Repression of the Protein Kinase PIM3 by an Mtorc1-Regulated Microrna

Total Page:16

File Type:pdf, Size:1020Kb

Repression of the Protein Kinase PIM3 by an Mtorc1-Regulated Microrna Repression of the protein kinase PIM3 by an mTORC1-regulated microRNA The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:39987987 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA Repression of the protein kinase PIM3 by an mTORC1-regulated microRNA A dissertation presented by Ilana Ashley Kelsey to The Division of Medical Sciences in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the subject of Biological and Biomedical Sciences Harvard University Cambridge, Massachusetts August 2017 © 2017 Ilana Ashley Kelsey All rights reserved. Dissertation Advisor: Brendan Manning Ilana Ashley Kelsey Repression of the protein kinase PIM3 by an mTORC1-regulated microRNA Abstract The mechanistic target of rapamycin complex 1 (mTORC1) is a central regulator of cell growth that is often aberrantly activated in cancer. However, mTORC1 inhibitors, such as rapamycin, have limited effectiveness as single agent cancer therapies, with feedback mechanisms inherent to the signaling network thought to diminish the anti-tumor effects of mTORC1 inhibition. The goals of this dissertation were to characterize pro-survival effectors activated upon mTORC1 inhibition, and to determine the functional significance of these downstream targets, including relevance to the development of targeted therapies in combination with mTORC1 inhibitors. I identify the repression of protein kinase and proto-oncogene PIM3 downstream of mTORC1 signaling. PIM3 expression is suppressed in cells with loss of the tuberous sclerosis complex (TSC) tumor suppressors, which exhibit growth factor-independent activation of mTORC1, and in the mouse liver upon feeding-induced activation of mTORC1. Inhibition of mTORC1 with rapamycin induces PIM3 transcript and protein levels in a variety of settings. Suppression of PIM3 involves the sterol regulatory element-binding (SREBP) transcription factors SREBP1 and 2, whose processing and mRNA expression are stimulated by mTORC1 signaling. I found that PIM3 repression is mediated by miR-33, an intronic microRNA encoded within the SREBP loci, the expression of which is decreased with rapamycin. I sought to better understand the functional implications of miR-33 induction by mTORC1, and the subsequent induction of PIM3 upon mTORC1 inhibition. Specifically, I show that PIM inhibition in combination with mTOR inhibitors may be a promising therapy in some cancer settings. I also identify several additional mTORC1-regulated miR-33 targets that contribute to cell survival and metabolism, iii including PIM1, which is closely related to PIM3. Finally, I explore the metabolic changes affected by PIM inhibition, providing an additional rationale for the regulation of PIM3 by mTORC1. Collectively, these studies identify a pro-survival kinase that is activated upon mTORC1 inhibition while highlighting the importance of further characterization of miR-33 targets altered downstream of mTORC1. Our results will guide future studies of mTORC1-regulated microRNAs and pro-survival pathways, with potential implications for the effects of mTORC1 inhibitors in TSC, cancer, and the many other disease settings influenced by aberrant mTORC1 signaling. iv TABLE OF CONTENTS ABSTRACT iii LIST OF FIGURES viii GLOSSARY OF TERMS x DEDICATION xv ACKNOWLEDGEMENTS xvi CHAPTER 1: INTRODUCTION 1 1.1 mTORC1 is a major regulator of cell growth and metabolism 1.1.1 Overview 1.1.2 Upstream regulation of mTORC1 1.1.3 Downstream processes regulated by mTORC1 1.1.4 mTORC1 control of coding and