Skip to main content
Top
Published in: Gastric Cancer 5/2020

01-09-2020 | Gastric Cancer | Original Article

Phosphoproteomic analysis identifies CLK1 as a novel therapeutic target in gastric cancer

Authors: Niraj Babu, Sneha M. Pinto, Manjusha Biswas, Tejaswini Subbannayya, Manoj Rajappa, Sonali V. Mohan, Jayshree Advani, Pavithra Rajagopalan, Gajanan Sathe, Nazia Syed, Vinod D. Radhakrishna, Oliyarasi Muthusamy, Sanjay Navani, Rekha V. Kumar, Gopal Gopisetty, Thangarajan Rajkumar, Padhma Radhakrishnan, Saravanan Thiyagarajan, Akhilesh Pandey, Harsha Gowda, Pradip Majumder, Aditi Chatterjee

Published in: Gastric Cancer | Issue 5/2020

Login to get access

Abstract

Background

Phosphorylation is an important regulatory mechanism of protein activity in cells. Studies in various cancers have reported perturbations in kinases resulting in aberrant phosphorylation of oncoproteins and tumor suppressor proteins.

Methods

In this study, we carried out quantitative phosphoproteomic analysis of gastric cancer tissues and corresponding xenograft samples. Using these data, we employed bioinformatics analysis to identify aberrant signaling pathways. We further performed molecular inhibition and silencing of the upstream regulatory kinase in gastric cancer cell lines and validated its effect on cellular phenotype. Through an ex vivo technology utilizing patient tumor and blood sample, we sought to understand the therapeutic potential of the kinase by recreating the tumor microenvironment.

Results

Using mass spectrometry-based high-throughput analysis, we identified 1,344 phosphosites and 848 phosphoproteins, including differential phosphorylation of 177 proteins (fold change cut-off ≥ 1.5). Our data showed that a subset of differentially phosphorylated proteins belonged to splicing machinery. Pathway analysis highlighted Cdc2-like kinase (CLK1) as upstream kinase. Inhibition of CLK1 using TG003 and CLK1 siRNA resulted in a decreased cell viability, proliferation, invasion and migration as well as modulation in the phosphorylation of SRSF2. Ex vivo experiments which utilizes patient’s own tumor and blood to recreate the tumor microenvironment validated the use of CLK1 as a potential target for gastric cancer treatment.

