Early Growth Response 1 Acts As a Tumor Suppressor in Vivo and in Vitro Via Regulation of P53

Total Page:16

File Type:pdf, Size:1020Kb

Early Growth Response 1 Acts As a Tumor Suppressor in Vivo and in Vitro Via Regulation of P53 Research Article Early Growth Response 1 Acts as a Tumor Suppressor In vivo and In vitro via Regulation of p53 Anja Krones-Herzig,1 Shalu Mittal,1 Kelly Yule,1 Hongyan Liang,2 Chris English,1 Rafael Urcis,1 Tarun Soni,1 Eileen D. Adamson,2 and Dan Mercola1,3 1Sidney Kimmel Cancer Center, San Diego, California and 2The Burnham Institute; 3Cancer Center, University of California at San Diego, La Jolla, California Abstract human tumor cell lines express little or no Egr1 in contrast to their normal counterparts (9–12). Furthermore, Egr1 is decreased or The early growth response 1 (Egr1) gene is a transcription factor that acts as both a tumor suppressor and a tumor undetectable in small cell lung tumors, human breast tumors promoter. Egr1-null mouse embryo fibroblasts bypass repli- (11, 13), and human gliomas (12). Reexpression of Egr1 in human tumor cells inhibits transformation. The mechanism of suppression cative senescence and exhibit a loss of DNA damage response h and an apparent immortal growth, suggesting loss of p53 involves the direct induction of TGF- 1 leading to an autocrine- functions. Stringent expression analysis revealed 266 tran- mediated suppression of transformation (8), increased fibronectin, scripts with >2-fold differential expression in Egr1-null mouse and plasminogen activator inhibitor (9). Egr1 also has been embryo fibroblasts, including 143 known genes. Of the 143 implicated in the regulation of p53 in human melanoma cells genes, program-assisted searching revealed 66 informative leading to apoptosis (14–16), and the proapoptotic tumor genes linked to Egr1. All 66 genes could be placed on a single suppressor gene PTEN also is directly regulated by Egr1 (17). Recently, we have identified and established a new role of Egr1 as regulatory network consisting of three branch points of known a ‘‘gatekeeper’’ of p53-dependent growth-regulatory mechanisms in Egr1 target genes: TGFb1, IL6,andIGFI. Moreover, 19 replicative senescence and cell growth (18). This result was additional genes that are known targets of p53 were identified, revealed by examination of primary mouse embryo fibroblasts indicating that p53 is a fourth branch point. Electrophoretic (MEF) isolated from Egr1-null mice developed previously by mobility shift assay as well as chromatin immunoprecipitation Lee et al. (19) and Topilko et al. (20). Egr1-null cells from either confirmed that p53 is a direct target of Egr1. Because deficient strain express no Egr1 protein and much reduced p53 protein. p53 expression causes tumors in mice, we tested the role of These cells completely bypass replicative senescence in culture, Egr1 in a two-step skin carcinogenesis study (144 mice) that thereby appearing to be immortal without passing through a revealed a uniformly accelerated development of skin tumors ‘‘crisis’’ stage. Moreover, these cells fail to arrest following treatment in Egr1-null mice (P < 0.005). These studies reveal a new role with DNA-damaging agents, such as g-irradiation (18) and 12-O- for Egr1 as an in vivo tumor suppressor. (Cancer Res 2005; tetradecanoylphorbol-13-acetate (TPA; see below). Replicative 65(12): 5133-43) senescence and the DNA damage response of the Egr1-null cells may be restored by infection with an Egr1-expressing retrovirus in Introduction contrast to p53-null MEFs. In the Egr1-null cells, p53 is not The transcription factor early growth response 1 (Egr1)isa expressed and the p53 gene remains entirely free of mutation to member of the immediate-early gene family and encodes a 59-kDa high passage numbers. In contrast, the wild-type (WT) cells express phosphoprotein observed at f80 kDa by electrophoresis. Egr1 is normal levels of p53, arrest at low passage in normal tissue culture, involved in the regulation of cell growth and differentiation in and undergo crisis. WT cells that survive crisis inevitably exhibit response to signals, such as mitogens, growth factors, and stress mutations of the p53 gene. We speculated that Egr1 exerts a stimuli (1–4). Egr1 has a COOH-terminal DNA-binding domain gatekeeper function over p53, thereby allowing the null cells to consisting of three zinc fingers that regulates transcription through bypass replicative senescence and avoid the mutation association GC-rich elements. The 9-bp DNA consensus-binding sequence with the survival of crisis. However, the exact mechanism whereby GCG(G/T)GGGCG has been identified in the promoter of several Egr1 functions in the regulation of p53-dependent growth arrest is growth-regulatory genes (5). Besides the highly conserved DNA- unknown, and there is no evidence that Egr1 mediates these roles binding domain, Egr1 also contains a nuclear localization signal, in a whole organism. two activator domains, and one repressor domain (6). In addition, Here, we examined gene expression in the genetically defined Egr1 binds to regulatory proteins called NAB1 and NAB2 (nerve Egr1-deficient MEFs. These cells exhibit at least 266 significant growth factor-I A-binding protein) that repress its transcriptional differences in transcript expression compared with WT cells, many activity (7). of which could be recognized as Egr1 target genes or downstream Analysis of certain human tumor cells and tissues indicate that signal transduction mediators. Indeed, an extended regulatory Egr1 exhibits prominent tumor suppressor function (5, 8–11). Many network based on four ‘‘nodes’’ could be constructed. Three nodes are upstream of numerous recognized Egr1-regulated genes. However, a fourth node, and potential Egr1 target gene, p53,is Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). upstream of at least 19 genes not previously recognized as Egr1- A. Krones-Herzig and S. Mittal contributed equally to this work. regulated genes but are well-recognized mediators of p53 function. Requests for reprints: Dan Mercola, Sidney Kimmel Cancer Center, 10835 Altman Electrophoretic mobility shift assay (EMSA) and chromatin Row, San Diego, CA 92121. Phone: 858-450-5990, ext. 370; Fax: 858-450-3251; E-mail: [email protected]. immunoprecipitation (ChIP) experiments directly confirmed the I2005 American Association for Cancer Research. binding of the p53 promoter by Egr1 in cells both in vitro and www.aacrjournals.org 5133 Cancer Res 2005; 65: (12). June 15, 2005 Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2005 American Association for Cancer Research. Cancer Research in vivo. Moreover, TPA-induced skin tumors appeared rapidly in TGFb1, TGFb1I, GRO1, IGFII, THSP2, and procollagen XI. Primers were Egr1-null mice compared with WT and heterozygous littermates. selected for these genes with the aid of Primer Express software (Applied These studies indicate that Egr1 regulates an extensive network of Biosystems) and are available on request. The results were normalized to genes that include direct regulation of p53 at the level of RNA and the relative amounts of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). protein expression. Our observations indicate that this mechanism Chromatin immunoprecipitation assay. WT and null MEFs and may function in vitro and in vivo to control DNA damage–induced human prostate cancer DU145 cells were treated with 1% formaldehyde for growth arrest. Loss of this mechanism in vivo may be related to 20 minutes to cross-link protein to binding sites on DNA. Egr1-containing increased skin tumor formation in Egr1-deficient mice. fragments were recovered by immunoprecipitation as described earlier (24). The ChIP-captured DNA was then screened for p53 promoter fragments by PCR analysis using the following p53 specific primers: (mouse) 5V- Materials and Methods GTAGAGTAAGCCCCCGGAAG-3V and 5V-GGTTACCGGGATTCAAAACA-3V Cells and cell culture. MEFs were prepared as described earlier (21) (the amplified fragments correspond to À925 and À424 region of the mouse from 19-day-old embryos from Egr1 WT, Egr1-null, and Egr1 heterozygous p53 promoter from AF287146); (human) 5V-TGGGAGTTGTAGTCTGAA- mice kindly provided by Dr. J. Milbrandt (Washington University Medical CGCTTC-3V and 5V-GAGAAGCTCAAAACTTTTAGCGCC-3V (the amplified School, St. Louis, MO) (19). Cultures of WT and Egr1-null MEFs were fragments correspond to À693 to +89 region of the human p53 promoter established and maintained in parallel for >60 passages. The predicted from X54156). Genomic DNA was used as a control for the amplification genotype and expression properties of the MEFs derived from Egr1-null and efficiency of each primer pair. heterozygous mice were confirmed by PCR-based analysis of DNA and RNA Electrophoretic mobility shift assay. WT and Egr1-null MEFs were and by Western analysis for protein expression as described previously (18). grown in tissue culture to confluence, incubated in serum-free medium for Oligonucleotide microarray analysis. Total cellular RNA was isolated 1 hour, and then supplemented with 20% fetal bovine serum for 1 hour. by using RNeasy kits (Qiagen Inc., Valencia, CA) and quantified. The total Cells were then lysed in hypotonic buffer (10 mmol/L HEPES, 25 mmol/L RNA quality was determined using the standard Affymetrix protocol. For KCl, 1 mmol/L EDTA, 1 mmol/L EGTA) containing 1:100 protease hybridization, total RNA (10 Ag) from WT or Egr1-null MEFs was reverse inhibitor cocktail (Sigma, St. Louis, MO) on ice for 10 minutes. Samples transcribed using an oligo(dT) primer harboring a T7 RNA polymerase were centrifuged at 1,500 rpm for 7 minutes and the pellets were promoter at the 5V end (Genset, San Diego, CA). Following second-strand incubated on ice for 30 minutes in nuclear extract buffer (50 mmol/L Tris- synthesis, biotinylated cRNA probes were produced. The probes were then HCl, 420 mmol/L KCl, 5 mmol/L MgCl2, 0.1 mmol/L EDTA) with 1:100 fragmented and hybridized to Affymetrix MGU75Av2 array representing protease inhibitor cocktail. The samples were centrifuged at 5,000 rpm for 12,566 mouse transcripts using the standard Affymetrix protocol (http:// 30 minutes at 4jC and the supernatants were removed for protein www.affymetrix.com).