non-coding RNA expression 1.1.5 mTORC1 signaling in disease 1.2 The PIM kinase family in signaling and disease 1.2.1 Overview 1.2.2 Upstream regulation of PIM kinases 1.2.3 Downstream targets of PIM kinases 1.2.4 PIM kinases in cancer 1.3 Specific Aims and Overview of the Dissertation 1.4 References v CHAPTER 2: mTORC1 SUPPRESSES PIM3 EXPRESSION VIA miR-33 ENCODED BY THE SREBP LOCI 50 2.1 Abstract 2.2 Introduction 2.3 Materials and Methods 2.4 Results 2.4.1 PIM3 expression is repressed downstream of mTORC1 and induced by mTOR inhibitors 2.4.2 A survey of mTORC1-regulated transcription factors identifies SREBP1 and 2 as upstream of PIM3 2.4.3 miR-33, an intronic microRNA within the SREBP loci, targets PIM3 downstream of mTORC1 2.5 Discussion 2.6 Acknowledgements 2.7 Author Contributions 2.8 References CHAPTER 3: BIOLOGICAL SIGNIFICANCE OF miR-33 INDUCTION AND PIM REPRESSION BY mTORC1 75 3.1 Abstract 3.2 Introduction vi 3.3 Materials and Methods 3.4 Results 3.4.1 miR-33-targeted transcripts are changing downstream of mTORC1 3.4.2 Additive effects of dual inhibition of mTORC1 and PIM kinases in Tsc2-/- MEFs 3.4.3 Implications for PIM effects on metabolism, particularly cellular NAD+ levels 3.5 Discussion 3.6 Acknowledgements 3.7 References CHAPTER 4: CONCLUSIONS 102 4.1 Overview 4.2 Challenges of targeting the mTORC1 signaling axis in disease 4.2.1 Overview 4.2.2 Crosstalk and feedback pathways contribute to rewiring of the signaling network in response to specific inhibitors 4.2.3 Redundancies in the greater mTORC1 network enable sustained signaling 4.2.4 Resistance to targeted therapies is intrinsic to the wiring of the network 4.3 Future directions 4.4 References vii LIST OF FIGURES FIGURE 1.1 – Regulation of mTORC1 by a variety of upstream inputs 4 FIGURE 1.2 – Amino acid sensing and mTORC1 activation at the lysosome 7 FIGURE 1.3 – mTORC1 stimulates anabolic pathways to support cell growth and proliferation 9 FIGURE 1.4 – mTORC1 controls a vast transcriptional program in order to regulate anabolic 13 processes and stimulate cell growth FIGURE 1.5 – microRNA regulation downstream of mTORC1 16 FIGURE 1.6 – A wide network of transcription factors regulates the expression of the PIM kinases 20 FIGURE 1.7 – The PIM kinases promote cell survival through a variety of pathways 24 FIGURE 1.8 – The PIM kinases stimulate Myc expression and transcription at multiple steps 27 FIGURE 2.1 – PIM3 is repressed downstream of mTORC1 57 FIGURE 2.2 – PIM3 repression by mTORC1 is observed in a variety of human cancer settings 59 FIGURE 2.3 – Identification of the mTORC1 effectors SREBP1 and 2 as being upstream of 61 PIM3 regulation FIGURE 2.4 – PIM3 is induced upon inhibition of SREBP1 and 2 63 FIGURE 2.5 – An SREBP-intronic microRNA, miR-33, targets PIM3 expression downstream 64 of mTORC1 FIGURE 3.1 – miR-33 targets are induced by rapamycin 81 FIGURE 3.2 – Combined inhibition of PIM3 and mTORC1 has an additive effect in MEFs 83 FIGURE 3.3 – Metabolomic profiling in high/low PIM3 settings reveals changes in NAD+ levels 86 and synthesis upon PIM inhibition FIGURE 3.4 – Supporting metabolomics data for Figure 3.3 88 viii FIGURE 4.1 – Graphical summary of the dissertation 104 FIGURE 4.2 – Feedback pathways in the mTORC1 signaling network 106 FIGURE 4.3 – Pathway convergence on shared effectors 110 ix GLOSSARY OF TERMS 25-HC 25-hydroxycholesterol 4E-BP eIF4E binding proteins ABCA1 ATP-binding cassette transporter A1 AMP adenosine monophosphate AMPK AMP kinase ATF4 activating transcription factor 4 ASK1 apoptosis signaling kinase 1 ATG7 autophagy-related 7 C/EBP-α CCAAT/enhancer-binding protein-α CAD carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, dihydroorotase