Conclusions

Our data indicates that CLK1 plays a crucial role in the regulation of splicing process in gastric cancer and that CLK1 can act as a novel therapeutic target in gastric cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–E386.PubMed Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–E386.PubMed
2.
go back to reference Schirren R, Reim D, Novotny AR. Adjuvant and/or neoadjuvant therapy for gastric cancer? A perspective review. Ther Adv Med Oncol. 2015;7(1):39–48.PubMedPubMedCentral Schirren R, Reim D, Novotny AR. Adjuvant and/or neoadjuvant therapy for gastric cancer? A perspective review. Ther Adv Med Oncol. 2015;7(1):39–48.PubMedPubMedCentral
3.
go back to reference Ott K, Sendler A, Becker K, Dittler HJ, Helmberger H, Busch R, et al. Neoadjuvant chemotherapy with cisplatin, 5-FU, and leucovorin (PLF) in locally advanced gastric cancer: a prospective phase II study. Gastric Cancer. 2003;6(3):159–67.PubMed Ott K, Sendler A, Becker K, Dittler HJ, Helmberger H, Busch R, et al. Neoadjuvant chemotherapy with cisplatin, 5-FU, and leucovorin (PLF) in locally advanced gastric cancer: a prospective phase II study. Gastric Cancer. 2003;6(3):159–67.PubMed
4.
go back to reference Rustum YM. Biochemical rationale for the 5-fluorouracil leucovorin combination and update of clinical experience. J Chemother. 1990;2(Suppl 1):5–11.PubMed Rustum YM. Biochemical rationale for the 5-fluorouracil leucovorin combination and update of clinical experience. J Chemother. 1990;2(Suppl 1):5–11.PubMed
5.
go back to reference Apicella M, Corso S, Giordano S. Targeted therapies for gastric cancer: failures and hopes from clinical trials. Oncotarget. 2017;8(34):57654–69.PubMedPubMedCentral Apicella M, Corso S, Giordano S. Targeted therapies for gastric cancer: failures and hopes from clinical trials. Oncotarget. 2017;8(34):57654–69.PubMedPubMedCentral
6.
go back to reference Bang YJ, Kang YK, Kang WK, Boku N, Chung HC, Chen JS, et al. Phase II study of sunitinib as second-line treatment for advanced gastric cancer. Investig New Drugs. 2011;29(6):1449–588. Bang YJ, Kang YK, Kang WK, Boku N, Chung HC, Chen JS, et al. Phase II study of sunitinib as second-line treatment for advanced gastric cancer. Investig New Drugs. 2011;29(6):1449–588.
7.
go back to reference Papaetis GS, Syrigos KN. Sunitinib: a multitargeted receptor tyrosine kinase inhibitor in the era of molecular cancer therapies. BioDrugs. 2009;23(6):377–89.PubMed Papaetis GS, Syrigos KN. Sunitinib: a multitargeted receptor tyrosine kinase inhibitor in the era of molecular cancer therapies. BioDrugs. 2009;23(6):377–89.PubMed
8.
go back to reference Erdem GU, Bozkaya Y, Ozdemir NY, Demirci NS, Yazici O, Zengin N. 5-Fluorouracil, leucovorin, and irinotecan (FOLFIRI) as a third-line chemotherapy treatment in metastatic gastric cancer, after failure of fluoropyrimidine, platinum, anthracycline, and taxane. Bosn J Basic Med Sci. 2018;18(2):170–7.PubMedPubMedCentral Erdem GU, Bozkaya Y, Ozdemir NY, Demirci NS, Yazici O, Zengin N. 5-Fluorouracil, leucovorin, and irinotecan (FOLFIRI) as a third-line chemotherapy treatment in metastatic gastric cancer, after failure of fluoropyrimidine, platinum, anthracycline, and taxane. Bosn J Basic Med Sci. 2018;18(2):170–7.PubMedPubMedCentral
9.
go back to reference Moehler M, Gepfner-Tuma I, Maderer A, Thuss-Patience PC, Ruessel J, Hegewisch-Becker S, et al. Sunitinib added to FOLFIRI versus FOLFIRI in patients with chemorefractory advanced adenocarcinoma of the stomach or lower esophagus: a randomized, placebo-controlled phase II AIO trial with serum biomarker program. BMC Cancer. 2016;16:699.PubMedPubMedCentral Moehler M, Gepfner-Tuma I, Maderer A, Thuss-Patience PC, Ruessel J, Hegewisch-Becker S, et al. Sunitinib added to FOLFIRI versus FOLFIRI in patients with chemorefractory advanced adenocarcinoma of the stomach or lower esophagus: a randomized, placebo-controlled phase II AIO trial with serum biomarker program. BMC Cancer. 2016;16:699.PubMedPubMedCentral
