Significance of Serine Threonine Tyrosine Kinase 1 As a Drug Resistance Factor and Therapeutic Predictor in Acute Leukemia

Total Page:16

File Type:pdf, Size:1020Kb

Significance of Serine Threonine Tyrosine Kinase 1 As a Drug Resistance Factor and Therapeutic Predictor in Acute Leukemia INTERNATIONAL JOURNAL OF ONCOLOGY 45: 1867-1874, 2014 Significance of serine threonine tyrosine kinase 1 as a drug resistance factor and therapeutic predictor in acute leukemia Shinya Nirasawa, DAISUKE Kobayashi, TAKASHI KONDOH, KAGEAKI Kuribayashi, MAKI TANAKA, NOZOMI YANAGIHARA and NAOKI Watanabe Department of Clinical Laboratory Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan Received June 13, 2014; Accepted July 30, 2014 DOI: 10.3892/ijo.2014.2633 Abstract. Alterations in the mRNA expression or the tutively in patients before therapy and that promote natural mutation of previously reported tyrosine kinases have been resistance. detected only in a limited number of patients with acute The overexpression and mutation of various tyrosine leukemia. In this study, we examined whether the widely kinases contributes to the development of acute leukemia. For expressed serine threonine tyrosine kinase 1 (STYK1)/novel example, the overexpression or mutation of tyrosine kinases, oncogene with kinase domain (NOK) acts as a drug resis- including Flt3, c-kit, platelet-derived growth factor receptor tance factor in acute leukemia. The transfection of leukemic and Bcr-Abl has been reported (1-3). Once these tyrosine HL-60 cells with an STYK1 expression vector resulted in kinases are activated, they transmit major molecules such as the resistance to doxorubicin and etoposide and decreased phosphatidylinositol 3-kinase (PI3K) and mitogen-activated drug-induced caspase-3/7 activity and sub-G1 popula- protein kinase (MAPK) (1-3). This not only induces prolif- tion. To investigate the mechanism of STYK1-induced eration of leukemia cells, but also renders them resistant to drug resistance, microarray analysis was performed using various anticancer drugs. Therefore, several kinase inhibi- HL-60 cells transfected with control or STYK1 expression tors are used clinically and some were reported to serve as vectors. Three tyrosine kinases (EphA4, FLT4 and STK31), predictive factors for the patient response to treatment or prog- two NF-κB inducers (MAPK4 and TNF-RSF11A), and two nosis (4-6). However, these abnormalities are detected only in a genes essential for stem cell replication (SALL4 and NOV) limited number of patients with acute leukemia. Furthermore, were identified as novel STYK1-induced genes. In addition energy-dependent, rapid drug efflux and multidrug-resistance to the data using cell line, a comparison of the leukemic molecules are the major factors for the resistance of several patients who did and did not respond to therapy revealed that leukemic cells; however, drug retention does not always STYK1 expression before therapy was significantly higher correlate with cytotoxicity and these molecules are not always in the non-responder group compared with the group that expressed before therapy. responded completely. These results suggest that STYK1 is We recently reported that serine threonine tyro- a novel drug resistance factor and could be a predictor of the sine kinase 1 (STYK1)/novel oncogene with kinase therapeutic response in acute leukemia. domain (NOK), a receptor protein tyrosine kinase (RPTK)- like protein, is widely expressed in patients with leukemia (7). Introduction We also demonstrated that STYK1 expression was decreased after chemotherapy in all patients, and was important in Anticancer drug resistance is evoked by various mechanisms, the proliferation of leukemia cells. STYK1 was reported to but there is no universal marker that plays a role in drug be a tumorigenesis-inducing factor by Liu et al (8). STYK1 resistance and that is useful to predict the therapeutic effect overexpression results in the growth factor-independent in different disease types. To characterize such markers, it proliferation of murine bone-marrow-derived lymphoid BaF3 is important to identify candidates that are expressed consti- cells and surface adhesion-independent growth and colony formation in NIH3T3 and BaF3 cells (8). STYK1 shares 20-30% homology with members of the fibroblast growth factor receptor and platelet-derived growth factor receptor families (8). RPTKs usually bind to specific