CDC25A/C cell division cycle 25 homolog A and C C-TAK1 Cdc25 C-associated kinase 1 DEPTOR DEP domain-containing mTOR-interacting protein eEF2K eukaryotic elongation factor 2 kinase EGF epidermal growth factor EGFR EGF receptor eIF4B eukaryotic translation initiation factor 4B eIF4E eukaryotic translation initiation factor 4E ER endoplasmic reticulum ERK/MAPK mitogen-activated protein kinase FAS fatty acid synthase FBS fetal bovine serum FKBP12 FK506 binding protein of 12 kDa FOXO forkhead box O x GAB1/2 GRB2-associated binder 1 and 2 GAP GTPase-activating protein GEF guanine nucleotide exchange GRB10 growth factor receptor bound protein 10 GSK3 glycogen synthase kinase 3 HCC hepatocellular carcinoma HIF-1α hypoxia-inducible factor alpha HK2 hexokinase 2 HOXA9 homeobox protein A9 HSP90 heat shock protein 90β IGF1 insulin-like growth factor 1 IR insulin receptor IRS-1/2 insulin receptor substrates 1 and 2 INSIG insulin-induced gene JNK c-Jun N-terminal kinase KLF5 Kruppel-like factor 5 LAM lymphangioleiomyomatosis LC-MS/MS liquid chromatography-mass spectrometry/mass spectrometry Lipin1 phosphatidic acid phosphatase LPIN1 MAX Myc-associated factor X MEF mouse embryonic fibroblast MEK MAPK kinase mLST8 mTOR-associated protein, LST8 homologue mRNA messenger RNA MTHFD2 methylene tetrahydrofolate dehydrogenase 2 mTOR mechanistic target of rapamycin xi mTORC1 mTOR complex 1 mTORC2 mTOR complex 2 MuLV Moloney murine leukemia virus NADH nicotinamide adenine dinucleotide nCoR1 nuclear receptor co-repressor 1 NF1 neurofibromin 1 NF-Κb nuclear factor- Κb Nrf1 nuclear factor, erythroid 2-like 1 p21Cip1/WAF1 cyclin dependent kinase inhibitor 1A p27KIP1 cyclin dependent kinase inhibitor 1B PA phosphatidic acid Pax-5 paired box gene 5 PDCD4 programmed cell death 4 PDGF platelet-derived growth factor PDGFR PDGF receptor PDK1 phosphinositide-dependent kinase-1 PGC1α peroxisome proliferator-activated receptor gamma coactivator-1 PI propidium iodide PI3K phosphoinositide 3-kinase PIM proviral
Recommended publications
  • Identification of PIM1 Substrates Reveals a Role for NDRG1
    ARTICLE https://doi.org/10.1038/s42003-020-01528-6 OPEN Identification of PIM1 substrates reveals a role for NDRG1 phosphorylation in prostate cancer cellular migration and invasion Russell J. Ledet1,2,3,5, Sophie E. Ruff1,2,3,5, Yu Wang1,2, Shruti Nayak4, Jeffrey A. Schneider1,2,3, ✉ ✉ 1234567890():,; Beatrix Ueberheide1,4, Susan K. Logan1,2 & Michael J. Garabedian 2,3 PIM1 is a serine/threonine kinase that promotes and maintains prostate tumorigenesis. While PIM1 protein levels are elevated in prostate cancer relative to local disease, the mechanisms by which PIM1 contributes to oncogenesis have not been fully elucidated. Here, we performed a direct, unbiased chemical genetic screen to identify PIM1 substrates in prostate cancer cells. The PIM1 substrates we identified were involved in a variety of oncogenic processes, and included N-Myc Downstream-Regulated Gene 1 (NDRG1), which has reported roles in sup- pressing cancer cell invasion and metastasis. NDRG1 is phosphorylated by PIM1 at serine 330 (pS330), and the level of NDRG1 pS330 is associated higher grade prostate tumors. We have shown that PIM1 phosphorylation of NDRG1 at S330 reduced its stability, nuclear localization, and interaction with AR, resulting in enhanced cell migration and invasion. 1 Departments of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA. 2 Department of Urology, New York University School of Medicine, New York, NY 10016, USA. 3 Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA. 4 Proteomics Laboratory, New York University School of Medicine, New York, NY 10016, USA.
