The Role of MAP4K4 in Cardiac Muscle Cell Death

Total Page:16

File Type:pdf, Size:1020Kb

The Role of MAP4K4 in Cardiac Muscle Cell Death The role of MAP4K4 in cardiac muscle cell death Micaela M. Jenkins CID: 00855768 National Heart and Lung Institute Faculty of Medicine Imperial College London A Thesis submitted to Imperial College London for Doctor of Philosophy 1 National Heart and Lung Institute Word count: 65,028 2 Acknowledgements I express my sincerest thanks to Professor Michael Schneider and Professor Sian Harding for enabling the opportunity to work on this exciting project as well as for all their help, support and advice during course of my studies. I am also very grateful to the British Heart Foundation for awarding me the studentship to undertake this research. I would like to acknowledge the extensive work carried out by the MAP4K4 team past and present, which provided the foundations for this study. I am especially grateful to Lorna Fiedler for all her scientific and moral support all the way through from my MRes days to this day. Lorna’s insight into the project has been absolutely invaluable to me as has her guidance and support. I am extremely grateful to Dr Michela Noseda for imparting to me the research and technical skills necessary to complete the work hereby presented as well as for the daily support and guidance on every respect. I am indebted to Tom Owen, Eleanor Humphrey and Carolina Pinto Ricardo not only for their generous technical and academic assistance but also for accompanying me in the happy as well as hard moments during these three years. I would also like to thank all the members of the Schneider and Harding laboratories for their support and advice and for generally making my days at work so enjoyable. I would also like to acknowledge the Carling group for having provided me with training on the Seahorse platform and kindly allowed me to use their equipment on a regular basis. Special thanks to my family, friends and particularly Quin. It was your encouragement and support that took me through these years and always made me see the light at the end of the tunnel. Finally, I would like to dedicate this thesis to my mother and grandfather, my everlasting sources of inspiration and strength. 3 Declaration of Originality I declare that the work hereby presented was conducted by the author, except where indicated by special reference in the text, and that no part of the dissertation has been submitted for any other degree. Some of the results contained in this thesis have been presented at conferences and seminars. Micaela M. Jenkins 4 Copyright Declaration ‘The copyright of this thesis rests with the author and is made available under a Creative Commons Attribution Non-Commercial No Derivatives licence. Researchers are free to copy, distribute or transmit the thesis on the condition that they attribute it, that they do not use it for commercial purposes and that they do not alter, transform or build upon it. For any reuse or redistribution, researchers must make clear to others the licence terms of this work’ 5 Table of contents Acknowledgements ................................................................................................................... 3 Declaration of Originality ........................................................................................................... 4 Copyright Declaration................................................................................................................ 5 Table of contents ...................................................................................................................... 6 List of Figures ......................................................................................................................... 10 List of Tables .......................................................................................................................... 14 List of abbreviations ................................................................................................................ 15 Abstract................................................................................................................................... 19 1. Introduction ......................................................................................................................... 20 1.1 Heart failure as a socio-economic problem ................................................................ 21 1.2 Pathobiology of heart failure ...................................................................................... 24 1.3 Cell death signalling pathways ................................................................................... 28 1.3.1 Apoptosis ............................................................................................................ 29 1.3.2 Necrosis .............................................................................................................. 33 1.3.3 Autophagy ........................................................................................................... 38 1.4 Cell death in myocardial infarction and heart failure ................................................... 39 1.4.1 Apoptosis in myocardial infarction and heart failure ........................................... 41 1.4.2 Necrosis in myocardial infarction and heart failure .............................................. 44 1.4.3 Autophagy in myocardial infarction and heart failure ........................................... 45 1.5 Metabolism ................................................................................................................ 49 1.6 Metabolism in myocardial infarction and heart failure ................................................ 