In Vivo Antitumor Activity of MEK and Phosphatidylinositol 3-Kinase Inhibitors in Basal-Like Breast Cancer Models Klaus P

Total Page:16

File Type:pdf, Size:1020Kb

In Vivo Antitumor Activity of MEK and Phosphatidylinositol 3-Kinase Inhibitors in Basal-Like Breast Cancer Models Klaus P Cancer Therapy: Preclinical In vivo Antitumor Activity of MEK and Phosphatidylinositol 3-Kinase Inhibitors in Basal-Like Breast Cancer Models Klaus P. Hoeflich,2 Carol O’Brien,1Zachary Boyd,1Guy Cavet,3 Steve Guerrero,3 Kenneth Jung,3 To m Ja n u a r i o , 1 Heidi Savage,1Elizabeth Punnoose,1Tom Truong,2 Wei Zhou,2 Leanne Berry,2 Lesley Murray,2 Lukas Amler,1Marcia Belvin,4 Lori S. Friedman,4 and Mark R. Lackner1 Abstract Purpose: The pathways underlying basal-like breast cancer are poorly understood, and as yet, there is no approved targeted therapy for this disease.We investigated the role of mitogen- activated protein kinase kinase (MEK) and phosphatidylinositol 3-kinase (PI3K) inhibitors as targeted therapies for basal-like breast cancer. Experimental Design: We used pharmacogenomic analysis of a large panel of breast cancer cell lines with detailed accompanying molecular information to identify molecular predictors of response to a potent and selective inhibitor of MEK and also to define molecular mechanisms underlying combined MEK and PI3K targeting in basal-like breast cancer. Hypotheses were confirmed by testing in multiple tumor xenograft models. Results:We found that basal-like breast cancer models have an activated RAS-like transcriptional program and show greater sensitivity to a selective inhibitor of MEK compared with models rep- resentative of other breast cancer subtypes.We also showed that loss of PTEN is a negative pre- dictor of response to MEK inhibition, that treatment with a selective MEK inhibitor caused up-regulation of PI3K pathway signaling, and that dual blockade of both PI3K and MEK/extracel- lular signal ^ regulated kinase signaling synergized to potently impair the growth of basal-like breast cancer models in vitro and in vivo. Conclusions: Our studies suggest that single-agent MEK inhibition is a promising therapeutic modality for basal-like breast cancers with intact PTEN, and also provide a basis for rational combination of MEK and PI3K inhibitors in basal-like cancers with both intact and deleted PTEN. Recent molecular profiling studies have suggested that breast poor prognosis if untreated, and significant clinical benefit cancers represent at least three distinct diseases with a common from the HER2 targeting monoclonal antibody trastuzumab tissue of origin but different underlying biological aberrations. (Herceptin, Genentech; ref. 3). Basal-like breast cancers Gene expression studies have shown that primary breast tumors typically lack expression of HER2, ER, and progesterone may be classified into at least three major subtypes by receptor and hence overlap with tumors immunohistochemi- unsupervised hierarchical clustering analysis and that the cally defined as ‘‘triple-negative’’ tumors (4). Basal-like breast subtypes have different prognostic outcomes in terms of patient cancers have a poor prognosis and currently have not been survival (1). Luminal breast cancers are typically estrogen shown to respond to any targeted therapy and thus represent a receptor (ER) positive and characterized by coordinate expres- particularly pressing unmet medical need (5). Interestingly, the sion of a number of epithelial specific genes, a relatively good majority of basal-like breast cancers express high levels of the prognosis, and good response rates to targeted hormonal epidermal growth factor receptor (EGFR) and have an EGFR- therapies (2). HER2-positive breast cancers are characterized by associated gene expression signature (6). Because growth HER2 high-level gene amplification of the oncogene, relatively factor stimulation of EGFR results in potent activation of the signaling pathway composed of RAS, RAF, mitogen-activated protein kinase (MAPK) kinase (MEK), and extracellular Authors’ Affiliations: 1Development Oncology Diagnostics, 2Translational 3 4 signal–regulated kinase (ERK; ref. 7), we sought to determine Oncology, Bioinformatics, and Cancer Signaling Pathways, Genentech, Inc., South San Francisco, California whether the downstream arm of this pathway might be a target Received2/9/09;revised3/19/09;accepted3/24/09;publishedOnlineFirst6/30/09. of