10.
go back to reference Pompili L, Porru M, Caruso C, Biroccio A, Leonetti C. Patient-derived xenografts: a relevant preclinical model for drug development. J Exp Clin Cancer Res. 2016;35(1):189.PubMedPubMedCentral Pompili L, Porru M, Caruso C, Biroccio A, Leonetti C. Patient-derived xenografts: a relevant preclinical model for drug development. J Exp Clin Cancer Res. 2016;35(1):189.PubMedPubMedCentral
11.
go back to reference Black DL. Mechanisms of alternative pre-messenger RNA splicing. Annu Rev Biochem. 2003;72:291–336.PubMed Black DL. Mechanisms of alternative pre-messenger RNA splicing. Annu Rev Biochem. 2003;72:291–336.PubMed
12.
go back to reference Shkreta L, Bell B, Revil T, Venables JP, Prinos P, Elela SA, et al. Cancer-associated perturbations in alternative pre-messenger RNA splicing. Cancer Treat Res. 2013;158:41–944.PubMed Shkreta L, Bell B, Revil T, Venables JP, Prinos P, Elela SA, et al. Cancer-associated perturbations in alternative pre-messenger RNA splicing. Cancer Treat Res. 2013;158:41–944.PubMed
13.
go back to reference Scotti MM, Swanson MS. RNA mis-splicing in disease. Nat Rev Genet. 2016;17(1):19–32.PubMed Scotti MM, Swanson MS. RNA mis-splicing in disease. Nat Rev Genet. 2016;17(1):19–32.PubMed
14.
go back to reference Mermoud JE, Cohen PT, Lamond AI. Regulation of mammalian spliceosome assembly by a protein phosphorylation mechanism. EMBO J. 1994;13(23):5679–88.PubMedPubMedCentral Mermoud JE, Cohen PT, Lamond AI. Regulation of mammalian spliceosome assembly by a protein phosphorylation mechanism. EMBO J. 1994;13(23):5679–88.PubMedPubMedCentral
15.
go back to reference Amin EM, Oltean S, Hua J, Gammons MV, Hamdollah-Zadeh M, Welsh GI, et al. WT1 mutants reveal SRPK1 to be a downstream angiogenesis target by altering VEGF splicing. Cancer Cell. 2011;20(6):768–80.PubMedPubMedCentral Amin EM, Oltean S, Hua J, Gammons MV, Hamdollah-Zadeh M, Welsh GI, et al. WT1 mutants reveal SRPK1 to be a downstream angiogenesis target by altering VEGF splicing. Cancer Cell. 2011;20(6):768–80.PubMedPubMedCentral
16.
go back to reference Shultz JC, Goehe RW, Wijesinghe DS, Murudkar C, Hawkins AJ, Shay JW, et al. Alternative splicing of caspase 9 is modulated by the phosphoinositide 3-kinase/Akt pathway via phosphorylation of SRp30a. Cancer Res. 2010;70(22):9185–96.PubMedPubMedCentral Shultz JC, Goehe RW, Wijesinghe DS, Murudkar C, Hawkins AJ, Shay JW, et al. Alternative splicing of caspase 9 is modulated by the phosphoinositide 3-kinase/Akt pathway via phosphorylation of SRp30a. Cancer Res. 2010;70(22):9185–96.PubMedPubMedCentral
17.
go back to reference Majumder B, Baraneedharan U, Thiyagarajan S, Radhakrishnan P, Narasimhan H, Dhandapani M, et al. Predicting clinical response to anticancer drugs using an ex vivo platform that captures tumour heterogeneity. Nat Commun. 2015;6:6169.PubMed Majumder B, Baraneedharan U, Thiyagarajan S, Radhakrishnan P, Narasimhan H, Dhandapani M, et al. Predicting clinical response to anticancer drugs using an ex vivo platform that captures tumour heterogeneity. Nat Commun. 2015;6:6169.PubMed
18.
go back to reference Radhakrishnan A, Nanjappa V, Raja R, Sathe G, Puttamallesh VN, Jain AP, et al. Corrigendum: a dual specificity kinase, DYRK1A, as a potential therapeutic target for head and neck squamous cell carcinoma. Sci Rep. 2017;7:46864.PubMedPubMedCentral Radhakrishnan A, Nanjappa V, Raja R, Sathe G, Puttamallesh VN, Jain AP, et al. Corrigendum: a dual specificity kinase, DYRK1A, as a potential therapeutic target for head and neck squamous cell carcinoma. Sci Rep. 2017;7:46864.PubMedPubMedCentral
19.