    [Show full text]
  • The Legionella Kinase Legk7 Exploits the Hippo Pathway Scaffold Protein MOB1A for Allostery and Substrate Phosphorylation
    The Legionella kinase LegK7 exploits the Hippo pathway scaffold protein MOB1A for allostery and substrate phosphorylation Pei-Chung Leea,b,1, Ksenia Beyrakhovac,1, Caishuang Xuc, Michal T. Bonieckic, Mitchell H. Leea, Chisom J. Onub, Andrey M. Grishinc, Matthias P. Machnera,2, and Miroslaw Cyglerc,2 aDivision of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892; bDepartment of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, MI 48202; and cDepartment of Biochemistry, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada Edited by Ralph R. Isberg, Tufts University School of Medicine, Boston, MA, and approved May 1, 2020 (received for review January 12, 2020) During infection, the bacterial pathogen Legionella pneumophila Active LATS1/2 phosphorylate the cotranscriptional regulator manipulates a variety of host cell signaling pathways, including YAP1 (yes-associated protein 1) and its homolog TAZ (tran- the Hippo pathway which controls cell proliferation and differen- scriptional coactivator with PDZ-binding motif). Phosphorylated tiation in eukaryotes. Our previous studies revealed that L. pneu- YAP1 and TAZ are prevented from entering the nucleus by being mophila encodes the effector kinase LegK7 which phosphorylates either sequestered in the cytosol through binding to 14-3-3 pro- MOB1A, a highly conserved scaffold protein of the Hippo path- teins or targeted for proteolytic degradation (6, 8). Consequently, way. Here, we show that MOB1A, in addition to being a substrate the main outcome of signal transduction along the Hippo pathway of LegK7, also functions as an allosteric activator of its kinase is changes in gene expression (6).
    [Show full text]
  • Pim1 Inhibition As a Novel Therapeutic Strategy for Alzheimer's Disease
    Velazquez et al. Molecular Neurodegeneration (2016) 11:52 DOI 10.1186/s13024-016-0118-z RESEARCH ARTICLE Open Access Pim1 inhibition as a novel therapeutic strategy for Alzheimer’s disease Ramon Velazquez1, Darren M. Shaw1,2, Antonella Caccamo1 and Salvatore Oddo1,2* Abstract Background: Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder worldwide. Clinically, AD is characterized by impairments of memory and cognitive functions. Accumulation of amyloid-β (Aβ) and neurofibrillary tangles are the prominent neuropathologies in patients with AD. Strong evidence indicates that an imbalance between production and degradation of key proteins contributes to the pathogenesis of AD. The mammalian target of rapamycin (mTOR) plays a key role in maintaining protein homeostasis as it regulates both protein synthesis and degradation. A key regulator of mTOR activity is the proline-rich AKT substrate 40 kDa (PRAS40), which directly binds to mTOR and reduces its activity. Notably, AD patients have elevated levels of phosphorylated PRAS40, which correlate with Aβ and tau pathologies as well as cognitive deficits. Physiologically, PRAS40 phosphorylation is regulated by Pim1, a protein kinase of the protoconcogene family. Here, we tested the effects of a selective Pim1 inhibitor (Pim1i), on spatial reference and working memory and AD-like pathology in 3xTg-AD mice. Results: We have identified a Pim1i that crosses the blood brain barrier and reduces PRAS40 phosphorylation. Pim1i-treated 3xTg-AD mice performed significantly better than their vehicle treated counterparts as well as non-transgenic mice. Additionally, 3xTg-AD Pim1i-treated mice showed a reduction in soluble and insoluble Aβ40 and Aβ42 levels, as well as a 45.2 % reduction in Aβ42 plaques within the hippocampus.