therapeutic intervention in breast cancer. The costs of publication of this article were defrayed in part by the payment of page The protein kinase MEK occupies a crucial signaling node advertisement charges. This article must therefore be hereby marked in accordance downstream of RAS and RAF and directly upstream of ERK and with 18 U.S.C. Section 1734 solely to indicate this fact. Note: Supplementary data for this article are available at Clinical Cancer Research as such has been the subject of intensive drug discovery Online (http://clincancerres.aacrjournals.org/). activities (8). Genetic and biochemical analyses of MEK K. Hoeflich, C. O’Brien, and Z. Boyd contributed equally to this work. function have suggested that MEK activity is necessary for the Requests for reprints: Mark R. Lackner, Genentech, Inc., 1DNA Way, South San transforming and proliferative effects of this pathway, suggest- Francisco, CA 94080. Phone: 650-467-1846; Fax: 650-225-7571; E-mail: [email protected]. ing that therapeutics that completely inhibit MEK function may F 2009 American Association for Cancer Research. have utility in the treatment of cancers driven by activation of doi:10.1158/1078-0432.CCR-09-0317 the RAS/RAF axis. Such efforts have led to the development of www.aacrjournals.org 4649 Clin Cancer Res 2009;15(14) July 15, 2009 Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2009 American Association for Cancer Research. Cancer Therapy: Preclinical cancer based on in vitro analyses (16). Our studies are consistent Translational Relevance with those findings and, moreover, offer extensive in vivo confirmation of the important role MEK signaling plays in We find that basal-like breast cancer models are sensitive basal-like breast cancer. In addition, we also show that activation in vitro in vivo and to the inhibitory effects of mitogen- of the PI3K/Akt pathway by loss of the tumor suppressor PTEN activated protein kinase kinase (MEK) inhibition relative to occurs in a subset of basal-like breast cancer cell line models and luminal and HER2-amplified tumors and identify the tumor is associated with diminished response to MEK inhibition, and suppressor PTEN as a negative predictor factor for that siRNA ablation of PTEN in responsive models blocks cell response to MEK inhibition.We also find that a combination cycle and proliferative changes triggered by MEK inhibition. of MEK and phosphatidylinositol 3-kinase inhibitors Based on the apparent role of PTEN and activation of the PI3K/ results in enhanced efficacy in both PTEN wild-type and Akt pathway in mediating resistance to MEK inhibition, we use PTEN-null basal-like tumors and provide a mechanistic both in vitro and in vivo combination studies with potent and explanation for this observation. Thus, our studies provide selective MEK and PI3K inhibitors to show that simultaneous a rational hypothesis for biomarker evaluation and patient treatment with a MEK inhibitor and a potent and selective stratification in clinical trials seeking to evaluate the efficacy inhibitor of PI3K results in synergistic decreases in cell viability, of MEK and phosphatidylinositol 3-kinase inhibitor enhanced down-regulation of cyclin D1, and increased apoptosis treatment in this poor-prognosis tumor type. specifically in basal-like breast cancer models. molecules such as PD0325901 and AZD6244 that act through a Materials and Methods novel noncompetitive allosteric mechanism of inhibition by binding to a site adjacent to the ATP-binding site and locking Cell lines and cell viability experiments. Details and sources of all the kinase in a closed and catalytically inactive conformation cell lines used in this study have been described previously (11, 17) and (9). This pathway is frequently activated by mutations in RAS are also detailed in Supplementary Materials and Methods. All studies family members or BRAF in a number of solid tumor types, and in this report describing MEK inhibition have used the potent and elegant studies in cell lines and mouse tumor models have selective MEK1/2 inhibitor PD0325901 (Pfizer) to evaluate the effects suggested that mutations in BRAF and, to a lesser extent, KRAS of MEK inhibition in breast cancer preclinical models, with the exception of a small number of confirmatory studies described in may be useful to stratify patients enrolled in clinical trials of Supplementary materials that use a second potent and selective MEK MEK