Eukaryotic Mrna Literature Flyer-20200806

Total Page:16

File Type:pdf, Size:1020Kb

Eukaryotic Mrna Literature Flyer-20200806 Published in Nature Communications Armadillo Repeat Containing 12 Promotes Neuroblastoma Progression Through Interaction with Retinoblastoma Binding Protein 4 Background Armadillo (ARM) family proteins play an important role in tumor progression. This paper identified ARMC12 as an ARM member associated with neuroblastoma progression. 1 Materials 6 · Tumor cells (1×10 ) 2 RNA Isolation · Total RNA extracted using TRIzol® Research Pipeline 3 Library Construction · mRNA libraries for RNA-seq 4 Sequencing · Illumina platform 5 Bioinformatic Analysis · Differential expression · GSEA (Gene Set Enrichment Analysis) Research Results Armadillo repeat containing 12 (ARMC12 ), an ARM member associated with NB progression, promotes the growth and aggressiveness of NB cell lines. Mechanistically, ARMC12 physically interacts with retinoblastoma binding protein 4 (RBBP4) to facilitate the formation and activity of polycomb repressive complex 2 (PRC2), resulting in transcriptional repression of tumor suppressive genes. Blocking the interaction between ARMC12 and RBBP4 by cell-penetrating inhibitory peptide activates downstream gene expression and suppresses the tumorigenesis and aggressiveness of NB cells. Both ARMC12 and RBBP4 are upregulated in NB tissues, and are associated with unfavorable outcomes for patients. Analysis Showcase Novogene’s RNA-seq analysis helped to screen differentially expressed genes upon ARMC12 overexpression. Five genes, CADM1 , EGLN3 , HRK , HS6ST3 , and SMAD9 , were identified and further proved to be suppressed by ARMC12 , facilitating the growth and aggressiveness of NB cells. a k Moc ARMC12 100 Correlated genes Correlated genes HS6ST2 (GSE16476) (GSE62564) Down: Up: 327 3798 HS6ST3 CADM1 3224 2901 443 82 0 SMAD9 RNA-Seq ChIP-X EGLN3 105 MBNL3 (padj) 0 50 4493 0 10 HRK CHPT1 –1.04 852 14 0 MYO1B –0.69 –Log SETD7 –0.35 PPP1R3C 0.00 155 4 PLS1 0.35 0.69 1.3 TMEM45B 420 SLC7A11 1.04 0 4 8 –8 –4 12 16 –16 –12 Log2 (fold change) Volcano plots (left panel), Venn diagram (middle panel), and heatmap (right panel) show the alteration of gene expression in ARMC12 overexpressed or normal expressed cells. Red indicates high expression, and blue indicates low expression in the heatmap. Reference Subramanian, A. et al . Gene set enrichment analysis: A knowledge-basedapproach for interpreting genome-wide expres- sion profiles. Proc. Natl. Acad.Sci. USA 102, 15545‒15550 (2005). Powered Literature by Novogene Year Journal Title 2018 Cell Targeting Epigenetic Crosstalk as a Therapeutic Strategy for EZH2 -Aberrant Solid Tumors Armadillo repeat containing 12 promotes neuroblastoma progression through interaction 2018 Nat Commun with retinoblastoma binding protein 4 E6 protein expressed by high-risk HPV activates super-enhancers of the EGFR and c-MET 2018 Cancer Res oncogenes by destabilizing the histone demethylase KDM5C Pold3 is required for genomic stability and telomere integrity in embryonic stem cells Nucleic Acids Res 2018 and meiosis Single-Base Methylome Analysis Reveals Dynamic Epigenomic Differences Associated 2018 Plant Biotechnol J with Water Deficit in Apple A Critical Role of Presynaptic Cadherin/Catenin/ p140Cap Complexes in Stabilizing 2017 Neuron Spines and Functional Synapses in the Neocortex The genome of the medicinal plant Macleaya cordata provides new insights into 2017 Mol Plant benzylisoquinoline alkaloids metabolism SIRT6 Controls Hematopoietic Stem Cell Homeostasis through Epigenetic Regulation 2016 Cell Stem Cell of Wnt Signaling Novogene Corporation Inc. 8801 Folsom Blvd #290, Sacramento, CA 95826 916-252-0068-383 [email protected] en.novogene.com Follow us on LinkedIn.