ligands to stimu- Correspondence to: Dr Naoki Watanabe, Department of Clinical late tyrosine phosphorylation and growth signal transduction. Laboratory Medicine, Sapporo Medical University School of Medicine, South-1, West-16, Chuo-Ku, Sapporo 060-8543, Japan Interestingly, STYK1 has a single putative transmembrane E-mail: [email protected] domain and an intracellular tyrosine kinase domain, but it lacks an extracellular domain. Therefore, STYK1 expression Key words: serine threonine tyrosine kinase 1, mRNA expression, may itself trigger self-phosphorylation and transmit growth chemo-resistance, prediction, acute leukemia signals without ligand binding. PI3K and MAPK are reported as activated growth signals by STYK1, suggesting that these signals might induce the expression of various molecules 1868 Nirasawa et al: Role of STYK1 in leukemia such as inhibitor of apoptosis proteins (IAPs) and NF-κB Microarrays. Total RNA was prepared from transfected cells, signals to stimulate resistance against anticancer drugs. and then subjected to industrial analysis. Quality control Based on these observations, we hypothesized that the widely check and global gene expression profiling was performed expressed STYK1 may play key roles in the drug resistance by TakaraBio. Inc. (Otsu, Japan) using SurePrint G3 Human of hematopoietic malignancies. However, STYK1 expres- GE 8x60 K Microarrays (both from Agilent Technologies, sion has not yet been reported to act as a resistance factor in Santa Clara, CA, USA) following the Agilent one-color leukemic cells; therefore, whether its expression is related to microarray-based gene expression analysis protocol. The therapeutic outcome in patients with acute leukemia remains slides were scanned using an Agilent Technologies Microarray unclear. Scanner, and the images were processed using Agilent Feature In this study, to determine whether STYK1 functions as Extraction software, version 10.7.3.1. a resistance factor against anticancer drugs, we transfected cells with either a control or STYK1 expression vector and RNA extraction and quantification of STYK1 mRNA expres- compared drug sensitivity and the activation of signaling sion. The expression of STYK1 mRNA was determined by pathways. We next examined STYK1 mRNA expression levels performing qRT-PCR on an ABI PRISM 7700 sequence in patients with acute leukemia and compared its expression detection system (Applied Biosystems, Foster City, CA, in two groups with completely different therapeutic outcomes. USA). Total RNA was isolated, and the concentration was To ensure the applicability of the mRNA expression measure- determined using the GeneQuant DNA/RNA Calculator ments to clinical practice, we used peripheral blood and bone (Amersham Pharmacia Biotech, Uppsala, Sweden). The marrow blood samples without purifying blasts. gene-specific primers and fluorescent hybridization probes used for quantitative PCR were as follows: STYK1 forward Materials and methods primer, 5'-CAT CTT TCG AGC CAA TAT GAA CAC-3'; reverse primer, 5'-TGG AAT TGG ATT CGC CCT AA-3'; Cell culture and anticancer drugs. The human myelogenous and probe, 5'-(FAM) CCA GCT GGG CTC CAT GAG leukemia cell lines HL-60 and K562 were obtained from GTA CAA GAT (TAMRA)-3'. Quantitative RT-PCR was the American Type Culture Collection (Manassas, VA, performed using the TaqMan One-Step RT-PCR Master Mix USA). The cells were cultured in RPMI-1640 (BioWhittaker, Reagents kit (Applied Biosystems). To compare the STYK1 Walkersville, MD, USA) supplemented with 10% heat-inacti- mRNA expression between different samples, the specific vated fetal bovine serum (FBS; Invitrogen, Carlsbad, CA, USA) mRNA was normalized to that of 18S ribosomal RNA at 37˚C in a humidified atmosphere of 5% CO2. Doxorubicin (rRNA) to obtain a ratio. The 18S rRNA expression was and etoposide were purchased from Sigma-Aldrich (St. Louis, determined using TaqMan Ribosomal RNA control reagents MO, USA). (Applied Biosystems), according to the protocol provided by the manufacturer. For each experiment, a calibration curve Patients and samples. A total of 34 peripheral or bone was prepared using control RNA from K562 cells. Briefly, marrow blood samples were examined. Fresh peripheral a computer algorithm was used to analyze the emission blood samples from 22 patients who had been diagnosed with of reporter dye and quenching dye emission during PCR acute leukemia were obtained