inhibitors (10). We sought to address whether MEK inhibitor (9), purchased from Synthesis MedChem.5 For cell viability inhibition might have therapeutic value in breast cancer studies, cells were plated in quadruplicate at a density of 3,000 per well through evaluation of a large panel of breast cancer cell lines in 384-well plates in normal growth medium and allowed to adhere and tumor xenograft models that have been characterized in overnight. Compounds were added in 10 concentrations based on a detail at the molecular level and shown to reflect many of the 3-fold dilution series. Cell viability was measured 72 h later using the genomic alterations commonly found in breast tumors (11). CellTiter-Glo Luminescent Cell Viability Assay (Promega). The concen- We also set out to examine the relationship between RAS/ tration of drug resulting in 50% of maximal inhibition (EC50) was RAF/MEK/ERK pathway activity and phosphatidylinositol calculated from a four-parameter
Recommended publications
  • Deregulated Gene Expression Pathways in Myelodysplastic Syndrome Hematopoietic Stem Cells
    Leukemia (2010) 24, 756–764 & 2010 Macmillan Publishers Limited All rights reserved 0887-6924/10 $32.00 www.nature.com/leu ORIGINAL ARTICLE Deregulated gene expression pathways in myelodysplastic syndrome hematopoietic stem cells A Pellagatti1, M Cazzola2, A Giagounidis3, J Perry1, L Malcovati2, MG Della Porta2,MJa¨dersten4, S Killick5, A Verma6, CJ Norbury7, E Hellstro¨m-Lindberg4, JS Wainscoat1 and J Boultwood1 1LRF Molecular Haematology Unit, NDCLS, John Radcliffe Hospital, Oxford, UK; 2Department of Hematology Oncology, University of Pavia Medical School, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; 3Medizinische Klinik II, St Johannes Hospital, Duisburg, Germany; 4Division of Hematology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden; 5Department of Haematology, Royal Bournemouth Hospital, Bournemouth, UK; 6Albert Einstein College of Medicine, Bronx, NY, USA and 7Sir William Dunn School of Pathology, University of Oxford, Oxford, UK To gain insight into the molecular pathogenesis of the the World Health Organization.6,7 Patients with refractory myelodysplastic syndromes (MDS), we performed global gene anemia (RA) with or without ringed sideroblasts, according to expression profiling and pathway analysis on the hemato- poietic stem cells (HSC) of 183 MDS patients as compared with the the French–American–British classification, were subdivided HSC of 17 healthy controls. The most significantly deregulated based on the presence or absence of multilineage dysplasia. In pathways in MDS include interferon signaling, thrombopoietin addition, patients with RA with excess blasts (RAEB) were signaling and the Wnt pathways. Among the most signifi- subdivided into two categories, RAEB1 and RAEB2, based on the cantly deregulated gene pathways in early MDS are immuno- percentage of bone marrow blasts.
    [Show full text]
  • Mouse Rras2 Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Rras2 Knockout Project (CRISPR/Cas9) Objective: To create a Rras2 knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Rras2 gene (NCBI Reference Sequence: NM_025846 ; Ensembl: ENSMUSG00000055723 ) is located on Mouse chromosome 7. 6 exons are identified, with the ATG start codon in exon 1 and the TAA stop codon in exon 6 (Transcript: ENSMUST00000069449). Exon 3~5 will be selected as target site. Cas9 and gRNA will be co-injected into fertilized eggs for KO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Homozygote and heterozygote null mice are lymphopenic, resulting from diminished homeostatic proliferation and impaired T cell and B cell survival. Mice homozygous for a gene trap insertion exhibit retinal degeneration, and increased total body mass and total body fat. Exon 3 starts from about 32.19% of the coding region. Exon 3~5 covers 54.08% of the coding region. The size of effective KO region: ~8729 bp. The KO region does not have any other known gene. Page 1 of 8 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele 5' gRNA region gRNA region 3' 1 3 4 5 6 Legends Exon of mouse Rras2 Knockout region Page 2 of 8 https://www.alphaknockout.com Overview of the Dot Plot (up) Window size: 15 bp Forward Reverse Complement Sequence 12 Note: The 1302 bp section upstream of Exon 3 is aligned with itself to determine if there are tandem repeats. Tandem repeats are found in the dot plot matrix.