Recommended publications
  • Table S1. List of Proteins in the BAHD1 Interactome
    Table S1. List of proteins in the BAHD1 interactome BAHD1 nuclear partners found in this work yeast two-hybrid screen Name Description Function Reference (a) Chromatin adapters HP1α (CBX5) chromobox homolog 5 (HP1 alpha) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins (20-23) HP1β (CBX1) chromobox homolog 1 (HP1 beta) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins HP1γ (CBX3) chromobox homolog 3 (HP1 gamma) Binds histone H3 methylated on lysine 9 and chromatin-associated proteins MBD1 methyl-CpG binding domain protein 1 Binds methylated CpG dinucleotide and chromatin-associated proteins (22, 24-26) Chromatin modification enzymes CHD1 chromodomain helicase DNA binding protein 1 ATP-dependent chromatin remodeling activity (27-28) HDAC5 histone deacetylase 5 Histone deacetylase activity (23,29,30) SETDB1 (ESET;KMT1E) SET domain, bifurcated 1 Histone-lysine N-methyltransferase activity (31-34) Transcription factors GTF3C2 general transcription factor IIIC, polypeptide 2, beta 110kDa Required for RNA polymerase III-mediated transcription HEYL (Hey3) hairy/enhancer-of-split related with YRPW motif-like DNA-binding transcription factor with basic helix-loop-helix domain (35) KLF10 (TIEG1) Kruppel-like factor 10 DNA-binding transcription factor with C2H2 zinc finger domain (36) NR2F1 (COUP-TFI) nuclear receptor subfamily 2, group F, member 1 DNA-binding transcription factor with C4 type zinc finger domain (ligand-regulated) (36) PEG3 paternally expressed 3 DNA-binding transcription factor with
    [Show full text]
  • An Integrative Study
    Published OnlineFirst January 12, 2010; DOI: 10.1158/1535-7163.MCT-09-0321 Molecular Spotlight on Molecular Profiling Cancer Therapeutics Multifactorial Regulation of E-Cadherin Expression: An Integrative Study William C. Reinhold1, Mark A. Reimers1,2, Philip Lorenzi1,3, Jennifer Ho1, Uma T. Shankavaram1,4, Micah S. Ziegler1, Kimberly J. Bussey1,5, Satoshi Nishizuka1,6, Ogechi Ikediobi1,7, Yves G. Pommier1, and John N. Weinstein1,3 Abstract E-cadherin (E-cad) is an adhesion molecule associated with tumor invasion and metastasis. Its down- regulation is associated with poor prognosis for many epithelial tumor types. We have profiled E-cad in the NCI-60 cancer cell lines at the DNA, RNA, and protein levels using six different microarray platforms plus bisulfite sequencing. Here we consider the effects on E-cad expression of eight potential regulatory factors: E-cad promoter DNA methylation, the transcript levels of six transcriptional repressors (SNAI1, SNAI2, TCF3, TCF8, TWIST1, and ZFHX1B), and E-cad DNA copy number. Combined bioinformatic and pharmacological analyses indicate the following ranking of influence on E-cad expression: (1) E-cad pro- moter methylation appears predominant, is strongly correlated with E-cad expression, and shows a 20% to 30% threshold above which E-cad expression is silenced; (2) TCF8 expression levels correlate with (−0.62) and predict (P < 0.00001) E-cad expression; (3) SNAI2 and ZFHX1B expression levels correlate positively with each other (+0.83) and also correlate with (−0.32 and −0.30, respectively) and predict (P =0.03and 0.01, respectively) E-cad expression; (4) TWIST1 correlates with (−0.34) but does not predict E-cad expres- sion; and (5) SNAI1 expression, TCF3 expression, and E-cad DNA copy number do not correlate with or predict E-cad expression.