Recommended publications
  • Hypoxia and Oxygen-Sensing Signaling in Gene Regulation and Cancer Progression
    International Journal of Molecular Sciences Review Hypoxia and Oxygen-Sensing Signaling in Gene Regulation and Cancer Progression Guang Yang, Rachel Shi and Qing Zhang * Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; [email protected] (G.Y.); [email protected] (R.S.) * Correspondence: [email protected]; Tel.: +1-214-645-4671 Received: 6 October 2020; Accepted: 29 October 2020; Published: 31 October 2020 Abstract: Oxygen homeostasis regulation is the most fundamental cellular process for adjusting physiological oxygen variations, and its irregularity leads to various human diseases, including cancer. Hypoxia is closely associated with cancer development, and hypoxia/oxygen-sensing signaling plays critical roles in the modulation of cancer progression. The key molecules of the hypoxia/oxygen-sensing signaling include the transcriptional regulator hypoxia-inducible factor (HIF) which widely controls oxygen responsive genes, the central members of the 2-oxoglutarate (2-OG)-dependent dioxygenases, such as prolyl hydroxylase (PHD or EglN), and an E3 ubiquitin ligase component for HIF degeneration called von Hippel–Lindau (encoding protein pVHL). In this review, we summarize the current knowledge about the canonical hypoxia signaling, HIF transcription factors, and pVHL. In addition, the role of 2-OG-dependent enzymes, such as DNA/RNA-modifying enzymes, JmjC domain-containing enzymes, and prolyl hydroxylases, in gene regulation of cancer progression, is specifically reviewed. We also discuss the therapeutic advancement of targeting hypoxia and oxygen sensing pathways in cancer. Keywords: hypoxia; PHDs; TETs; JmjCs; HIFs 1. Introduction Molecular oxygen serves as a co-factor in many biochemical processes and is fundamental for aerobic organisms to maintain intracellular ATP levels [1,2].
    [Show full text]
  • Interplay Between Epigenetics and Metabolism in Oncogenesis: Mechanisms and Therapeutic Approaches
    OPEN Oncogene (2017) 36, 3359–3374 www.nature.com/onc REVIEW Interplay between epigenetics and metabolism in oncogenesis: mechanisms and therapeutic approaches CC Wong1, Y Qian2,3 and J Yu1 Epigenetic and metabolic alterations in cancer cells are highly intertwined. Oncogene-driven metabolic rewiring modifies the epigenetic landscape via modulating the activities of DNA and histone modification enzymes at the metabolite level. Conversely, epigenetic mechanisms regulate the expression of metabolic genes, thereby altering the metabolome. Epigenetic-metabolomic interplay has a critical role in tumourigenesis by coordinately sustaining cell proliferation, metastasis and pluripotency. Understanding the link between epigenetics and metabolism could unravel novel molecular targets, whose intervention may lead to improvements in cancer treatment. In this review, we summarized the recent discoveries linking epigenetics and metabolism and their underlying roles in tumorigenesis; and highlighted the promising molecular targets, with an update on the development of small molecule or biologic inhibitors against these abnormalities in cancer. Oncogene (2017) 36, 3359–3374; doi:10.1038/onc.2016.485; published online 16 January 2017 INTRODUCTION metabolic genes have also been identified as driver genes It has been appreciated since the early days of cancer research mutated in some cancers, such as isocitrate dehydrogenase 1 16 17 that the metabolic profiles of tumor cells differ significantly from and 2 (IDH1/2) in gliomas and acute myeloid leukemia (AML), 18 normal cells. Cancer cells have high metabolic demands and they succinate dehydrogenase (SDH) in paragangliomas and fuma- utilize nutrients with an altered metabolic program to support rate hydratase (FH) in hereditary leiomyomatosis and renal cell 19 their high proliferative rates and adapt to the hostile tumor cancer (HLRCC).