    [Show full text]
  • The Capacity of Long-Term in Vitro Proliferation of Acute Myeloid
    The Capacity of Long-Term in Vitro Proliferation of Acute Myeloid Leukemia Cells Supported Only by Exogenous Cytokines Is Associated with a Patient Subset with Adverse Outcome Annette K. Brenner, Elise Aasebø, Maria Hernandez-Valladares, Frode Selheim, Frode Berven, Ida-Sofie Grønningsæter, Sushma Bartaula-Brevik and Øystein Bruserud Supplementary Material S2 of S31 Table S1. Detailed information about the 68 AML patients included in the study. # of blasts Viability Proliferation Cytokine Viable cells Change in ID Gender Age Etiology FAB Cytogenetics Mutations CD34 Colonies (109/L) (%) 48 h (cpm) secretion (106) 5 weeks phenotype 1 M 42 de novo 241 M2 normal Flt3 pos 31.0 3848 low 0.24 7 yes 2 M 82 MF 12.4 M2 t(9;22) wt pos 81.6 74,686 low 1.43 969 yes 3 F 49 CML/relapse 149 M2 complex n.d. pos 26.2 3472 low 0.08 n.d. no 4 M 33 de novo 62.0 M2 normal wt pos 67.5 6206 low 0.08 6.5 no 5 M 71 relapse 91.0 M4 normal NPM1 pos 63.5 21,331 low 0.17 n.d. yes 6 M 83 de novo 109 M1 n.d. wt pos 19.1 8764 low 1.65 693 no 7 F 77 MDS 26.4 M1 normal wt pos 89.4 53,799 high 3.43 2746 no 8 M 46 de novo 26.9 M1 normal NPM1 n.d. n.d. 3472 low 1.56 n.d. no 9 M 68 MF 50.8 M4 normal D835 pos 69.4 1640 low 0.08 n.d.
    [Show full text]
  • RRAS2 Rabbit Polyclonal Antibody – TA308124 | Origene
    OriGene Technologies, Inc. 9620 Medical Center Drive, Ste 200 Rockville, MD 20850, US Phone: +1-888-267-4436 [email protected] EU: [email protected] CN: [email protected] Product datasheet for TA308124 RRAS2 Rabbit Polyclonal Antibody Product data: Product Type: Primary Antibodies Applications: IHC, WB Recommended Dilution: IHC:1:100-1:1000; WB:1:1000-1:10000 Reactivity: Human, Mouse (Predicted: Bovine, Rhesus Monkey, Rat) Host: Rabbit Isotype: IgG Clonality: Polyclonal Immunogen: Recombinant fragment corresponding to a region within amino acids 1 and 204 of TC21 (Uniprot ID#P62070) Formulation: 0.1M Tris, 0.1M Glycine, 10% Glycerol (pH7). 0.01% Thimerosal was added as a preservative. Concentration: lot specific Purification: Purified by antigen-affinity chromatography. Conjugation: Unconjugated Storage: Store at -20°C as received. Stability: Stable for 12 months from date of receipt. Predicted Protein Size: 23 kDa Gene Name: related RAS viral (r-ras) oncogene homolog 2 Database Link: NP_001096139 Entrez Gene 66922 MouseEntrez Gene 365355 RatEntrez Gene 22800 Human P62070 Synonyms: TC21 Note: Seq homology of immunogen across species: Rat (100%), Rhesus Monkey (100%), Bovine (100%) Protein Families: Druggable Genome Protein Pathways: MAPK signaling pathway, Regulation of actin cytoskeleton, Tight junction This product is to be used for laboratory only. Not for diagnostic or therapeutic use. View online » ©2021 OriGene Technologies, Inc., 9620 Medical Center Drive, Ste 200, Rockville, MD 20850, US 1 / 3 RRAS2 Rabbit Polyclonal Antibody – TA308124 Product images: Sample (50 ug of whole cell lysate). A: mouse brain. 12% SDS PAGE. TA308124 diluted at 1:5000. Sample (30 ug of whole cell lysate).