go back to reference Syed N, Chavan S, Sahasrabuddhe NA, Renuse S, Sathe G, Nanjappa V, et al. Silencing of high-mobility group box 2 (HMGB2) modulates cisplatin and 5-fluorouracil sensitivity in head and neck squamous cell carcinoma. Proteomics. 2015;15(2–3):383–93.PubMedPubMedCentral Syed N, Chavan S, Sahasrabuddhe NA, Renuse S, Sathe G, Nanjappa V, et al. Silencing of high-mobility group box 2 (HMGB2) modulates cisplatin and 5-fluorouracil sensitivity in head and neck squamous cell carcinoma. Proteomics. 2015;15(2–3):383–93.PubMedPubMedCentral
20.
go back to reference Radhakrishnan A, Nanjappa V, Raja R, Sathe G, Puttamallesh VN, Jain AP, et al. A dual specificity kinase, DYRK1A, as a potential therapeutic target for head and neck squamous cell carcinoma. Sci Rep. 2016;6:36132.PubMedPubMedCentral Radhakrishnan A, Nanjappa V, Raja R, Sathe G, Puttamallesh VN, Jain AP, et al. A dual specificity kinase, DYRK1A, as a potential therapeutic target for head and neck squamous cell carcinoma. Sci Rep. 2016;6:36132.PubMedPubMedCentral
21.
go back to reference Subbannayya T, Leal-Rojas P, Barbhuiya MA, Raja R, Renuse S, Sathe G, et al. Macrophage migration inhibitory factor—a therapeutic target in gallbladder cancer. BMC Cancer. 2015;15:843.PubMedPubMedCentral Subbannayya T, Leal-Rojas P, Barbhuiya MA, Raja R, Renuse S, Sathe G, et al. Macrophage migration inhibitory factor—a therapeutic target in gallbladder cancer. BMC Cancer. 2015;15:843.PubMedPubMedCentral
22.
go back to reference Subbannayya Y, Syed N, Barbhuiya MA, Raja R, Marimuthu A, Sahasrabuddhe N, et al. Calcium calmodulin dependent kinase kinase 2—a novel therapeutic target for gastric adenocarcinoma. Cancer Biol Ther. 2015;16(2):336–45.PubMedPubMedCentral Subbannayya Y, Syed N, Barbhuiya MA, Raja R, Marimuthu A, Sahasrabuddhe N, et al. Calcium calmodulin dependent kinase kinase 2—a novel therapeutic target for gastric adenocarcinoma. Cancer Biol Ther. 2015;16(2):336–45.PubMedPubMedCentral
23.
go back to reference Cormier N, Yeo A, Fiorentino E, Paxson J. Optimization of the wound scratch assay to detect changes in murine mesenchymal stromal cell migration after damage by soluble cigarette smoke extract. J Vis Exp. 2015;106:e53414. Cormier N, Yeo A, Fiorentino E, Paxson J. Optimization of the wound scratch assay to detect changes in murine mesenchymal stromal cell migration after damage by soluble cigarette smoke extract. J Vis Exp. 2015;106:e53414.
24.
go back to reference Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9(7):671–5.PubMedPubMedCentral Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9(7):671–5.PubMedPubMedCentral
25.
go back to reference Radhakrishnan P, Baraneedharan U, Veluchamy S, Dhandapani M, Pinto DD, Thiyagarajan S, et al. Inhibition of rapamycin-induced AKT activation elicits differential antitumor response in head and neck cancers. Cancer Res. 2013;73(3):1118–27.PubMed Radhakrishnan P, Baraneedharan U, Veluchamy S, Dhandapani M, Pinto DD, Thiyagarajan S, et al. Inhibition of rapamycin-induced AKT activation elicits differential antitumor response in head and neck cancers. Cancer Res. 2013;73(3):1118–27.PubMed
26.
go back to reference Brijwani N, Jain M, Dhandapani M, Zahed F, Mukhopadhyay P, Biswas M, et al. Rationally co-targeting divergent pathways in KRAS wild-type colorectal cancers by CANscript technology reveals tumor dependence on Notch and Erbb2. Sci Rep. 2017;7(1):1502.PubMedPubMedCentral Brijwani N, Jain M, Dhandapani M, Zahed F, Mukhopadhyay P, Biswas M, et al. Rationally co-targeting divergent pathways in KRAS wild-type colorectal cancers by CANscript technology reveals tumor dependence on Notch and Erbb2. Sci Rep. 2017;7(1):1502.PubMedPubMedCentral
27.