51 1.7 The protein kinase complement of the human genome .............................................. 55 1.7.1 Classification and mode of action ........................................................................ 55 1.7.2 Structure ............................................................................................................. 58 1.7.3 Mitogen-activated protein kinases (MAPKs) ........................................................ 59 1.8 MAPKs in heart failure ............................................................................................... 63 1.8.1 MAP kinases in heart failure ................................................................................ 63 1.8.2 MAP3 kinases in heart failure .............................................................................. 67 1.8.3 MAP4K kinases in heart failure ........................................................................... 70 1.9 Induced pluripotent stem cells (iPSCs) as a tool for drug discovery ........................... 75 1.9.1 Characterisation of iPSC-derived cardiomyocytes ............................................... 77 1.9.2 Engineered heart tissue (EHT) as a platform for drug evaluation ........................ 78 2. The aim ....................................................................................................................... 81 2. Methods .............................................................................................................................. 84 6 2.1 hiPSC-CM culture ...................................................................................................... 85 2.2 MAP4K4 pharmacological inhibition ........................................................................... 86 2.3 Cell death induction ................................................................................................... 89 2.4 Cell death assays ...................................................................................................... 89 2.5 Immunocytostaining (2D) ........................................................................................... 90 2.6 Mitochondrial respiration analysis .............................................................................. 92 2.7 Protein quantification ................................................................................................. 93 2.8 hiPSC-CMs thawing for human engineered heart tissue (hEHTs) generation ............ 93 2.9 EHT generation ......................................................................................................... 94 2.10 EHT solutions .......................................................................................................... 94 2.11 Force calculation in EHTs and contractile assessment ............................................ 96 2.12 Immunocytochemistry .............................................................................................. 98 2.13 Calcium dynamics assessment ................................................................................ 99 2.14 Statistical Analysis ................................................................................................. 100 3. Results .............................................................................................................................. 101 Effects of MAP4K4 inhibition on cell death in 2-dimensional human iPSC-CM culture. ........ 102 3.1 Optimisation of conditions for human cardiac muscle cell death induced by H2O2 ... 104 3.2 Effect of the MAP4K4 inhibitor IC4-001 on plasma membrane disruption induced by H2O2 .............................................................................................................................. 106 3.3 Effect of the MAP4K4inhibitor IC4-001 on caspase-3 activation induced by H2O2 .... 108 3.4 Effect of the MAP4K4 inhibitor IC4-001 on BID cleavage induced by H2O2 .............. 111 3.5
Recommended publications
  • Characterization of a Novel MAP4K4-SASH1 Kinase Cascade Regulating Breast Cancer Tumorigenesis and Metastasis
    Characterization of a Novel MAP4K4-SASH1 Kinase Cascade Regulating Breast Cancer Tumorigenesis and Metastasis Yadong Li Guizhou Medical University Daoqiu Wu The Aliated Hospital of Guizhou Medical University Jing Hou Guizhou Provincial People's Hospital Jing Zhang The Aliated Hospital of Guizhou Medical University Xing Zeng Chongqing Medical University Lian Chen Guizhou Medical University Xin Wan Guizhou Medical University Zhixiong Wu Guizhou medical university Jinyun Wang Guizhou Medical University Ke Wang Yongchuan Hospital of Chongqing Dan Yang The Aliated Hospital of Guizhou Medical University Hongyu Chen Guizhou Medical University Zexi Xu Guizhou medical university Lei Jia Guizhou Medical University Qianfan Liu Guizhou medical university Zhongshu Kuang Page 1/30 Fudan university Geli Jiang Chongqing Cancer Hospital Hui Zhang Chongqing Zhongshan Hospital Jie Luo Chongqing Cancer Hospital Wei Li Chongqing Cancer Hospital Xue Zou The Aliated Hospital of Guizhou Medical University Xiaohua Zeng Chongqing Cancer Hospital Ding'an Zhou ( [email protected] ) Guizhou Medical University https://orcid.org/0000-0002-6614-9321 Research Keywords: SASH1, MAP4K4, Tumorigenesis, Metastasis, Hormone-dependent breast cancers Posted Date: November 5th, 2020 DOI: https://doi.org/10.21203/rs.3.rs-101160/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 2/30 Abstract Background: The SAM and SH3 domain containing protein 1(SASH1) was previously described as a candidate tumor-suppressor gene in breast cancer and colon cancer to mediate tumor metastasis and tumor growth. Howeverthe underlying mechanisms by which SASH1 implements breast cancer tumorigenesis and the question why SASH1 is downregulated in most solid cancers remain unexplored.