    [Show full text]
  • LIAO-DISSERTATION-2019.Pdf (4.688Mb)
    A Genetic Interaction Analysis Identifies Novel Cancer Driver Modifiers and a Combination Therapy The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Liao, Sida. 2019. A Genetic Interaction Analysis Identifies Novel Cancer Driver Modifiers and a Combination Therapy. Doctoral dissertation, Harvard University, Graduate School of Arts & Sciences. Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:42029794 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 A genetic interaction analysis identifies novel cancer driver modifiers and a combination therapy A dissertation presented by Sida Liao 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 April 2019 © 2019 Sida Liao All rights reserved. Dissertation Advisor: Dr. Stephen J. Elledge Sida Liao A genetic interaction analysis identifies novel cancer driver modifiers and a combination therapy Abstract A large number of cancer drivers have been identified through tumor sequencing efforts but how they interact and the degree to which they can substitute for each other has not been systematically explored. To comprehensively investigate how cancer drivers genetically interact, I searched for modifiers of EGFR dependency by performing CRISPR, shRNA and expression screens in a non-small cell lung cancer model. I elucidated a broad spectrum of TSGs and OGs that can genetically modify proliferation and survival of cancer cells when EGFR signaling is altered.
    [Show full text]
  • Role and Regulation of the P53-Homolog P73 in the Transformation of Normal Human Fibroblasts
    Role and regulation of the p53-homolog p73 in the transformation of normal human fibroblasts Dissertation zur Erlangung des naturwissenschaftlichen Doktorgrades der Bayerischen Julius-Maximilians-Universität Würzburg vorgelegt von Lars Hofmann aus Aschaffenburg Würzburg 2007 Eingereicht am Mitglieder der Promotionskommission: Vorsitzender: Prof. Dr. Dr. Martin J. Müller Gutachter: Prof. Dr. Michael P. Schön Gutachter : Prof. Dr. Georg Krohne Tag des Promotionskolloquiums: Doktorurkunde ausgehändigt am Erklärung Hiermit erkläre ich, dass ich die vorliegende Arbeit selbständig angefertigt und keine anderen als die angegebenen Hilfsmittel und Quellen verwendet habe. Diese Arbeit wurde weder in gleicher noch in ähnlicher Form in einem anderen Prüfungsverfahren vorgelegt. Ich habe früher, außer den mit dem Zulassungsgesuch urkundlichen Graden, keine weiteren akademischen Grade erworben und zu erwerben gesucht. Würzburg, Lars Hofmann Content SUMMARY ................................................................................................................ IV ZUSAMMENFASSUNG ............................................................................................. V 1. INTRODUCTION ................................................................................................. 1 1.1. Molecular basics of cancer .......................................................................................... 1 1.2. Early research on tumorigenesis ................................................................................. 3 1.3. Developing
    [Show full text]
  • Transcriptomic and Epigenomic Characterization of the Developing Bat Wing
    ARTICLES OPEN Transcriptomic and epigenomic characterization of the developing bat wing Walter L Eckalbar1,2,9, Stephen A Schlebusch3,9, Mandy K Mason3, Zoe Gill3, Ash V Parker3, Betty M Booker1,2, Sierra Nishizaki1,2, Christiane Muswamba-Nday3, Elizabeth Terhune4,5, Kimberly A Nevonen4, Nadja Makki1,2, Tara Friedrich2,6, Julia E VanderMeer1,2, Katherine S Pollard2,6,7, Lucia Carbone4,8, Jeff D Wall2,7, Nicola Illing3 & Nadav Ahituv1,2 Bats are the only mammals capable of powered flight, but little is known about the genetic determinants that shape their wings. Here we generated a genome for Miniopterus natalensis and performed RNA-seq and ChIP-seq (H3K27ac and H3K27me3) analyses on its developing forelimb and hindlimb autopods at sequential embryonic stages to decipher the molecular events that underlie bat wing development. Over 7,000 genes and several long noncoding RNAs, including Tbx5-as1 and Hottip, were differentially expressed between forelimb and hindlimb, and across different stages. ChIP-seq analysis identified thousands of regions that are differentially modified in forelimb and hindlimb. Comparative genomics found 2,796 bat-accelerated regions within H3K27ac peaks, several of which cluster near limb-associated genes. Pathway analyses highlighted multiple ribosomal proteins and known limb patterning signaling pathways as differentially regulated and implicated increased forelimb mesenchymal condensation in differential growth. In combination, our work outlines multiple genetic components that likely contribute to bat wing formation, providing insights into this morphological innovation. The order Chiroptera, commonly known as bats, is the only group of To characterize the genetic differences that underlie divergence in mammals to have evolved the capability of flight.