go back to reference Tyanova S, Temu T, Sinitcyn P, Carlson A, Hein MY, Geiger T, et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat Methods. 2016;13(9):731–40.PubMed Tyanova S, Temu T, Sinitcyn P, Carlson A, Hein MY, Geiger T, et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat Methods. 2016;13(9):731–40.PubMed
28.
go back to reference Cho S, Hoang A, Sinha R, Zhong XY, Fu XD, Krainer AR, et al. Interaction between the RNA binding domains of Ser–Arg splicing factor 1 and U1-70K snRNP protein determines early spliceosome assembly. Proc Natl Acad Sci USA. 2011;108(20):8233–8.PubMedPubMedCentral Cho S, Hoang A, Sinha R, Zhong XY, Fu XD, Krainer AR, et al. Interaction between the RNA binding domains of Ser–Arg splicing factor 1 and U1-70K snRNP protein determines early spliceosome assembly. Proc Natl Acad Sci USA. 2011;108(20):8233–8.PubMedPubMedCentral
29.
go back to reference Fu XD, Maniatis T. The 35-kDa mammalian splicing factor SC35 mediates specific interactions between U1 and U2 small nuclear ribonucleoprotein particles at the 3′ splice site. Proc Natl Acad Sci USA. 1992;89(5):1725–9.PubMedPubMedCentral Fu XD, Maniatis T. The 35-kDa mammalian splicing factor SC35 mediates specific interactions between U1 and U2 small nuclear ribonucleoprotein particles at the 3′ splice site. Proc Natl Acad Sci USA. 1992;89(5):1725–9.PubMedPubMedCentral
30.
go back to reference Zhou Z, Fu XD. Regulation of splicing by SR proteins and SR protein-specific kinases. Chromosoma. 2013;122(3):191–207.PubMedPubMedCentral Zhou Z, Fu XD. Regulation of splicing by SR proteins and SR protein-specific kinases. Chromosoma. 2013;122(3):191–207.PubMedPubMedCentral
31.
go back to reference Mai S, Qu X, Li P, Ma Q, Cao C, Liu X. Global regulation of alternative RNA splicing by the SR-rich protein RBM39. Biochim Biophys Acta. 2016;1859(8):1014–24.PubMed Mai S, Qu X, Li P, Ma Q, Cao C, Liu X. Global regulation of alternative RNA splicing by the SR-rich protein RBM39. Biochim Biophys Acta. 2016;1859(8):1014–24.PubMed
32.
go back to reference Hsu TY, Simon LM, Neill NJ, Marcotte R, Sayad A, Bland CS, et al. The spliceosome is a therapeutic vulnerability in MYC-driven cancer. Nature. 2015;525(7569):384–8.PubMedPubMedCentral Hsu TY, Simon LM, Neill NJ, Marcotte R, Sayad A, Bland CS, et al. The spliceosome is a therapeutic vulnerability in MYC-driven cancer. Nature. 2015;525(7569):384–8.PubMedPubMedCentral
33.
go back to reference Liu Y, Conaway L, Rutherford Bethard J, Al-Ayoubi AM, Thompson Bradley A, Zheng H, et al. Phosphorylation of the alternative mRNA splicing factor 45 (SPF45) by Clk1 regulates its splice site utilization, cell migration and invasion. Nucleic Acids Res. 2013;41(9):4949–62.PubMedPubMedCentral Liu Y, Conaway L, Rutherford Bethard J, Al-Ayoubi AM, Thompson Bradley A, Zheng H, et al. Phosphorylation of the alternative mRNA splicing factor 45 (SPF45) by Clk1 regulates its splice site utilization, cell migration and invasion. Nucleic Acids Res. 2013;41(9):4949–62.PubMedPubMedCentral
34.
go back to reference Zhang L, Yang H, Zhang W, Liang Z, Huang Q, Xu G, et al. Clk1-regulated aerobic glycolysis is involved in glioma chemoresistance. J Neurochem. 2017;142(4):574–88.PubMed Zhang L, Yang H, Zhang W, Liang Z, Huang Q, Xu G, et al. Clk1-regulated aerobic glycolysis is involved in glioma chemoresistance. J Neurochem. 2017;142(4):574–88.PubMed
35.
go back to reference Muraki M, Ohkawara B, Hosoya T, Onogi H, Koizumi J, Koizumi T, et al. Manipulation of alternative splicing by a newly developed inhibitor of Clks. J Biol Chem. 2004;279(23):24246–544.PubMed Muraki M, Ohkawara B, Hosoya T, Onogi H, Koizumi J, Koizumi T, et al. Manipulation of alternative splicing by a newly developed inhibitor of Clks. J Biol Chem. 2004;279(23):24246–544.PubMed
36.