    [Show full text]
  • Mutant IDH, (R)-2-Hydroxyglutarate, and Cancer
    Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press REVIEW What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer Julie-Aurore Losman1 and William G. Kaelin Jr.1,2,3 1Department of Medical Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA; 2Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA Mutations in metabolic enzymes, including isocitrate whether altered cellular metabolism is a cause of cancer dehydrogenase 1 (IDH1) and IDH2, in cancer strongly or merely an adaptive response of cancer cells in the face implicate altered metabolism in tumorigenesis. IDH1 of accelerated cell proliferation is still a topic of some and IDH2 catalyze the interconversion of isocitrate and debate. 2-oxoglutarate (2OG). 2OG is a TCA cycle intermediate The recent identification of cancer-associated muta- and an essential cofactor for many enzymes, including tions in three metabolic enzymes suggests that altered JmjC domain-containing histone demethylases, TET cellular metabolism can indeed be a cause of some 5-methylcytosine hydroxylases, and EglN prolyl-4-hydrox- cancers (Pollard et al. 2003; King et al. 2006; Raimundo ylases. Cancer-associated IDH mutations alter the enzymes et al. 2011). Two of these enzymes, fumarate hydratase such that they reduce 2OG to the structurally similar (FH) and succinate dehydrogenase (SDH), are bone fide metabolite (R)-2-hydroxyglutarate [(R)-2HG]. Here we tumor suppressors, and loss-of-function mutations in FH review what is known about the molecular mechanisms and SDH have been identified in various cancers, in- of transformation by mutant IDH and discuss their im- cluding renal cell carcinomas and paragangliomas.
    [Show full text]
  • Protein Identities in Evs Isolated from U87-MG GBM Cells As Determined by NG LC-MS/MS
    Protein identities in EVs isolated from U87-MG GBM cells as determined by NG LC-MS/MS. No. Accession Description Σ Coverage Σ# Proteins Σ# Unique Peptides Σ# Peptides Σ# PSMs # AAs MW [kDa] calc. pI 1 A8MS94 Putative golgin subfamily A member 2-like protein 5 OS=Homo sapiens PE=5 SV=2 - [GG2L5_HUMAN] 100 1 1 7 88 110 12,03704523 5,681152344 2 P60660 Myosin light polypeptide 6 OS=Homo sapiens GN=MYL6 PE=1 SV=2 - [MYL6_HUMAN] 100 3 5 17 173 151 16,91913397 4,652832031 3 Q6ZYL4 General transcription factor IIH subunit 5 OS=Homo sapiens GN=GTF2H5 PE=1 SV=1 - [TF2H5_HUMAN] 98,59 1 1 4 13 71 8,048185945 4,652832031 4 P60709 Actin, cytoplasmic 1 OS=Homo sapiens GN=ACTB PE=1 SV=1 - [ACTB_HUMAN] 97,6 5 5 35 917 375 41,70973209 5,478027344 5 P13489 Ribonuclease inhibitor OS=Homo sapiens GN=RNH1 PE=1 SV=2 - [RINI_HUMAN] 96,75 1 12 37 173 461 49,94108966 4,817871094 6 P09382 Galectin-1 OS=Homo sapiens GN=LGALS1 PE=1 SV=2 - [LEG1_HUMAN] 96,3 1 7 14 283 135 14,70620005 5,503417969 7 P60174 Triosephosphate isomerase OS=Homo sapiens GN=TPI1 PE=1 SV=3 - [TPIS_HUMAN] 95,1 3 16 25 375 286 30,77169764 5,922363281 8 P04406 Glyceraldehyde-3-phosphate dehydrogenase OS=Homo sapiens GN=GAPDH PE=1 SV=3 - [G3P_HUMAN] 94,63 2 13 31 509 335 36,03039959 8,455566406 9 Q15185 Prostaglandin E synthase 3 OS=Homo sapiens GN=PTGES3 PE=1 SV=1 - [TEBP_HUMAN] 93,13 1 5 12 74 160 18,68541938 4,538574219 10 P09417 Dihydropteridine reductase OS=Homo sapiens GN=QDPR PE=1 SV=2 - [DHPR_HUMAN] 93,03 1 1 17 69 244 25,77302971 7,371582031 11 P01911 HLA class II histocompatibility antigen,
    [Show full text]
  • Endocrine Abstracts Vol 65
    Endocrine Abstracts November 2019 Volume 65 ISSN 1479-6848 (online) Society for Endocrinology BES 2019 11–13 November 2019, Brighton published by Online version available at bioscientifica www.endocrine-abstracts.org Volume 65 Endocrine Abstracts November 2019 Society for Endocrinology BES 2019 11–13 November 2019, Brighton VOLUME EDITORS The abstracts submitted were marked by the Abstract Marking panel, selected by the Programme Organising Committee. Programme Committee D Bassett (Programme Secretary) (London) Laura Matthews (Leeds) Andrew Childs (Programme Co-ordinator) (London) Carla Moran (Cambridge) Nils Krone (Programme Co-ordinator) (Sheffield) Annice Mukherjee (Salford) Helen Simpson (Programme Co-ordinator) (London) Francesca Spiga (Bristol) Davide Calebiro (Birmingham) Jeremy Tomlinson (Oxford) Ben Challis (Cambridge) Jennifer Walsh (Sheffield) Mandy Drake (Edinburgh) Abstract Marking Panel Ramzi Ajjan (Leeds) Neil Gittoes (Birmingham) John Newell-Price (Sheffield) Richard Anderson (Edinburgh) Helena Gleeson (Birmingham) Mark Nixon (Edinburgh) Ruth Andrew (Edinburgh) Philippa Hanson (London) Finbarr O’Harte (Ulster) Weibke Arlt (Birmingham) Martin Hewison (Birmingham) Adrian Park (Cambridge) Mo Aye (Hull) Claire Higham (Manchester) Simon Pearce (Newcastle) Tom Barber (Warwick) Steve Hillier (Edinburgh) Andrew Powlson (Cambridge) Duncan Bassett (London) Andy James (Newcastle) Teresa Rea (Belfast) Roger Brown (Edinburgh) Channa Jayasena (London) Martin Read (Birmingham) Paul Carroll (London) Niki Karavitaki (Oxford) Aled Rees (Cardiff)
    [Show full text]
  • Potential Genotoxicity from Integration Sites in CLAD Dogs Treated Successfully with Gammaretroviral Vector-Mediated Gene Therapy
    Gene Therapy (2008) 15, 1067–1071 & 2008 Nature Publishing Group All rights reserved 0969-7128/08 $30.00 www.nature.com/gt SHORT COMMUNICATION Potential genotoxicity from integration sites in CLAD dogs treated successfully with gammaretroviral vector-mediated gene therapy M Hai1,3, RL Adler1,3, TR Bauer Jr1,3, LM Tuschong1, Y-C Gu1,XWu2 and DD Hickstein1 1Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA and 2Laboratory of Molecular Technology, Scientific Applications International Corporation-Frederick, National Cancer Institute-Frederick, Frederick, Maryland, USA Integration site analysis was performed on six dogs with in hematopoietic stem cells. Integrations clustered around canine leukocyte adhesion deficiency (CLAD) that survived common insertion sites more frequently than random. greater than 1 year after infusion of autologous CD34+ bone Despite potential genotoxicity from RIS, to date there has marrow cells transduced with a gammaretroviral vector been no progression to oligoclonal hematopoiesis and no expressing canine CD18. A total of 387 retroviral insertion evidence that vector integration sites influenced cell survival sites (RIS) were identified in the peripheral blood leukocytes or proliferation. Continued follow-up in disease-specific from the six dogs at 1 year postinfusion. A total of 129 RIS animal models such as CLAD will be required to provide an were identified in CD3+ T-lymphocytes and 102 RIS in accurate estimate
    [Show full text]
  • An Enhanced Chemopreventive Effect of Methyl Donor S-Adenosylmethionine in Combination with 25-Hydroxyvitamin D in Blocking Mammary Tumor Growth and Metastasis
    Bone Research www.nature.com/boneres ARTICLE OPEN An enhanced chemopreventive effect of methyl donor S-adenosylmethionine in combination with 25-hydroxyvitamin D in blocking mammary tumor growth and metastasis Niaz Mahmood1, Ani Arakelian1, William J. Muller2, Moshe Szyf3 and Shafaat A. Rabbani1 Therapeutic targeting of metastatic breast cancer still remains a challenge as the tumor cells are highly heterogenous and exploit multiple pathways for their growth and metastatic spread that cannot always be targeted by a single-agent monotherapy regimen. Therefore, a rational approach through simultaneous targeting of several pathways may provide a better anti-cancer therapeutic effect. We tested this hypothesis using a combination of two nutraceutical agents S-adenosylmethionine (SAM) and Vitamin D (Vit. D) prohormone [25-hydroxyvitamin D; ‘25(OH)D’] that are individually known to exert distinct changes in the expression of genes involved in tumor growth and metastasis. Our results show that both SAM and 25(OH)D monotherapy significantly reduced proliferation and clonogenic survival of a panel of breast cancer cell lines in vitro and inhibited tumor growth, lung metastasis, and breast tumor cell colonization to the skeleton in vivo. However, these effects were significantly more pronounced in the combination setting. RNA-Sequencing revealed that the transcriptomic footprint on key cancer-related signaling pathways is broader in the combination setting than any of the monotherapies. Furthermore, comparison of the differentially expressed genes from our transcriptome analyses with publicly available cancer-related dataset demonstrated that the combination treatment 1234567890();,: upregulates genes from immune-related pathways that are otherwise downregulated in bone metastasis in vivo.