at Sapporo Medical University amplification, and the intensity of the fluorescence signals Hospital. Fresh bone marrow blood samples were also from each PCR cycle was detected. The amplification curves obtained from 12 patients with acute leukemia. Ten patients obtained from serial dilutions of control RNA were prepared, with acute lymphoblastic leukemia, 20 with acute myeloid and the optimal signal intensity (threshold) was selected leukemia and 4 with acute mixed-lineage leukemia were manually in the exponential phase of the curves. The PCR assessed. A total of 25 patients were newly diagnosed and 9 cycle number (threshold cycle, CT) at each concentration of were relapsed cases. After informed consent was obtained, starting RNA was determined to draw the calibration curve, blood samples were freshly prepared immediately before which was constructed as a xy plot [with the log of the input therapy, hemolyzed using a lysis reagent (Ortho Diagnostic amount
Recommended publications
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • N-Glycan Trimming in the ER and Calnexin/Calreticulin Cycle
    Neurotransmitter receptorsGABA and A postsynapticreceptor activation signal transmission Ligand-gated ion channel transport GABAGABA Areceptor receptor alpha-5 alpha-1/beta-1/gamma-2 subunit GABA A receptor alpha-2/beta-2/gamma-2GABA receptor alpha-4 subunit GABAGABA receptor A receptor beta-3 subunitalpha-6/beta-2/gamma-2 GABA-AGABA receptor; A receptor alpha-1/beta-2/gamma-2GABA receptoralpha-3/beta-2/gamma-2 alpha-3 subunit GABA-A GABAreceptor; receptor benzodiazepine alpha-6 subunit site GABA-AGABA-A receptor; receptor; GABA-A anion site channel (alpha1/beta2 interface) GABA-A receptor;GABA alpha-6/beta-3/gamma-2 receptor beta-2 subunit GABAGABA receptorGABA-A receptor alpha-2receptor; alpha-1 subunit agonist subunit GABA site Serotonin 3a (5-HT3a) receptor GABA receptorGABA-C rho-1 subunitreceptor GlycineSerotonin receptor subunit3 (5-HT3) alpha-1 receptor GABA receptor rho-2 subunit GlycineGlycine receptor receptor subunit subunit alpha-2 alpha-3 Ca2+ activated K+ channels Metabolism of ingested SeMet, Sec, MeSec into H2Se SmallIntermediateSmall conductance conductance conductance calcium-activated calcium-activated calcium-activated potassium potassium potassiumchannel channel protein channel protein 2 protein 1 4 Small conductance calcium-activatedCalcium-activated potassium potassium channel alpha/beta channel 1 protein 3 Calcium-activated potassiumHistamine channel subunit alpha-1 N-methyltransferase Neuraminidase Pyrimidine biosynthesis Nicotinamide N-methyltransferase Adenosylhomocysteinase PolymerasePolymeraseHistidine basic
    [Show full text]
  • YAP and TAZ Mediators at the Crossroad Between Metabolic and Cellular Reprogramming
    H OH metabolites OH Review YAP and TAZ Mediators at the Crossroad between Metabolic and Cellular Reprogramming Giorgia Di Benedetto, Silvia Parisi , Tommaso Russo and Fabiana Passaro * Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 40, 80138 Napoli, Italy; [email protected] (G.D.B.); [email protected] (S.P.); [email protected] (T.R.) * Correspondence: [email protected] Abstract: Cell reprogramming can either refer to a direct conversion of a specialized cell into another or to a reversal of a somatic cell into an induced pluripotent stem cell (iPSC). It implies a peculiar modification of the epigenetic asset and gene regulatory networks needed for a new cell, to better fit the new phenotype of the incoming cell type. Cellular reprogramming also implies a metabolic rearrangement, similar to that observed upon tumorigenesis, with a transition from oxidative phosphorylation to aerobic glycolysis. The induction of a reprogramming process requires a nexus of signaling pathways, mixing a range of local and systemic information, and accumulating evidence points to the crucial role exerted by the Hippo pathway components Yes-Associated Protein (YAP) and Transcriptional Co-activator with PDZ-binding Motif (TAZ). In this review, we will first provide a synopsis of the Hippo pathway and its function during reprogramming and tissue regeneration, then we introduce the latest knowledge on the interplay between YAP/TAZ Citation: Di Benedetto, G.; Parisi, S.; and metabolism and, finally, we discuss the possible role of YAP/TAZ in the orchestration of the Russo, T.; Passaro, F. YAP and TAZ metabolic switch upon cellular reprogramming.
