (O.H.); (J.A.); (F.G.)
| | - Jennifer Arrondeau
- Department of Medical Oncology, Cochin Hospital AP-HP, 75014 Paris, France; (O.H.); (J.A.); (F.G.)
| | - Michel Vidal
- Department of Pharmacokinetics and Pharmacochemistry, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France; (D.B.); (A.P.); (N.K.); (M.V.)
- UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, University of Paris, PRES Sorbonne Paris Cité, CARPEM, 75006 Paris, France
| | - Francois Goldwasser
- Department of Medical Oncology, Cochin Hospital AP-HP, 75014 Paris, France; (O.H.); (J.A.); (F.G.)
| | - Eve Maubec
- Department of Dermatology, Avicenne Hospital AP-HP, 93000 Bobigny, France; (V.H.); (E.M.)
| | - Nicolas Dupin
- Department of Dermatology, Cochin Hospital AP-HP, 75014 Paris, France; (S.G.); (N.K.); (C.L.); (N.F.); (N.D.); (S.A.)
- Cochin Institute, INSERM U1016, University of Paris, 75014 Paris, France;
| | - Selim Aractingi
- Department of Dermatology, Cochin Hospital AP-HP, 75014 Paris, France; (S.G.); (N.K.); (C.L.); (N.F.); (N.D.); (S.A.)
- Cochin Institute, INSERM U1016, University of Paris, 75014 Paris, France;
| | - Monia Guidi
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (C.C.); (M.G.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland;
| | - Benoit Blanchet
- Department of Pharmacokinetics and Pharmacochemistry, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France; (D.B.); (A.P.); (N.K.); (M.V.)
- UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, University of Paris, PRES Sorbonne Paris Cité, CARPEM, 75006 Paris, France
- Correspondence: ; Tel.: +331-5841-2313; Fax: +331-5841-2315
| |
Collapse
|
7
|
A Targeted Quantitative Proteomic Method Revealed a Substantial Reprogramming of Kinome during Melanoma Metastasis. Sci Rep 2020; 10:2485. [PMID: 32051510 PMCID: PMC7015909 DOI: 10.1038/s41598-020-59572-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/31/2020] [Indexed: 12/15/2022] Open
Abstract
Kinases are involved in numerous critical cell signaling processes, and dysregulation in kinase signaling is implicated in many types of human cancers. In this study, we applied a parallel-reaction monitoring (PRM)-based targeted proteomic method to assess kinome reprogramming during melanoma metastasis in three pairs of matched primary/metastatic human melanoma cell lines. Around 300 kinases were detected in each pair of cell lines, and the results showed that Janus kinase 3 (JAK3) was with reduced expression in the metastatic lines of all three pairs of melanoma cells. Interrogation of The Cancer Genome Atlas (TCGA) data showed that reduced expression of JAK3 is correlated with poorer prognosis in melanoma patients. Additionally, metastatic human melanoma cells/tissues exhibited diminished levels of JAK3 mRNA relative to primary melanoma cells/tissues. Moreover, JAK3 suppresses the migration and invasion of cultured melanoma cells by modulating the activities of matrix metalloproteinases 2 and 9 (MMP-2 and MMP-9). In summary, our targeted kinome profiling method provided by far the most comprehensive dataset for kinome reprogramming associated with melanoma progression, which builds a solid foundation for examining the functions of other kinases in melanoma metastasis. Moreover, our results reveal a role of JAK3 as a potential suppressor for melanoma metastasis.