go back to reference Edmond V, Merdzhanova G, Gout S, Brambilla E, Gazzeri S, Eymin B. A new function of the splicing factor SRSF2 in the control of E2F1-mediated cell cycle progression in neuroendocrine lung tumors. Cell Cycle. 2013;12(8):1267–78.PubMedPubMedCentral Edmond V, Merdzhanova G, Gout S, Brambilla E, Gazzeri S, Eymin B. A new function of the splicing factor SRSF2 in the control of E2F1-mediated cell cycle progression in neuroendocrine lung tumors. Cell Cycle. 2013;12(8):1267–78.PubMedPubMedCentral
37.
go back to reference Razanau A, Xie J. Emerging mechanisms and consequences of calcium regulation of alternative splicing in neurons and endocrine cells. Cell Mol Life Sci. 2013;70(23):4527–36.PubMed Razanau A, Xie J. Emerging mechanisms and consequences of calcium regulation of alternative splicing in neurons and endocrine cells. Cell Mol Life Sci. 2013;70(23):4527–36.PubMed
38.
go back to reference Radhakrishnan A, Nanjappa V, Raja R, Sathe G, Chavan S, Nirujogi RS, et al. Dysregulation of splicing proteins in head and neck squamous cell carcinoma. Cancer Biol Ther. 2016;17(2):219–29.PubMedPubMedCentral Radhakrishnan A, Nanjappa V, Raja R, Sathe G, Chavan S, Nirujogi RS, et al. Dysregulation of splicing proteins in head and neck squamous cell carcinoma. Cancer Biol Ther. 2016;17(2):219–29.PubMedPubMedCentral
40.
go back to reference Wiredja DD, Ayati M, Mazhar S, Sangodkar J, Maxwell S, Schlatzer D, et al. Phosphoproteomics profiling of nonsmall cell lung cancer cells treated with a novel phosphatase activator. Proteomics. 2017;17(22):1700214. Wiredja DD, Ayati M, Mazhar S, Sangodkar J, Maxwell S, Schlatzer D, et al. Phosphoproteomics profiling of nonsmall cell lung cancer cells treated with a novel phosphatase activator. Proteomics. 2017;17(22):1700214.
41.
go back to reference Francavilla C, Lupia M, Tsafou K, Villa A, Kowalczyk K, Rakownikow Jersie-Christensen R, et al. Phosphoproteomics of primary cells reveals druggable kinase signatures in ovarian cancer. Cell Rep. 2017;18(13):3242–56.PubMedPubMedCentral Francavilla C, Lupia M, Tsafou K, Villa A, Kowalczyk K, Rakownikow Jersie-Christensen R, et al. Phosphoproteomics of primary cells reveals druggable kinase signatures in ovarian cancer. Cell Rep. 2017;18(13):3242–56.PubMedPubMedCentral
42.
go back to reference Alli-Shaik A, Wee S, Lim LHK, Gunaratne J. Phosphoproteomics reveals network rewiring to a pro-adhesion state in annexin-1-deficient mammary epithelial cells. Breast Cancer Res. 2017;19(1):132.PubMedPubMedCentral Alli-Shaik A, Wee S, Lim LHK, Gunaratne J. Phosphoproteomics reveals network rewiring to a pro-adhesion state in annexin-1-deficient mammary epithelial cells. Breast Cancer Res. 2017;19(1):132.PubMedPubMedCentral
43.
go back to reference Guo T, Lee SS, Ng WH, Zhu Y, Gan CS, Zhu J, et al. Global molecular dysfunctions in gastric cancer revealed by an integrated analysis of the phosphoproteome and transcriptome. Cell Mol Life Sci. 2011;68(11):1983–2002.PubMed Guo T, Lee SS, Ng WH, Zhu Y, Gan CS, Zhu J, et al. Global molecular dysfunctions in gastric cancer revealed by an integrated analysis of the phosphoproteome and transcriptome. Cell Mol Life Sci. 2011;68(11):1983–2002.PubMed
44.
go back to reference Lin LL, Huang HC, Juan HF. Discovery of biomarkers for gastric cancer: a proteomics approach. J Proteom. 2012;75(11):3081–97. Lin LL, Huang HC, Juan HF. Discovery of biomarkers for gastric cancer: a proteomics approach. J Proteom. 2012;75(11):3081–97.
45.
go back to reference Zahari MS, Wu X, Pinto SM, Nirujogi RS, Kim MS, Fetics B, et al. Phosphoproteomic profiling of tumor tissues identifies HSP27 Ser82 phosphorylation as a robust marker of early ischemia. Sci Rep. 2015;5:13660.PubMedPubMedCentral Zahari MS, Wu X, Pinto SM, Nirujogi RS, Kim MS, Fetics B, et al. Phosphoproteomic profiling of tumor tissues identifies HSP27 Ser82 phosphorylation as a robust marker of early ischemia. Sci Rep. 2015;5:13660.PubMedPubMedCentral
46.