    [Show full text]
  • IGF2BP1 Is a Targetable SRC/MAPK-Dependent Driver Of
    bioRxiv preprint doi: https://doi.org/10.1101/2020.06.19.159905; this version posted June 20, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 IGF2BP1 is a targetable SRC/MAPK-dependent driver of invasive growth in ovarian 2 cancer 3 4 Authors: Nadine Bley1*$#, Annekatrin Schott1*, Simon Müller1, Danny Misiak1, Marcell 5 Lederer1, Tommy Fuchs1, Chris Aßmann1, Markus Glaß1, Christian Ihling2, Andrea Sinz2, 6 Nikolaos Pazaitis3, Claudia Wickenhauser3, Martina Vetter4, Olga Ungurs4, Hans-Georg 7 Strauss4, Christoph Thomssen4, Stefan Hüttelmaier1$. 8 9 10 Addresses: 11 1 Sect. Molecular Cell Biology, Inst. of Molecular Medicine, Charles Tanford Protein Center, 12 Medical Faculty, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3A, 06120 Halle, 13 Germany 14 2 Dept. of Pharmaceutical Chemistry & Bioanalytics, Inst. of Pharmacy, Charles Tanford 15 Protein Center, Martin Luther University Halle-Wittenberg, Kurt-Mothes Str. 3A, 06120 Halle, 16 Germany 17 3 Inst. of Pathology, Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger 18 Str. 14, 06112 Halle, Germany 19 4 Clinics for Gynecology, Medical Faculty, Martin Luther University Halle-Wittenberg, Ernst- 20 Grube-Str. 40, 06120 Halle, Germany 21 22 23 * These authors contributed equally to this work 24 $ shared corresponding authorship 25 # Correspondence: should be addressed to Nadine Bley 26 Running title: IGF2BP1-directed AJ disassembly depends on SRC activation 27 Disclosure of Potential Conflicts of Interests: the authors declare no conflicts of interest.
    [Show full text]
  • Investigation of the Underlying Hub Genes and Molexular Pathogensis in Gastric Cancer by Integrated Bioinformatic Analyses
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Investigation of the underlying hub genes and molexular pathogensis in gastric cancer by integrated bioinformatic analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract The high mortality rate of gastric cancer (GC) is in part due to the absence of initial disclosure of its biomarkers. The recognition of important genes associated in GC is therefore recommended to advance clinical prognosis, diagnosis and and treatment outcomes. The current investigation used the microarray dataset GSE113255 RNA seq data from the Gene Expression Omnibus database to diagnose differentially expressed genes (DEGs). Pathway and gene ontology enrichment analyses were performed, and a proteinprotein interaction network, modules, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. Finally, validation of hub genes was performed. The 1008 DEGs identified consisted of 505 up regulated genes and 503 down regulated genes.
    [Show full text]
  • Ponatinib Shows Potent Antitumor Activity in Small Cell Carcinoma of the Ovary Hypercalcemic Type (SCCOHT) Through Multikinase Inhibition Jessica D
    Published OnlineFirst February 9, 2018; DOI: 10.1158/1078-0432.CCR-17-1928 Cancer Therapy: Preclinical Clinical Cancer Research Ponatinib Shows Potent Antitumor Activity in Small Cell Carcinoma of the Ovary Hypercalcemic Type (SCCOHT) through Multikinase Inhibition Jessica D. Lang1,William P.D. Hendricks1, Krystal A. Orlando2, Hongwei Yin1, Jeffrey Kiefer1, Pilar Ramos1, Ritin Sharma3, Patrick Pirrotte3, Elizabeth A. Raupach1,3, Chris Sereduk1, Nanyun Tang1, Winnie S. Liang1, Megan Washington1, Salvatore J. Facista1, Victoria L. Zismann1, Emily M. Cousins4, Michael B. Major4, Yemin Wang5, Anthony N. Karnezis5, Aleksandar Sekulic1,6, Ralf Hass7, Barbara C. Vanderhyden8, Praveen Nair9, Bernard E. Weissman2, David G. Huntsman5,10, and Jeffrey M. Trent1 Abstract Purpose: Small cell carcinoma of the ovary, hypercalcemic type three SWI/SNF wild-type ovarian cancer cell lines. We further (SCCOHT) is a rare, aggressive ovarian cancer in young women identified ponatinib as the most effective clinically approved that is universally driven by loss of the SWI/SNF ATPase subunits RTK inhibitor. Reexpression of SMARCA4 was shown to confer SMARCA4 and SMARCA2. A great need exists for effective targeted a 1.7-fold increase in resistance to ponatinib. Subsequent therapies for SCCOHT. proteomic assessment of ponatinib target modulation in Experimental Design: To identify underlying therapeutic vul- SCCOHT cell models confirmed inhibition of nine known nerabilities in SCCOHT, we conducted high-throughput siRNA ponatinib target kinases alongside 77 noncanonical ponatinib and drug screens. Complementary proteomics approaches pro- targets in SCCOHT. Finally, ponatinib delayed tumor dou- filed kinases inhibited by ponatinib. Ponatinib was tested for bling time 4-fold in SCCOHT-1 xenografts while reducing efficacy in two patient-derived xenograft (PDX) models and one final tumor volumes in SCCOHT PDX models by 58.6% and cell-line xenograft model of SCCOHT.