    [Show full text]
  • Downloaded from the UCSC PRO-Seq Library Preparation and Sequencing
    ARTICLE https://doi.org/10.1038/s41467-021-21687-2 OPEN Multi-omics analysis reveals contextual tumor suppressive and oncogenic gene modules within the acute hypoxic response ✉ ✉ Zdenek Andrysik1,2, Heather Bender1,2, Matthew D. Galbraith 1,2 & Joaquin M. Espinosa 1,2,3 Cellular adaptation to hypoxia is a hallmark of cancer, but the relative contribution of hypoxia- inducible factors (HIFs) versus other oxygen sensors to tumorigenesis is unclear. We employ 1234567890():,; a multi-omics pipeline including measurements of nascent RNA to characterize transcrip- tional changes upon acute hypoxia. We identify an immediate early transcriptional response that is strongly dependent on HIF1A and the kinase activity of its cofactor CDK8, includes indirect repression of MYC targets, and is highly conserved across cancer types. HIF1A drives this acute response via conserved high-occupancy enhancers. Genetic screen data indicates that, in normoxia, HIF1A displays strong cell-autonomous tumor suppressive effects through a gene module mediating mTOR inhibition. Conversely, in advanced malignancies, expression of a module of HIF1A targets involved in collagen remodeling is associated with poor prog- nosis across diverse cancer types. In this work, we provide a valuable resource for investi- gating context-dependent roles of HIF1A and its targets in cancer biology. 1 Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA. 2 Linda Crnic Institute for Down Syndrome, School of Medicine, University
    [Show full text]
  • Original Article Association of EGLN1 and EGLN3 Single-Nucleotide Polymorphisms with Chronic Obstructive Pulmonary Disease Risk in a Chinese Population
    Int J Clin Exp Med 2017;10(7):10866-10873 www.ijcem.com /ISSN:1940-5901/IJCEM0046332 Original Article Association of EGLN1 and EGLN3 single-nucleotide polymorphisms with chronic obstructive pulmonary disease risk in a Chinese population Si Fang*, Jinfan Qiu*, Huapeng Yu, Huizhen Fan, Xiping Wu, Zekui Fang, Qixiao Shen, Shuyu Chen Department of Respiration, Zhujiang Hospital, Southern Medical University, Guangzhou, China. *Equal contribu- tors. Received December 11, 2016; Accepted March 31, 2017; Epub July 15, 2017; Published July 30, 2017 Abstract: Hypoxia is implicated in the process of chronic inflammation, which is the characteristic pathogenesis of COPD (chronic obstructive pulmonary disease). EGLNs (EglN prolyl hydroxylase) that connect oxygen sensing to the activation of HIF-1 (hypoxia inducible factor-1) have been proven to play a role in the development of COPD. The ob- jective of this study was to explore the role of EGLN1 and EGLN3 SNPs (single-nucleotide polymorphisms) in suscep- tibility and development of COPD in a Chinese cohort. Seven SNPs were chosen from two genes (EGLN1 and EGLN3) in 217 cases and 447 controls. The relationship between SNPs and COPD risk was analyzed by using genetic model analysis. Among the seven SNPs, rs11156819 in EGLN3 exhibited a significant association with COPD risk with an OR of 1.392 (95% CI=1.085-1.787, P=0.009). In the log-additive model, the minor allele T of rs11156819 in EGLN3 significantly increased the risk of COPD (OR=1.58, 95% CI=1.14-2.19,P =0.006) after adjusting for gender, age and smoking status.