    [Show full text]
  • Comprehensive Identification of Proteins in Hodgkin Lymphoma
    Laboratory Investigation (2007) 87, 1113–1124 & 2007 USCAP, Inc All rights reserved 0023-6837/07 $30.00 Comprehensive identification of proteins in Hodgkin lymphoma-derived Reed–Sternberg cells by LC-MS/MS Jeremy C Wallentine1, Ki Kwon Kim1, Charles E Seiler III1, Cecily P Vaughn2, David K Crockett2, Sheryl R Tripp2, Kojo SJ Elenitoba-Johnson1,2 and Megan S Lim1,2 Mass spectrometry-based proteomics in conjunction with liquid chromatography and bioinformatics analysis provides a highly sensitive and high-throughput approach for the identification of proteins. Hodgkin lymphoma is a form of malignant lymphoma characterized by the proliferation of Reed–Sternberg cells and background reactive lymphocytes. Comprehensive analysis of proteins expressed and released by Reed–Sternberg cells would assist in the discovery of potential biomarkers and improve our understanding of its pathogenesis. The subcellular proteome of the three cellular compartments from L428 and KMH2 Hodgkin lymphoma-derived cell lines were fractionated, and analyzed by reverse- phase liquid chromatography coupled with electrospray ionization tandem mass spectrometry. Additionally, proteins released by Hodgkin lymphoma-derived L428 cells were extracted from serum-free culture media and analyzed. Peptide spectra were analyzed using TurboSEQUESTs against the UniProt protein database (5.26.05; 188 712 entries). A subset of the identified proteins was validated by Western blot analysis, immunofluorescence microscopy and im- munohistochemistry. A total of 1945 proteins were identified with 785 from the cytosolic fraction, 305 from the membrane fraction, 441 from the nuclear fraction and 414 released proteins using a minimum of two peptide identi- fications per protein and an error rate of o5.0%.
    [Show full text]
  • Rnai Screen Identifies a Synthetic Lethal Interaction Between PIM1 Overexpression and PLK1 Inhibition
    Published OnlineFirst April 25, 2014; DOI: 10.1158/1078-0432.CCR-13-3116 Clinical Cancer Cancer Therapy: Preclinical Research RNAi Screen Identifies a Synthetic Lethal Interaction between PIM1 Overexpression and PLK1 Inhibition Riet van der Meer1, Ha Yong Song2, Seong-Hoon Park2, Sarki A. Abdulkadir1, and Meejeon Roh2 Abstract Purpose: To identify genes whose depletion is detrimental to Pim1-overexpressing prostate cancer cells and to validate this finding in vitro and in vivo. Experimental Design: RNAi screening was used to identify genes whose depletion is detrimental to Pim1-overexpressing cells. Our finding was validated using shRNA or PLK1-specific inhibitor BI 2536. Xenograft studies were performed using both PLK1-knockdown cells and BI 2536 to investigate the effects of PLK1 inhibition on tumorigenesis in Pim1-overexpressing cells. Finally, PLK1 and PIM1 expression patterns in human prostate tumors were examined by immunohistochemistry using tissue microarrays. Results: We identified the mitotic regulator polo-like kinase (PLK1) as a gene whose depletion is particularly detrimental to the viability of Pim1-overexpressing prostate cancer. Inhibition of PLK1 by shRNA or BI 2536 in Pim1-overexpressing prostate cancer xenograft models resulted in a dramatic inhibition of tumor progression. Notably, Pim1-overexpressing cells were more prone to mitotic arrest followed by apoptosis due to PLK1 inhibition than control cells. Furthermore, inhibition of PLK1 led to the reduction of MYC protein levels both in vitro and in vivo. Our data also suggest that PIM1 and PLK1 physically interact and PIM1 might phosphorylate PLK1. Finally, PLK1 and PIM1 are frequently co- expressed in human prostate tumors, and co-expression of PLK1 and PIM1 was significantly correlated to higher Gleason grades.
    [Show full text]
  • 1 Identification of PIM1 Substrates Reveals a Role for NDRG1
    bioRxiv preprint doi: https://doi.org/10.1101/2020.01.21.913962; this version posted January 22, 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. Identification of PIM1 substrates reveals a role for NDRG1 in prostate cancer cellular migration and invasion Russell J. Ledet1,2,3#, Sophie Ruff1,2,3#, Yu Wang1,2, Shruti Nayak4, Jeffrey A. Schneider1,2,3, Beatrix Ueberheide1,4, Susan K. Logan1,2*, Michael J. Garabedian2,3* 1Departments of Biochemistry and Molecular Pharmacology, 2Urology, and 3Microbiology, New York University School of Medicine, New York, NY 10016, USA. 4Proteomics Laboratory, New York University School of Medicine, New York, NY 10016, USA. # contributed equally to the work *Corresponding authors: Michael J. Garabedian, PhD Departments of Microbiology and Urology New York University School of Medicine Alexandria Center for Life Sciences 450 East 29th Street, Room 324 New York, NY 10016 Tel.; (212) 263-7662 Fax: (646) 501-4643 Email: [email protected] Susan K. Logan, PhD Departments of Urology, and Biochemistry and Molecular Pharmacology New York University School of Medicine Alexandria Center for Life Sciences 450 East 29th Street, Room 323 New York, NY 10016 Tel.; (212) 263-2921 Fax: (646) 501-4643 Email: [email protected] Keywords: Prostate Cancer, NDRG1, PIM1, phosphoproteomics, chemical genetics screen, protein phosphorylation 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.01.21.913962; this version posted January 22, 2020.