Collapse
|
8
|
Liu X, Miao W, Huang M, Li L, Dai X, Wang Y. Elevated Hexokinase II Expression Confers Acquired Resistance to 4-Hydroxytamoxifen in Breast Cancer Cells. Mol Cell Proteomics 2019; 18:2273-2284. [PMID: 31519767 PMCID: PMC6823848 DOI: 10.1074/mcp.ra119.001576] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/26/2019] [Indexed: 12/11/2022] Open
Abstract
Tamoxifen has been clinically used in treating estrogen receptor (ER)-positive breast cancer for over 30 years. The most challenging aspect associated with tamoxifen therapy is the development of resistance in initially responsive breast tumors. We applied a parallel-reaction monitoring (PRM)-based quantitative proteomic method to examine the differential expression of kinase proteins in MCF-7 and the isogenic tamoxifen-resistant (TamR) cells. We were able to quantify the relative protein expression levels of 315 kinases, among which hexokinase 2 (HK2) and mTOR were up-regulated in TamR MCF-7 cells. We also observed that the TamR MCF-7 cells exhibited elevated rate of glycolysis than the parental MCF-7 cells. In addition, we found that phosphorylation of S6K - a target of mTOR - was much lower in TamR MCF-7 cells, and this phosphorylation level could be restored upon genetic depletion or pharmacological inhibition of HK2. Reciprocally, the level of S6K phosphorylation was diminished upon overexpression of HK2 in MCF-7 cells. Moreover, we observed that HK2 interacts with mTOR, and this interaction inhibits mTOR activity. Lower mTOR activity led to augmented autophagy, which conferred resistance of MCF-7 cells toward tamoxifen. Together, our study demonstrates that elevated expression of HK2 promotes autophagy through inhibiting the mTOR-S6K signaling pathway and results in resistance of MCF-7 breast cancer cells toward tamoxifen; thus, our results uncovered, for the first time, HK2 as a potential therapeutic target for overcoming tamoxifen resistance.
Collapse
Affiliation(s)
- Xiaochuan Liu
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| | - Weili Miao
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| | - Ming Huang
- Environmental Toxicology Graduate Program, University of California, Riverside, Riverside, CA 92521
| | - Lin Li
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| | - Xiaoxia Dai
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, Riverside, CA 92521; Environmental Toxicology Graduate Program, University of California, Riverside, Riverside, CA 92521.
| |
Collapse
|
9
|
Miao W, Li L, Wang Y. High-Throughput Targeted Quantitative Analysis of the Interaction between HSP90 and Kinases. Anal Chem 2019; 91:11507-11509. [PMID: 31476117 DOI: 10.1021/acs.analchem.9b03320] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Kinases, which function in numerous cell signaling processes, are among the best characterized groups of client proteins for the 90-kDa heat shock protein (HSP90), a molecular chaperone that suppresses the aggregation and maintains the proper folding of its substrate proteins (i.e., clients). No high-throughput proteomic method, however, has been developed for the characterizations of the interactions between HSP90 and the human kinome. Herein, by employing a parallel-reaction monitoring (PRM)-based targeted proteomic method, we found that 99 out of the 249 detected kinase proteins display diminished expression in cultured human cells upon treatment with ganetespib, a small-molecule HSP90 inhibitor. PRM analysis of kinase proteins in the affinity pull-down samples showed that 86 out of the 120 detected kinases are enriched from the CRISPR-engineered cells where a tandem affinity tag was conjugated with the C-terminus of endogenous HSP90β protein over the parental cells. Together, our results from the two complementary quantitative proteomic experiments offer systematic characterizations about the HSP90-kinase interactions at the entire proteome scale and reveal extensive interactions between HSP90 and kinase proteins in human cells.
Collapse
Affiliation(s)
- Weili Miao
- Department of Chemistry , University of California Riverside , Riverside , California 92521-0403 , United States
| | - Lin Li
- Department of Chemistry , University of California Riverside , Riverside , California 92521-0403 , United States
| | - Yinsheng Wang
- Department of Chemistry , University of California Riverside , Riverside , California 92521-0403 , United States
| |
Collapse
|
10
|
Miao W, Yuan J, Li L, Wang Y. Parallel-Reaction-Monitoring-Based Proteome-Wide Profiling of Differential Kinase Protein Expression during Prostate Cancer Metastasis in Vitro. Anal Chem 2019; 91:9893-9900. [PMID: 31241916 DOI: 10.1021/acs.analchem.9b01561] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Prostate cancer is the most common type of cancer in men, and kinases are heavily pursued as drug targets for anticancer therapy. In this study, we applied our recently reported parallel-reaction-monitoring (PRM)-based targeted proteomic method to examine the reprogramming of the human kinome associated with bone metastasis of prostate cancer in vitro. The method displayed superior sensitivity over the shotgun-proteomic approach, and it facilitated the quantification of the relative expression of 276 kinase proteins in a pair of bone metastatic prostate cancer cells. Among the differentially expressed kinases, mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) stimulates the migration and invasion of cultured prostate cancer cells, partially by modulating the activity of secreted matrix metalloproteinases 9 (MMP-9). We also found that the upregulation of MAP4K4 in metastatic prostate cancer cells is driven by the MYC proto-oncogene. Cumulatively, we identify MAP4K4 as a potential promoter for prostate cancer metastasis in vitro.
Collapse
|