    [Show full text]
  • PRKACA Mediates Resistance to HER2-Targeted Therapy in Breast Cancer Cells and Restores Anti-Apoptotic Signaling
    Oncogene (2015) 34, 2061–2071 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc ORIGINAL ARTICLE PRKACA mediates resistance to HER2-targeted therapy in breast cancer cells and restores anti-apoptotic signaling SE Moody1,2,3, AC Schinzel1, S Singh1, F Izzo1, MR Strickland1, L Luo1,2, SR Thomas3, JS Boehm3, SY Kim4, ZC Wang5,6 and WC Hahn1,2,3 Targeting HER2 with antibodies or small molecule inhibitors in HER2-positive breast cancer leads to improved survival, but resistance is a common clinical problem. To uncover novel mechanisms of resistance to anti-HER2 therapy in breast cancer, we performed a kinase open reading frame screen to identify genes that rescue HER2-amplified breast cancer cells from HER2 inhibition or suppression. In addition to multiple members of the MAPK (mitogen-activated protein kinase) and PI3K (phosphoinositide 3-kinase) signaling pathways, we discovered that expression of the survival kinases PRKACA and PIM1 rescued cells from anti-HER2 therapy. Furthermore, we observed elevated PRKACA expression in trastuzumab-resistant breast cancer samples, indicating that this pathway is activated in breast cancers that are clinically resistant to trastuzumab-containing therapy. We found that neither PRKACA nor PIM1 restored MAPK or PI3K activation after lapatinib or trastuzumab treatment, but rather inactivated the pro-apoptotic protein BAD, the BCl-2-associated death promoter, thereby permitting survival signaling through BCL- XL. Pharmacological blockade of BCL-XL/BCL-2 partially abrogated the rescue effects conferred by PRKACA and PIM1, and sensitized cells to lapatinib treatment. These observations suggest that combined targeting of HER2 and the BCL-XL/BCL-2 anti-apoptotic pathway may increase responses to anti-HER2 therapy in breast cancer and decrease the emergence of resistant disease.
    [Show full text]
  • MAPK4 Overexpression Promotes Tumor Progression Via Noncanonical Activation of AKT/Mtor Signaling
    The Journal of Clinical Investigation RESEARCH ARTICLE MAPK4 overexpression promotes tumor progression via noncanonical activation of AKT/mTOR signaling Wei Wang,1 Tao Shen,1 Bingning Dong,1 Chad J. Creighton,2,3 Yanling Meng,1 Wolong Zhou,1 Qing Shi,1 Hao Zhou,1 Yinjie Zhang,1 David D. Moore,1 and Feng Yang1 1Department of Molecular and Cellular Biology, 2Department of Medicine, and 3Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA. MAPK4 is an atypical MAPK. Currently, little is known about its physiological function and involvement in diseases, including cancer. A comprehensive analysis of 8887 gene expression profiles in The Cancer Genome Atlas (TCGA) revealed that MAPK4 overexpression correlates with decreased overall survival, with particularly marked survival effects in patients with lung adenocarcinoma, bladder cancer, low-grade glioma, and thyroid carcinoma. Interestingly, human tumor MAPK4 overexpression also correlated with phosphorylation of AKT, 4E-BP1, and p70S6K, independent of the loss of PTEN or mutation of PIK3CA. This led us to examine whether MAPK4 activates the key metabolic, prosurvival, and proliferative kinase AKT and mTORC1 signaling, independent of the canonical PI3K pathway. We found that MAPK4 activated AKT via a novel, concerted mechanism independent of PI3K. Mechanistically, MAPK4 directly bound and activated AKT by phosphorylation of the activation loop at threonine 308. It also activated mTORC2 to phosphorylate AKT at serine 473 for full activation. MAPK4 overexpression induced oncogenic outcomes, including transforming prostate epithelial cells into anchorage-independent growth, and MAPK4 knockdown inhibited cancer cell proliferation, anchorage-independent growth, and xenograft growth. We concluded that MAPK4 can promote cancer by activating the AKT/mTOR signaling pathway and that targeting MAPK4 may provide a novel therapeutic approach for cancer.