    [Show full text]
  • PIM1-Dependent Phosphorylation of Histone H3 at Serine 10 Is Required for MYC-Dependent Transcriptional Activation and Oncogenic Transformation
    ARTICLES PIM1-dependent phosphorylation of histone H3 at serine 10 is required for MYC-dependent transcriptional activation and oncogenic transformation Alessio Zippo1, Alessandra De Robertis1, Riccardo Serafini1 and Salvatore Oliviero1,2 The serine/threonine kinase human Pim1 (hereafter PIM1) cooperates with human c-Myc (hereafter MYC) in cell cycle progression and tumorigenesis. However, the nature of this cooperation is still unknown. Here we show that, after stimulation with growth factor, PIM1 forms a complex with the dimer of MYC with MAX (Myc-associated factor X) via the MYC BoxII (MBII) domain. MYC recruits PIM1 to the E boxes of the MYC-target genes FOSL1 (FRA-1) and ID2, and PIM1 phosphorylates serine 10 of histone H3 (H3S10) on the nucleosome at the MYC-binding sites, contributing to their transcriptional activation. MYC and PIM1 colocalize at sites of active transcription, and expression profile analysis revealed that PIM1 contributes to the regulation of 20% of the MYC-regulated genes. Moreover, PIM1-dependent H3S10 phosphorylation contributes to MYC transforming capacity. These results establish a new function for PIM1 as a MYC cofactor that phosphorylates the chromatin at MYC-target loci and suggest that nucleosome phosphorylation, at E boxes, contributes to MYC-dependent transcriptional activation and cellular transformation. Chromatin structure has a crucial function in eukaryotic gene that Pim1 is overexpressed in Myc-driven prostate tumours16. However, transcription. Several transcription activators have been shown the nature of the cooperation of c-Myc and Pim1 in cell transformation to recruit chromatin-modifying activities, which mediate either is still unknown. covalent modifications of histones or ATP-dependent mobiliza- MYC codes for a basic helix–loop–helix leucine zipper transcription tion of nucleosomes.
    [Show full text]
  • Characterization of a Protein Kinase B Inhibitor in Vitro and in Insulin-Treated Liver Cells Lisa Logie,1 Antonio J
    Original Article Characterization of a Protein Kinase B Inhibitor In Vitro and in Insulin-Treated Liver Cells Lisa Logie,1 Antonio J. Ruiz-Alcaraz,1 Michael Keane,2,3 Yvonne L. Woods,2 Jennifer Bain,2 Rudolfo Marquez,3 Dario R. Alessi,1 and Calum Sutherland1 OBJECTIVE—Abnormal expression of the hepatic gluconeo- genic genes (glucose-6-phosphatase [G6Pase] and PEPCK) con- tributes to hyperglycemia. These genes are repressed by insulin, rotein kinase B (PKB) is a member of the AGC but this process is defective in diabetic subjects. Protein kinase family of protein kinases (1–3). In mammals, B (PKB) is implicated in this action of insulin. An inhibitor of there are three isoforms (PKB␣, PKB␤, and PKB, Akt inhibitor (Akti)-1/2, was recently reported; however, PPKB␥) (1). PKB is activated following induction the specificity and efficacy against insulin-induced PKB was not of phosphatidylinositol 3 (PI3) kinase activity and the reported. Our aim was to characterize the specificity and efficacy resultant generation of the lipid second messengers PI of Akti-1/2 in cells exposed to insulin and then establish whether 3,4,5 trisphosphate and PI 3,4 bisphosphate (4). These inhibition of PKB is sufficient to prevent regulation of hepatic lipids bind to the PH domain of PKB, altering its confor- gene expression by insulin. mation and permitting access to upstream protein kinases RESEARCH DESIGN AND METHODS—Akti-1/2 was assayed (5). Phosphoinositide-dependent protein kinase-1 phos- against 70 kinases in vitro and its ability to block PKB activation phorylates PKB at Thr308 (6), and a second phosphoryla- in cells exposed to insulin fully characterized.