    [Show full text]
  • (12) Patent Application Publication (10) Pub. No.: US 2006/0128764 A1 Downes Et Al
    US 2006O128764A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2006/0128764 A1 DOWnes et al. (43) Pub. Date: Jun. 15, 2006 (54) NON-STEROIDAL FARNESOD X Publication Classification RECEPTOR MODULATORS AND METHODS FOR THE USE THEREOF (51) Int. Cl. A61K 31/4433 (2006.01) (75) Inventors: Michael R Downes, San Diego, CA A6II 3/44 (2006.01) (US); Ronald M Evans, La Jolla, CA A6II 3 L/353 (2006.01) (US) A6II 3/16 (2006.01) (52) U.S. Cl. ........................... 514/357: 514/456; 514/625 Correspondence Address: FOLEY & LARDNER LLP P.O. BOX 80278 (57) ABSTRACT SAN DIEGO, CA 92138-0278 (US) The efficient regulation of cholesterol synthesis, metabo (73) Assignee: THE SALK INSTITUTE FOR BIO lism, acquisition, and transport is an essential component of LOGICAL STUDES lipid homeostasis. The farnesoid X receptor (FXR) is a transcriptional sensor for bile acids, the primary product of (21) Appl. No.: 10/535,043 cholesterol metabolism. Accordingly, the development of potent, selective, Small molecule agonists, partial agonists, (22) PCT Fed: Nov. 14, 2003 and antagonists of FXR would be an important step in PCT No.: PCT/USO3/36137 further deconvoluting FXR physiology. In accordance with (86) the present invention, the identification of novel potent FXR Related U.S. Application Data activators is described. Two derivatives of invention com pounds, bearing stilbene or biaryl moieties, contain mem (60) Provisional application No. 60/426,664, filed on Nov. bers that are the most potent FXR agonists reported to date 15, 2002. in cell-based assays. These compounds are useful as chemi cal tools to further define the physiological role of FXR as (30) Foreign Application Priority Data well as therapeutic leads for the treatment of diseases linked to cholesterol, bile acids and their metabolism and homeo Sep.
    [Show full text]
  • The Prolyl Hydroxylase PHD3 Identifies Proinflammatory Macrophages and Its Expression Is Regulated by Activin A
    The Prolyl Hydroxylase PHD3 Identifies Proinflammatory Macrophages and Its Expression Is Regulated by Activin A This information is current as María M. Escribese, Elena Sierra-Filardi, Concha Nieto, of September 25, 2021. Rafael Samaniego, Carmen Sánchez-Torres, Takami Matsuyama, Elisabeth Calderon-Gómez, Miguel A. Vega, Azucena Salas, Paloma Sánchez-Mateos and Angel L. Corbí J Immunol published online 9 July 2012 http://www.jimmunol.org/content/early/2012/07/09/jimmun Downloaded from ol.1201064 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average by guest on September 25, 2021 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 © 2012 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published July 9, 2012, doi:10.4049/jimmunol.1201064 The Journal of Immunology The Prolyl Hydroxylase PHD3 Identifies Proinflammatory Macrophages and Its Expression Is Regulated by Activin A Marı´a M. Escribese,* Elena Sierra-Filardi,* Concha Nieto,* Rafael Samaniego,† Carmen Sa´nchez-Torres,‡ Takami Matsuyama,x Elisabeth Calderon-Go´mez,{ Miguel A.
    [Show full text]
  • Molecular Network-Based Identification of Competing Endogenous Rnas and Mrna Signatures That Predict Survival in Prostate Cancer
    Xu et al. J Transl Med (2018) 16:274 https://doi.org/10.1186/s12967-018-1637-x Journal of Translational Medicine RESEARCH Open Access Molecular network‑based identifcation of competing endogenous RNAs and mRNA signatures that predict survival in prostate cancer Ning Xu1,2, Yu‑Peng Wu2, Hu‑Bin Yin1, Xue‑Yi Xue2 and Xin Gou1* Abstract Background: The aim of the study is described the regulatory mechanisms and prognostic values of diferentially expressed RNAs in prostate cancer and construct an mRNA signature that predicts survival. Methods: The RNA profles of 499 prostate cancer tissues and 52 non-prostate cancer tissues from TCGA were ana‑ lyzed. The diferential expression of RNAs was examined using the edgeR package. Survival was analyzed by Kaplan– Meier method. microRNA (miRNA), messenger RNA (mRNA), and long non-coding RNA (lncRNA) networks from the miRcode database were constructed, based on the diferentially expressed RNAs between non-prostate and prostate cancer tissues. Results: A total of 773 lncRNAs, 1417 mRNAs, and 58 miRNAs were diferentially expressed between non-prostate and prostate cancer samples. The newly constructed ceRNA network comprised 63 prostate cancer-specifc lncRNAs, 13 miRNAs, and 18 mRNAs. Three of 63 diferentially expressed lncRNAs and 1 of 18 diferentially expressed mRNAs were signifcantly associated with overall survival in prostate cancer (P value < 0.05). After the univariate and multivari‑ ate Cox regression analyses, 4 mRNAs (HOXB5, GPC2, PGA5, and AMBN) were screened and used to establish a predic‑ tive model for the overall survival of patients. Our ROC curve analysis revealed that the 4-mRNA signature performed well.
    [Show full text]