    [Show full text]
  • The Human Gene Connectome As a Map of Short Cuts for Morbid Allele Discovery
    The human gene connectome as a map of short cuts for morbid allele discovery Yuval Itana,1, Shen-Ying Zhanga,b, Guillaume Vogta,b, Avinash Abhyankara, Melina Hermana, Patrick Nitschkec, Dror Friedd, Lluis Quintana-Murcie, Laurent Abela,b, and Jean-Laurent Casanovaa,b,f aSt. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065; bLaboratory of Human Genetics of Infectious Diseases, Necker Branch, Paris Descartes University, Institut National de la Santé et de la Recherche Médicale U980, Necker Medical School, 75015 Paris, France; cPlateforme Bioinformatique, Université Paris Descartes, 75116 Paris, France; dDepartment of Computer Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; eUnit of Human Evolutionary Genetics, Centre National de la Recherche Scientifique, Unité de Recherche Associée 3012, Institut Pasteur, F-75015 Paris, France; and fPediatric Immunology-Hematology Unit, Necker Hospital for Sick Children, 75015 Paris, France Edited* by Bruce Beutler, University of Texas Southwestern Medical Center, Dallas, TX, and approved February 15, 2013 (received for review October 19, 2012) High-throughput genomic data reveal thousands of gene variants to detect a single mutated gene, with the other polymorphic genes per patient, and it is often difficult to determine which of these being of less interest. This goes some way to explaining why, variants underlies disease in a given individual. However, at the despite the abundance of NGS data, the discovery of disease- population level, there may be some degree of phenotypic homo- causing alleles from such data remains somewhat limited. geneity, with alterations of specific physiological pathways under- We developed the human gene connectome (HGC) to over- come this problem.
    [Show full text]
  • Download a Subgraph Composed of the Initially 32 Raso- of Proteins Underlying Rasopathies in Order to Iden- Pathy Proteins Selected in This Study
    Montero‑Bullón et al. Orphanet J Rare Dis (2021) 16:303 https://doi.org/10.1186/s13023‑021‑01934‑x RESEARCH Open Access Integrated in silico MS‑based phosphoproteomics and network enrichment analysis of RASopathy proteins Javier‑Fernando Montero‑Bullón1, Óscar González‑Velasco2, María Isidoro‑García3,4,5,6 and Jesus Lacal3,7* Abstract Background: RASopathies are a group of syndromes showing clinical overlap caused by mutations in genes afect‑ ing the RAS‑MAPK pathway. Consequent disruption on cellular signaling leads and is driven by phosphoproteome remodeling. However, we still lack a comprehensive picture of the diferent key players and altered downstream efectors. Methods: An in silico interactome of RASopathy proteins was generated using pathway enrichment analysis/STRING tool, including identifcation of main hub proteins. We also integrated phosphoproteomic and immunoblotting stud‑ ies using previous published information on RASopathy proteins and their neighbors in the context of RASopathy syndromes. Data from Phosphosite database (www. phosp hosite. org) was collected in order to obtain the potential phosphosites subjected to regulation in the 27 causative RASopathy proteins. We compiled a dataset of dysregulated phosphosites in RASopathies, searched for commonalities between syndromes in harmonized data, and analyzed the role of phosphorylation in the syndromes by the identifcation of key players between the causative RASopathy proteins and the associated interactome. Results: In this study, we provide a curated data set of 27 causative RASopathy genes, identify up to 511 protein– protein associations using pathway enrichment analysis/STRING tool, and identify 12 nodes as main hub proteins. We found that a large group of proteins contain tyrosine residues and their biological processes include but are not limited to the nervous system.
    [Show full text]
  • Integrative Analysis of Disease Signatures Shows Inflammation Disrupts Juvenile Experience-Dependent Cortical Plasticity
    New Research Development Integrative Analysis of Disease Signatures Shows Inflammation Disrupts Juvenile Experience- Dependent Cortical Plasticity Milo R. Smith1,2,3,4,5,6,7,8, Poromendro Burman1,3,4,5,8, Masato Sadahiro1,3,4,5,6,8, Brian A. Kidd,2,7 Joel T. Dudley,2,7 and Hirofumi Morishita1,3,4,5,8 DOI:http://dx.doi.org/10.1523/ENEURO.0240-16.2016 1Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, 2Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, 3Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029, 4Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, 5Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, 6Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, 7Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, and 8Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029 Visual Abstract Throughout childhood and adolescence, periods of heightened neuroplasticity are critical for the development of healthy brain function and behavior. Given the high prevalence of neurodevelopmental disorders, such as autism, identifying disruptors of developmental plasticity represents an essential step for developing strategies for prevention and intervention. Applying a novel computational approach that systematically assessed connections between 436 transcriptional signatures of disease and multiple signatures of neuroplasticity, we identified inflammation as a common pathological process central to a diverse set of diseases predicted to dysregulate Significance Statement During childhood and adolescence, heightened neuroplasticity allows the brain to reorganize and adapt to its environment.