go back to reference Choi SY, Lin D, Gout PW, Collins CC, Xu Y, Wang Y. Lessons from patient-derived xenografts for better in vitro modeling of human cancer. Adv Drug Deliv Rev. 2014;79:222–37.PubMed Choi SY, Lin D, Gout PW, Collins CC, Xu Y, Wang Y. Lessons from patient-derived xenografts for better in vitro modeling of human cancer. Adv Drug Deliv Rev. 2014;79:222–37.PubMed
47.
go back to reference Damhofer H, Ebbing EA, Steins A, Welling L, Tol JA, Krishnadath KK, et al. Establishment of patient-derived xenograft models and cell lines for malignancies of the upper gastrointestinal tract. J Transl Med. 2015;13:115.PubMedPubMedCentral Damhofer H, Ebbing EA, Steins A, Welling L, Tol JA, Krishnadath KK, et al. Establishment of patient-derived xenograft models and cell lines for malignancies of the upper gastrointestinal tract. J Transl Med. 2015;13:115.PubMedPubMedCentral
48.
go back to reference Borodovsky A, McQuiston TJ, Stetson D, Ahmed A, Whitston D, Zhang J, et al. Generation of stable PDX derived cell lines using conditional reprogramming. Mol Cancer. 2017;16(1):177.PubMedPubMedCentral Borodovsky A, McQuiston TJ, Stetson D, Ahmed A, Whitston D, Zhang J, et al. Generation of stable PDX derived cell lines using conditional reprogramming. Mol Cancer. 2017;16(1):177.PubMedPubMedCentral
49.
go back to reference Zhang T, Zhang L, Fan S, Zhang M, Fu H, Liu Y, et al. Patient-derived gastric carcinoma xenograft mouse models faithfully represent human tumor molecular diversity. PLoS One. 2015;10(7):e0134493.PubMedPubMedCentral Zhang T, Zhang L, Fan S, Zhang M, Fu H, Liu Y, et al. Patient-derived gastric carcinoma xenograft mouse models faithfully represent human tumor molecular diversity. PLoS One. 2015;10(7):e0134493.PubMedPubMedCentral
50.
go back to reference Naro C, Sette C. Phosphorylation-mediated regulation of alternative splicing in cancer. Int J Cell Biol. 2013;2013:151839.PubMedPubMedCentral Naro C, Sette C. Phosphorylation-mediated regulation of alternative splicing in cancer. Int J Cell Biol. 2013;2013:151839.PubMedPubMedCentral
51.
go back to reference Colwill K, Pawson T, Andrews B, Prasad J, Manley JL, Bell JC, et al. The Clk/Sty protein kinase phosphorylates SR splicing factors and regulates their intranuclear distribution. EMBO J. 1996;15(2):265–75.PubMedPubMedCentral Colwill K, Pawson T, Andrews B, Prasad J, Manley JL, Bell JC, et al. The Clk/Sty protein kinase phosphorylates SR splicing factors and regulates their intranuclear distribution. EMBO J. 1996;15(2):265–75.PubMedPubMedCentral
52.
go back to reference Bonomi S, Gallo S, Catillo M, Pignataro D, Biamonti G, Ghigna C. Oncogenic alternative splicing switches: role in cancer progression and prospects for therapy. Int J Cell Biol. 2013;2013:962038.PubMedPubMedCentral Bonomi S, Gallo S, Catillo M, Pignataro D, Biamonti G, Ghigna C. Oncogenic alternative splicing switches: role in cancer progression and prospects for therapy. Int J Cell Biol. 2013;2013:962038.PubMedPubMedCentral
53.
go back to reference Bemmo A, Dias C, Rose AA, Russo C, Siegel P, Majewski J. Exon-level transcriptome profiling in murine breast cancer reveals splicing changes specific to tumors with different metastatic abilities. PLoS One. 2010;5(8):e11981.PubMedPubMedCentral Bemmo A, Dias C, Rose AA, Russo C, Siegel P, Majewski J. Exon-level transcriptome profiling in murine breast cancer reveals splicing changes specific to tumors with different metastatic abilities. PLoS One. 2010;5(8):e11981.PubMedPubMedCentral
54.
go back to reference Araki S, Dairiki R, Nakayama Y, Murai A, Miyashita R, Iwatani M, et al. Inhibitors of CLK protein kinases suppress cell growth and induce apoptosis by modulating pre-mRNA splicing. PLoS One. 2015;10(1):e0116929.PubMedPubMedCentral Araki S, Dairiki R, Nakayama Y, Murai A, Miyashita R, Iwatani M, et al. Inhibitors of CLK protein kinases suppress cell growth and induce apoptosis by modulating pre-mRNA splicing. PLoS One. 2015;10(1):e0116929.PubMedPubMedCentral