    [Show full text]
  • HHS Public Access Author Manuscript
    HHS Public Access Author manuscript Author Manuscript Author ManuscriptBreast Cancer Author Manuscript Res Treat Author Manuscript . Author manuscript; available in PMC 2016 June 01. Published in final edited form as: Breast Cancer Res Treat. 2015 June ; 151(2): 453–463. doi:10.1007/s10549-015-3401-8. Body mass index associated with genome-wide methylation in breast tissue Brionna Y. Hair1, Zongli Xu2, Erin L. Kirk1, Sophia Harlid2, Rupninder Sandhu3, Whitney R. Robinson1,3, Michael C. Wu4, Andrew F. Olshan1, Kathleen Conway1,3, Jack A. Taylor2, and Melissa A. Troester1 1 Department of Epidemiology, University of North Carolina at Chapel Hill, CB #7435, 2101 McGavran-Greenberg Hall, Chapel Hill, NC 27599-7435, USA 2 Epidemiology Branch, and Epigenomics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences (NIH), Research Triangle Park, NC, USA 3 Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA 4 Fred Hutchinson Cancer Research Center, Seattle, WA, USA Abstract Gene expression studies indicate that body mass index (BMI) is associated with molecular pathways involved in inflammation, insulin-like growth factor activation, and other carcinogenic processes in breast tissue. The goal of this study was to determine whether BMI is associated with gene methylation in breast tissue and to identify pathways that are commonly methylated in association with high BMI. Epigenome-wide methylation profiles were determined using the Illumina HumanMethylation450 BeadChip array in the non-diseased breast tissue of 81 women undergoing breast surgery between 2009 and 2013 at the University of North Carolina Hospitals. Multivariable, robust linear regression was performed to identify methylation sites associated with BMI at a false discovery rate q value <0.05.
    [Show full text]
  • Table 1 Top 100 Phosphorylated Substrates and Their Corresponding Kinases in Chondrosarcoma Cultures As Used for IPA Analysis
    Table 1 Top 100 phosphorylated substrates and their corresponding kinases in chondrosarcoma cultures as used for IPA analysis. Average Fold Adj intensity in Change p- chondrosarcoma Corresponding MSC value cultures Substrate Protein Psite kinase (log2) MSC 1043.42 RKKKVSSTKRH Cytohesin-1 S394 PKC 1.83 0.001 746.95 RKGYRSQRGHS Vitronectin S381 PKC 1.00 0.056 709.03 RARSTSLNERP Tuberin S939 AKT1 1.64 0.008 559.42 SPPRSSLRRSS Transcription elongation factor A-like1 S37 PKC; GSK3 0.18 0.684 515.29 LRRSLSRSMSQ Telethonin S157 Titin 0.77 0.082 510.00 MQPDNSSDSDY CD5 T434 PKA -0.35 0.671 476.27 GGRGGSRARNL Heterogeneous nuclear ribonucleoprotein K S302 PKCdelta 1.03 0.028 455.97 LKPGSSHRKTK Bruton's tyrosine kinase S180 PKCbeta 1.55 0.001 444.65 RRRMASMQRTG E1A binding protein p300 S1834 AKT; p70S6 kinase; pp90Rsk 0.53 0.195 Guanine nucleotide binding protein, alpha Z 440.26 HLRSESQRQRR polypeptide S27 PKC 0.88 0.199 6-phosphofructo-2-kinase/fructose-2,6- 424.12 RPRNYSVGSRP biphosphatase 2 S483 AKT 1.32 0.003 419.61 KKKIATRKPRF Metabotropic glutamate receptor 1 T695 PKC 1.75 0.001 391.21 DNSSDSDYDLH CD5 T453 Lck; Fyn -2.09 0.001 377.39 LRQLRSPRRAQ Ras associated protein Rab4 S204 CDC2 0.63 0.091 376.28 SSQRVSSYRRT Desmin S12 Aurora kinase B 0.56 0.255 369.05 ARIGGSRRERS EP4 receptor S354 PKC 0.29 0.543 RPS6 kinase alpha 3; PKA; 367.99 EPKRRSARLSA HMG14 S7 PKC -0.01 0.996 Peptidylglycine alpha amidating 349.08 SRKGYSRKGFD monooxygenase S930 PKC 0.21 0.678 347.92 RRRLSSLRAST Ribosomal protein S6 S236 PAK2 0.02 0.985 346.84 RSNPPSRKGSG Connexin
    [Show full text]
  • Taqman® Human Protein Kinase Array