    [Show full text]
  • Primepcr™Assay Validation Report
    PrimePCR™Assay Validation Report Gene Information Gene Name ras-related protein R-Ras2 Gene Symbol Rras2 Organism Rat Gene Summary Description Not Available Gene Aliases Not Available RefSeq Accession No. Not Available UniGene ID Rn.3271 Ensembl Gene ID ENSRNOG00000012258 Entrez Gene ID 365355 Assay Information Unique Assay ID qRnoCID0006243 Assay Type SYBR® Green Detected Coding Transcript(s) ENSRNOT00000017199 Amplicon Context Sequence TTCTTGTCTTTTTCTTTCCGGGTTGGTTCTGGTGAAGGAGGACATTCTTGTTCTTG AAACTTCCTGATAACTCGGACAAGTTCGTGGAAAGCTTGATCTACATTCATCCTG ATCTTTGCCGACGCCTCCATATACGTGACCTTGAGC Amplicon Length (bp) 117 Chromosome Location 1:185909667-185912813 Assay Design Intron-spanning Purification Desalted Validation Results Efficiency (%) 94 R2 0.9968 cDNA Cq 19.45 cDNA Tm (Celsius) 82 gDNA Cq 27.52 Specificity (%) 100 Information to assist with data interpretation is provided at the end of this report. Page 1/4 PrimePCR™Assay Validation Report Rras2, Rat Amplification Plot Amplification of cDNA generated from 25 ng of universal reference RNA Melt Peak Melt curve analysis of above amplification Standard Curve Standard curve generated using 20 million copies of template diluted 10-fold to 20 copies Page 2/4 PrimePCR™Assay Validation Report Products used to generate validation data Real-Time PCR Instrument CFX384 Real-Time PCR Detection System Reverse Transcription Reagent iScript™ Advanced cDNA Synthesis Kit for RT-qPCR Real-Time PCR Supermix SsoAdvanced™ SYBR® Green Supermix Experimental Sample qPCR Reference Total RNA Data Interpretation Unique Assay ID This is a unique identifier that can be used to identify the assay in the literature and online. Detected Coding Transcript(s) This is a list of the Ensembl transcript ID(s) that this assay will detect. Details for each transcript can be found on the Ensembl website at www.ensembl.org.
    [Show full text]
  • Full Text (PDF)
    The 3′ IgH Locus Control Region Is Sufficient to Deregulate a c- myc Transgene and Promote Mature B Cell Malignancies with a Predominant Burkitt-Like Phenotype This information is current as of September 29, 2021. Véronique Truffinet, Eric Pinaud, Nadine Cogné, Barbara Petit, Laurence Guglielmi, Michel Cogné and Yves Denizot J Immunol 2007; 179:6033-6042; ; doi: 10.4049/jimmunol.179.9.6033 http://www.jimmunol.org/content/179/9/6033 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2008/03/12/179.9.6033.DC1 Material http://www.jimmunol.org/ References This article cites 36 articles, 18 of which you can access for free at: http://www.jimmunol.org/content/179/9/6033.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision by guest on September 29, 2021 • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2007 by The American Association of Immunologists All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology The 3؅ IgH Locus Control Region Is Sufficient to Deregulate a c-myc Transgene and Promote Mature B Cell Malignancies with a Predominant Burkitt-Like Phenotype1 Ve´ronique Truffinet,* Eric Pinaud,* Nadine Cogne´,* Barbara Petit,*† Laurence Guglielmi,2* Michel Cogne´,* and Yves Denizot3* Burkitt lymphoma (BL) features translocations linking c-myc to an Ig locus.
    [Show full text]