55.
go back to reference Li P, Carter G, Romero J, Gower KM, Watson J, Patel NA, et al. Clk/STY (cdc2-like kinase 1) and Akt regulate alternative splicing and adipogenesis in 3T3-L1 pre-adipocytes. PLoS One. 2013;8(1):e53268.PubMedPubMedCentral Li P, Carter G, Romero J, Gower KM, Watson J, Patel NA, et al. Clk/STY (cdc2-like kinase 1) and Akt regulate alternative splicing and adipogenesis in 3T3-L1 pre-adipocytes. PLoS One. 2013;8(1):e53268.PubMedPubMedCentral
56.
go back to reference ElHady AK, Abdel-Halim M, Abadi AH, Engel M. Development of selective Clk1 and -4 inhibitors for cellular depletion of cancer-relevant proteins. J Med Chem. 2017;60(13):5377–91.PubMed ElHady AK, Abdel-Halim M, Abadi AH, Engel M. Development of selective Clk1 and -4 inhibitors for cellular depletion of cancer-relevant proteins. J Med Chem. 2017;60(13):5377–91.PubMed
57.
go back to reference Fedorov O, Huber K, Eisenreich A, Filippakopoulos P, King O, Bullock AN, et al. Specific CLK inhibitors from a novel chemotype for regulation of alternative splicing. Chem Biol. 2011;18(1):67–766.PubMedPubMedCentral Fedorov O, Huber K, Eisenreich A, Filippakopoulos P, King O, Bullock AN, et al. Specific CLK inhibitors from a novel chemotype for regulation of alternative splicing. Chem Biol. 2011;18(1):67–766.PubMedPubMedCentral
58.
go back to reference Eisenreich A, Zakrzewicz A, Huber K, Thierbach H, Pepke W, Goldin-Lang P, et al. Regulation of pro-angiogenic tissue factor expression in hypoxia-induced human lung cancer cells. Oncol Rep. 2013;30(1):462–70.PubMed Eisenreich A, Zakrzewicz A, Huber K, Thierbach H, Pepke W, Goldin-Lang P, et al. Regulation of pro-angiogenic tissue factor expression in hypoxia-induced human lung cancer cells. Oncol Rep. 2013;30(1):462–70.PubMed
59.
go back to reference Mavrou A, Brakspear K, Hamdollah-Zadeh M, Damodaran G, Babaei-Jadidi R, Oxley J, et al. Serine–arginine protein kinase 1 (SRPK1) inhibition as a potential novel targeted therapeutic strategy in prostate cancer. Oncogene. 2015;34(33):4311–9.PubMed Mavrou A, Brakspear K, Hamdollah-Zadeh M, Damodaran G, Babaei-Jadidi R, Oxley J, et al. Serine–arginine protein kinase 1 (SRPK1) inhibition as a potential novel targeted therapeutic strategy in prostate cancer. Oncogene. 2015;34(33):4311–9.PubMed
60.
go back to reference Cao W, Jamison SF, Garcia-Blanco MA. Both phosphorylation and dephosphorylation of ASF/SF2 are required for pre-mRNA splicing in vitro. RNA. 1997;3(12):1456–67.PubMedPubMedCentral Cao W, Jamison SF, Garcia-Blanco MA. Both phosphorylation and dephosphorylation of ASF/SF2 are required for pre-mRNA splicing in vitro. RNA. 1997;3(12):1456–67.PubMedPubMedCentral
61.
go back to reference Chanseok Shin JLM. The SR protein SRp38 represses splicing in M phase cells. Cell. 2002;111(3):10. Chanseok Shin JLM. The SR protein SRp38 represses splicing in M phase cells. Cell. 2002;111(3):10.
Metadata
Title
Phosphoproteomic analysis identifies CLK1 as a novel therapeutic target in gastric cancer
Authors
Niraj Babu
Sneha M. Pinto
Manjusha Biswas
Tejaswini Subbannayya
Manoj Rajappa
Sonali V. Mohan
Jayshree Advani
Pavithra Rajagopalan
Gajanan Sathe
Nazia Syed
Vinod D. Radhakrishna
Oliyarasi Muthusamy
Sanjay Navani
Rekha V. Kumar
Gopal Gopisetty
Thangarajan Rajkumar
Padhma Radhakrishnan
Saravanan Thiyagarajan
Akhilesh Pandey
Harsha Gowda
Pradip Majumder
Aditi Chatterjee
Publication date
01-09-2020
Publisher
Springer Singapore
Published in
Gastric Cancer / Issue 5/2020
Print ISSN: 1436-3291
Electronic ISSN: 1436-3305
DOI
https://doi.org/10.1007/s10120-020-01062-8

Other articles of this Issue 5/2020

Gastric Cancer 5/2020 Go to the issue