    TaqMan® Gene Signature Arrays TaqMan® Human Protein Kinase Array This array is part of a collection of TaqMan® Gene Signature these kinases are from receptor protein-tyrosine kinase (RPTK) Arrays that enable analysis of hundreds of TaqMan® Gene families: EGFR, InsulinR, PDGFR, VEGFR, FGFR, CCK, NGFR, Expression Assays on a micro fluidic card with minimal effort. HGFR, EPHR, AXL, TIE, RYK, DDR, RET, ROS, LTK, ROR and MUSK. The remaining 15 kinases are Ser/Thr kinases from the Protein kinases are one of the largest families of genes in kinase families: CAMKL, IRAK, Lmr, RIPK and STKR. eukaryotes. They belong to one superfamily containing a eukaryotic protein kinase catalytic domain. The ability of kinases We have also selected assays for 26 non-kinase genes in the to reversibly phosphorylate and regulate protein function Human Protein Kinase Array. These genes are involved in signal has been a subject of intense investigation. Kinases are transduction and mediate protein-protein interaction, transcrip- responsible for most of the signal transduction in eukaryotic tional regulation, neural development and cell adhesion. cells, affecting cellular processes including metabolism, References: angiogenesis, hemopoiesis, apoptosis, transcription and Manning, G., Whyte, D.B., Martinez, R., Hunter, T., and differentiation. Protein kinases are also involved in functioning Sudarsanam, S. 2002. The Protein Kinase Complement of the of the nervous and immune systems, in physiologic responses Human Genome. Science 298:1912–34. and in development. Imbalances in signal transduction due to accumulation of mutations or genetic alterations have Blume-Jensen, P. and Hunter, T. 2001. Oncogenic kinase been shown to result in malignant transformation.
    [Show full text]
  • Cardiac Fibroblast P38 MAPK: a Critical Regulator of Myocardial Remodeling
    Journal of Cardiovascular Development and Disease Review Cardiac Fibroblast p38 MAPK: A Critical Regulator of Myocardial Remodeling Neil A. Turner * and Nicola M. Blythe Discovery & Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds LS2 9JT, UK * Correspondence: [email protected] Received: 23 July 2019; Accepted: 6 August 2019; Published: 7 August 2019 Abstract: The cardiac fibroblast is a remarkably versatile cell type that coordinates inflammatory, fibrotic and hypertrophic responses in the heart through a complex array of intracellular and intercellular signaling mechanisms. One important signaling node that has been identified involves p38 MAPK; a family of kinases activated in response to stress and inflammatory stimuli that modulates multiple aspects of cardiac fibroblast function, including inflammatory responses, myofibroblast differentiation, extracellular matrix turnover and the paracrine induction of cardiomyocyte hypertrophy. This review explores the emerging importance of the p38 MAPK pathway in cardiac fibroblasts, describes the molecular mechanisms by which it regulates the expression of key genes, and highlights its potential as a therapeutic target for reducing adverse myocardial remodeling. Keywords: cardiac remodeling; extracellular matrix; fibroblast; fibrosis; heart; hypertrophy; inflammation; myofibroblast; p38 MAP kinase; signal transduction; stress-activated protein kinase; transcription factor 1. Introduction Although once viewed as relatively passive players that solely regulate extracellular matrix (ECM) remodeling, cardiac fibroblasts are now acknowledged as being the primary nodal regulators of multiple aspects of cardiac function under both physiological and pathophysiological conditions [1–4]. In addition to modulating ECM turnover, cardiac fibroblasts contribute to cardiac inflammation, angiogenesis and cardiomyocyte hypertrophy, and hence are viewed as an important potential therapeutic target for ameliorating adverse cardiac remodeling [5].
    [Show full text]