    [Show full text]
  • MAP4K Family Kinases Act in Parallel to MST1/2 to Activate LATS1/2 in the Hippo Pathway
    ARTICLE Received 19 Jun 2015 | Accepted 13 Aug 2015 | Published 5 Oct 2015 DOI: 10.1038/ncomms9357 OPEN MAP4K family kinases act in parallel to MST1/2 to activate LATS1/2 in the Hippo pathway Zhipeng Meng1, Toshiro Moroishi1, Violaine Mottier-Pavie2, Steven W. Plouffe1, Carsten G. Hansen1, Audrey W. Hong1, Hyun Woo Park1, Jung-Soon Mo1, Wenqi Lu1, Shicong Lu1, Fabian Flores1, Fa-Xing Yu3, Georg Halder2 & Kun-Liang Guan1 The Hippo pathway plays a central role in tissue homoeostasis, and its dysregulation contributes to tumorigenesis. Core components of the Hippo pathway include a kinase cascade of MST1/2 and LATS1/2 and the transcription co-activators YAP/TAZ. In response to stimulation, LATS1/2 phosphorylate and inhibit YAP/TAZ, the main effectors of the Hippo pathway. Accumulating evidence suggests that MST1/2 are not required for the regulation of YAP/TAZ. Here we show that deletion of LATS1/2 but not MST1/2 abolishes YAP/TAZ phosphorylation. We have identified MAP4K family members—Drosophila Happyhour homologues MAP4K1/2/3 and Misshapen homologues MAP4K4/6/7—as direct LATS1/2-activating kinases. Combined deletion of MAP4Ks and MST1/2, but neither alone, suppresses phosphorylation of LATS1/2 and YAP/TAZ in response to a wide range of signals. Our results demonstrate that MAP4Ks act in parallel to and are partially redundant with MST1/2 in the regulation of LATS1/2 and YAP/TAZ, and establish MAP4Ks as components of the expanded Hippo pathway. 1 Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, California 92093, USA.
    [Show full text]
  • 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]
  • The MAP4K4-STRIPAK Complex Promotes Growth and Tissue Invasion In
    bioRxiv preprint doi: https://doi.org/10.1101/2021.05.07.442906; this version posted May 8, 2021. 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. 1 The MAP4K4-STRIPAK complex promotes growth and tissue invasion in 2 medulloblastoma 3 Jessica Migliavacca1, Buket Züllig1, Charles Capdeville1, Michael Grotzer2 and Martin Baumgartner1,* 4 1 Division of Oncology, Children’s Research Center, University Children’s Hospital Zürich, Zürich, 5 Switzerland 6 2 Division of Oncology, University Children’s Hospital Zürich, Zürich, Switzerland 7 8 *e-mail: [email protected] 9 10 Abstract 11 Proliferation and motility are mutually exclusive biological processes associated with cancer that depend 12 on precise control of upstream signaling pathways with overlapping functionalities. We find that STRN3 13 and STRN4 scaffold subunits of the STRIPAK complex interact with MAP4K4 for pathway regulation in 14 medulloblastoma. Disruption of the MAP4K4-STRIPAK complex impairs growth factor-induced 15 migration and tissue invasion and stalls YAP/TAZ target gene expression and oncogenic growth. The 16 migration promoting functions of the MAP4K4-STRIPAK complex involve the activation of novel PKCs 17 and the phosphorylation of the membrane targeting S157 residue of VASP through MAP4K4. The anti- 18 proliferative effect of complex disruption is associated with reduced YAP/TAZ target gene expression 19 and results in repressed tumor growth in the brain tissue. This dichotomous functionality of the STRIPAK 20 complex in migration and proliferation control acts through MAP4K4 regulation in tumor cells and 21 provides relevant mechanistic insights into novel tumorigenic functions of the STRIPAK complex in 22 medulloblastoma.
    [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]
  • Application of a MYC Degradation
    SCIENCE SIGNALING | RESEARCH ARTICLE CANCER Copyright © 2019 The Authors, some rights reserved; Application of a MYC degradation screen identifies exclusive licensee American Association sensitivity to CDK9 inhibitors in KRAS-mutant for the Advancement of Science. No claim pancreatic cancer to original U.S. Devon R. Blake1, Angelina V. Vaseva2, Richard G. Hodge2, McKenzie P. Kline3, Thomas S. K. Gilbert1,4, Government Works Vikas Tyagi5, Daowei Huang5, Gabrielle C. Whiten5, Jacob E. Larson5, Xiaodong Wang2,5, Kenneth H. Pearce5, Laura E. Herring1,4, Lee M. Graves1,2,4, Stephen V. Frye2,5, Michael J. Emanuele1,2, Adrienne D. Cox1,2,6, Channing J. Der1,2* Stabilization of the MYC oncoprotein by KRAS signaling critically promotes the growth of pancreatic ductal adeno- carcinoma (PDAC). Thus, understanding how MYC protein stability is regulated may lead to effective therapies. Here, we used a previously developed, flow cytometry–based assay that screened a library of >800 protein kinase inhibitors and identified compounds that promoted either the stability or degradation of MYC in a KRAS-mutant PDAC cell line. We validated compounds that stabilized or destabilized MYC and then focused on one compound, Downloaded from UNC10112785, that induced the substantial loss of MYC protein in both two-dimensional (2D) and 3D cell cultures. We determined that this compound is a potent CDK9 inhibitor with a previously uncharacterized scaffold, caused MYC loss through both transcriptional and posttranslational mechanisms, and suppresses PDAC anchorage- dependent and anchorage-independent growth. We discovered that CDK9 enhanced MYC protein stability 62 through a previously unknown, KRAS-independent mechanism involving direct phosphorylation of MYC at Ser .