    [Show full text]
  • Exploration of CTCF Post-Translation Modifications Uncovers Serine-224 Phosphorylation by PLK1 at Pericentric Regions During
    TOOLS AND RESOURCES Exploration of CTCF post-translation modifications uncovers Serine-224 phosphorylation by PLK1 at pericentric regions during the G2/M transition Brian C Del Rosario1,2†, Andrea J Kriz1,2†, Amanda M Del Rosario3, Anthony Anselmo4, Christopher J Fry5, Forest M White3, Ruslan I Sadreyev4, Jeannie T Lee1,2* 1Department of Molecular Biology, Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, United States; 2Department of Genetics, Harvard Medical School, Boston, United States; 3Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States; 4Department of Molecular Biology, Massachusetts General Hospital, Boston, United States; 5Cell Signaling Technology, Danvers, United States Abstract The zinc finger CCCTC-binding protein (CTCF) carries out many functions in the cell. Although previous studies sought to explain CTCF multivalency based on sequence composition of binding sites, few examined how CTCF post-translational modification (PTM) could contribute to function. Here, we performed CTCF mass spectrometry, identified a novel phosphorylation site at Serine 224 (Ser224-P), and demonstrate that phosphorylation is carried out by Polo-like kinase 1 (PLK1). CTCF Ser224-P is chromatin-associated, mapping to at least a subset of known CTCF sites. CTCF Ser224-P accumulates during the G2/M transition of the cell cycle and is enriched at pericentric regions. The phospho-obviation mutant, S224A, appeared normal. However, the *For correspondence: phospho-mimic mutant, S224E, is detrimental to mouse embryonic stem cell colonies. While ploidy [email protected] and chromatin architecture appear unaffected, S224E mutants differentially express hundreds of †These authors contributed genes, including p53 and p21. We have thus identified a new CTCF PTM and provided evidence of equally to this work biological function.
    [Show full text]
  • PIM1 Promotes Survival of Cardiomyocytes by Upregulating C-Kit Protein Expression
    cells Article PIM1 Promotes Survival of Cardiomyocytes by Upregulating c-Kit Protein Expression David E. Ebeid y, Fareheh Firouzi y , Carolina Y. Esquer, Julian M. Navarrete, Bingyan J. Wang , Natalie A. Gude and Mark A. Sussman * Department of Biology, San Diego State University, San Diego, CA 92182, USA; [email protected] (D.E.E.); ffi[email protected] (F.F.); [email protected] (C.Y.E.); [email protected] (J.M.N.); [email protected] (B.J.W.); [email protected] (N.A.G.) * Correspondence: [email protected]; Tel.: +1-(619)-594-2983 These authors contributed equally to this work. y Received: 10 July 2020; Accepted: 27 August 2020; Published: 31 August 2020 Abstract: Enhancing cardiomyocyte survival is crucial to blunt deterioration of myocardial structure and function following pathological damage. PIM1 (Proviral Insertion site in Murine leukemia virus (PIM) kinase 1) is a cardioprotective serine threonine kinase that promotes cardiomyocyte survival and antagonizes senescence through multiple concurrent molecular signaling cascades. In hematopoietic stem cells, PIM1 interacts with the receptor tyrosine kinase c-Kit upstream of the ERK (Extracellular signal-Regulated Kinase) and Akt signaling pathways involved in cell proliferation and survival. The relationship between PIM1 and c-Kit activity has not been explored in the myocardial context. This study delineates the interaction between PIM1 and c-Kit leading to enhanced protection of cardiomyocytes from stress. Elevated c-Kit expression is induced in isolated cardiomyocytes from mice with cardiac-specific overexpression of PIM1. Co-immunoprecipitation and proximity ligation assay reveal protein–protein interaction between PIM1 and c-Kit. Following treatment with Stem Cell Factor, PIM1-overexpressing cardiomyocytes display elevated ERK activity consistent with c-Kit receptor activation.