    [Show full text]
  • Transcriptome Analysis of Human Diabetic Kidney Disease
    ORIGINAL ARTICLE Transcriptome Analysis of Human Diabetic Kidney Disease Karolina I. Woroniecka,1 Ae Seo Deok Park,1 Davoud Mohtat,2 David B. Thomas,3 James M. Pullman,4 and Katalin Susztak1,5 OBJECTIVE—Diabetic kidney disease (DKD) is the single cases, mild and then moderate mesangial expansion can be leading cause of kidney failure in the U.S., for which a cure has observed. In general, diabetic kidney disease (DKD) is not yet been found. The aim of our study was to provide an considered a nonimmune-mediated degenerative disease unbiased catalog of gene-expression changes in human diabetic of the glomerulus; however, it has long been noted that kidney biopsy samples. complement and immunoglobulins sometimes can be de- — tected in diseased glomeruli, although their role and sig- RESEARCH DESIGN AND METHODS Affymetrix expression fi arrays were used to identify differentially regulated transcripts in ni cance is not clear (4). 44 microdissected human kidney samples. The DKD samples were The understanding of DKD has been challenged by multi- significant for their racial diversity and decreased glomerular ple issues. First, the diagnosis of DKD usually is made using filtration rate (~20–30 mL/min). Stringent statistical analysis, using clinical criteria, and kidney biopsy often is not performed. the Benjamini-Hochberg corrected two-tailed t test, was used to According to current clinical practice, the development of identify differentially expressed transcripts in control and diseased albuminuria in patients with diabetes is sufficient to make the glomeruli and tubuli. Two different Web-based algorithms were fi diagnosis of DKD (5). We do not understand the correlation used to de ne differentially regulated pathways.
    [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]
  • Non-Target Genes Regulate Mirnas-Mediated Migration Steering of Colorectal Carcinoma
    Pathology & Oncology Research (2019) 25:559–566 https://doi.org/10.1007/s12253-018-0502-9 ORIGINAL ARTICLE Non-target Genes Regulate miRNAs-Mediated Migration Steering of Colorectal Carcinoma Sohair M. Salem1 & Ahmed R. Hamed2,3 & Alaaeldin G. Fayez1 & Ghada Nour Eldeen1,4 Received: 20 January 2018 /Accepted: 15 October 2018 /Published online: 25 October 2018 # Arányi Lajos Foundation 2018 Abstract MicroRNAs (miRNAs) trigger a two-layer regulatory network directly or through transcription factors and their co-regulators. Unlike miR-375, the role of miR-145 and miR-224 in inhibiting or driving cancer cell migration is controversial. This study is a step towards addressing the potential of miR-375, miR-145 and miR-224 expression modulation to inhibit colorectal carcinoma (CRC) cells migration in vitro through regulation of non-target genes VEGFA, TGFβ1, IGF1, CD105 and CD44. Transwell migration assay results revealed a significant subdue of migration ability of cells transfected with miR-375 and miR-145 mimics and miR-224 inhibitor. Real time PCR data showed that expression of VEGFA, TGFβ1, IGF1, CD105 and CD44 was downreg- ulated as a consequence of exogenous re-expression of miR-375 and inhibition of miR-224. On the other hand, ectopic expression of miR-145 did not affect VEGFA, TGFβ1 and CD44 expression, while it elevated CD105 and suppressed IGF1 expression. MAP4K4, a predicted target of miR-145, was validated as a target that could play a role in miR-145-mediated regulation of migration. At mRNA level, no change was observed in expression of MAP4K4 in cells with restored expression of miR-145, while western blotting analysis revealed a 25% reduction of protein level.
    [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]