    [Show full text]
  • Inhibition of ERK 1/2 Kinases Prevents Tendon Matrix Breakdown Ulrich Blache1,2,3, Stefania L
    www.nature.com/scientificreports OPEN Inhibition of ERK 1/2 kinases prevents tendon matrix breakdown Ulrich Blache1,2,3, Stefania L. Wunderli1,2,3, Amro A. Hussien1,2, Tino Stauber1,2, Gabriel Flückiger1,2, Maja Bollhalder1,2, Barbara Niederöst1,2, Sandro F. Fucentese1 & Jess G. Snedeker1,2* Tendon extracellular matrix (ECM) mechanical unloading results in tissue degradation and breakdown, with niche-dependent cellular stress directing proteolytic degradation of tendon. Here, we show that the extracellular-signal regulated kinase (ERK) pathway is central in tendon degradation of load-deprived tissue explants. We show that ERK 1/2 are highly phosphorylated in mechanically unloaded tendon fascicles in a vascular niche-dependent manner. Pharmacological inhibition of ERK 1/2 abolishes the induction of ECM catabolic gene expression (MMPs) and fully prevents loss of mechanical properties. Moreover, ERK 1/2 inhibition in unloaded tendon fascicles suppresses features of pathological tissue remodeling such as collagen type 3 matrix switch and the induction of the pro-fbrotic cytokine interleukin 11. This work demonstrates ERK signaling as a central checkpoint to trigger tendon matrix degradation and remodeling using load-deprived tissue explants. Tendon is a musculoskeletal tissue that transmits muscle force to bone. To accomplish its biomechanical function, tendon tissues adopt a specialized extracellular matrix (ECM) structure1. Te load-bearing tendon compart- ment consists of highly aligned collagen-rich fascicles that are interspersed with tendon stromal cells. Tendon is a mechanosensitive tissue whereby physiological mechanical loading is vital for maintaining tendon archi- tecture and homeostasis2. Mechanical unloading of the tissue, for instance following tendon rupture or more localized micro trauma, leads to proteolytic breakdown of the tissue with severe deterioration of both structural and mechanical properties3–5.
    [Show full text]
  • PRODUCTS and SERVICES Target List
    PRODUCTS AND SERVICES Target list Kinase Products P.1-11 Kinase Products Biochemical Assays P.12 "QuickScout Screening Assist™ Kits" Kinase Protein Assay Kits P.13 "QuickScout Custom Profiling & Panel Profiling Series" Targets P.14 "QuickScout Custom Profiling Series" Preincubation Targets Cell-Based Assays P.15 NanoBRET™ TE Intracellular Kinase Cell-Based Assay Service Targets P.16 Tyrosine Kinase Ba/F3 Cell-Based Assay Service Targets P.17 Kinase HEK293 Cell-Based Assay Service ~ClariCELL™ ~ Targets P.18 Detection of Protein-Protein Interactions ~ProbeX™~ Stable Cell Lines Crystallization Services P.19 FastLane™ Structures ~Premium~ P.20-21 FastLane™ Structures ~Standard~ Kinase Products For details of products, please see "PRODUCTS AND SERVICES" on page 1~3. Tyrosine Kinases Note: Please contact us for availability or further information. Information may be changed without notice. Expression Protein Kinase Tag Carna Product Name Catalog No. Construct Sequence Accession Number Tag Location System HIS ABL(ABL1) 08-001 Full-length 2-1130 NP_005148.2 N-terminal His Insect (sf21) ABL(ABL1) BTN BTN-ABL(ABL1) 08-401-20N Full-length 2-1130 NP_005148.2 N-terminal DYKDDDDK Insect (sf21) ABL(ABL1) [E255K] HIS ABL(ABL1)[E255K] 08-094 Full-length 2-1130 NP_005148.2 N-terminal His Insect (sf21) HIS ABL(ABL1)[T315I] 08-093 Full-length 2-1130 NP_005148.2 N-terminal His Insect (sf21) ABL(ABL1) [T315I] BTN BTN-ABL(ABL1)[T315I] 08-493-20N Full-length 2-1130 NP_005148.2 N-terminal DYKDDDDK Insect (sf21) ACK(TNK2) GST ACK(TNK2) 08-196 Catalytic domain
    [Show full text]