    [Show full text]
  • New Approaches to Functional Process Discovery in HPV 16-Associated Cervical Cancer Cells by Gene Ontology
    Cancer Research and Treatment 2003;35(4):304-313 New Approaches to Functional Process Discovery in HPV 16-Associated Cervical Cancer Cells by Gene Ontology Yong-Wan Kim, Ph.D.1, Min-Je Suh, M.S.1, Jin-Sik Bae, M.S.1, Su Mi Bae, M.S.1, Joo Hee Yoon, M.D.2, Soo Young Hur, M.D.2, Jae Hoon Kim, M.D.2, Duck Young Ro, M.D.2, Joon Mo Lee, M.D.2, Sung Eun Namkoong, M.D.2, Chong Kook Kim, Ph.D.3 and Woong Shick Ahn, M.D.2 1Catholic Research Institutes of Medical Science, 2Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul; 3College of Pharmacy, Seoul National University, Seoul, Korea Purpose: This study utilized both mRNA differential significant genes of unknown function affected by the display and the Gene Ontology (GO) analysis to char- HPV-16-derived pathway. The GO analysis suggested that acterize the multiple interactions of a number of genes the cervical cancer cells underwent repression of the with gene expression profiles involved in the HPV-16- cancer-specific cell adhesive properties. Also, genes induced cervical carcinogenesis. belonging to DNA metabolism, such as DNA repair and Materials and Methods: mRNA differential displays, replication, were strongly down-regulated, whereas sig- with HPV-16 positive cervical cancer cell line (SiHa), and nificant increases were shown in the protein degradation normal human keratinocyte cell line (HaCaT) as a con- and synthesis. trol, were used. Each human gene has several biological Conclusion: The GO analysis can overcome the com- functions in the Gene Ontology; therefore, several func- plexity of the gene expression profile of the HPV-16- tions of each gene were chosen to establish a powerful associated pathway, identify several cancer-specific cel- cervical carcinogenesis pathway.
    [Show full text]
  • Mediator of DNA Damage Checkpoint 1 (MDC1) Is a Novel Estrogen Receptor Co-Regulator in Invasive 6 Lobular Carcinoma of the Breast 7 8 Evelyn K
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.16.423142; this version posted December 16, 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 4.0 International license. 1 Running Title: MDC1 co-regulates ER in ILC 2 3 Research article 4 5 Mediator of DNA damage checkpoint 1 (MDC1) is a novel estrogen receptor co-regulator in invasive 6 lobular carcinoma of the breast 7 8 Evelyn K. Bordeaux1+, Joseph L. Sottnik1+, Sanjana Mehrotra1, Sarah E. Ferrara2, Andrew E. Goodspeed2,3, James 9 C. Costello2,3, Matthew J. Sikora1 10 11 +EKB and JLS contributed equally to this project. 12 13 Affiliations 14 1Dept. of Pathology, University of Colorado Anschutz Medical Campus 15 2Biostatistics and Bioinformatics Shared Resource, University of Colorado Comprehensive Cancer Center 16 3Dept. of Pharmacology, University of Colorado Anschutz Medical Campus 17 18 Corresponding author 19 Matthew J. Sikora, PhD.; Mail Stop 8104, Research Complex 1 South, Room 5117, 12801 E. 17th Ave.; Aurora, 20 CO 80045. Tel: (303)724-4301; Fax: (303)724-3712; email: [email protected]. Twitter: 21 @mjsikora 22 23 Authors' contributions 24 MJS conceived of the project. MJS, EKB, and JLS designed and performed experiments. JLS developed models 25 for the project. EKB, JLS, SM, and AEG contributed to data analysis and interpretation. SEF, AEG, and JCC 26 developed and performed informatics analyses. MJS wrote the draft manuscript; all authors read and revised the 27 manuscript and have read and approved of this version of the manuscript.
    [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 Direction of Cross Affects Obesity After Puberty in Male but Not Female
    Kärst et al. BMC Genomics (2015) 16:904 DOI 10.1186/s12864-015-2164-2 RESEARCH ARTICLE Open Access The direction of cross affects obesity after puberty in male but not female offspring Stefan Kärst1†, Danny Arends1†, Sebastian Heise1†, Jan Trost1, Marie-Laure Yaspo2, Vyacheslav Amstislavskiy2, Thomas Risch2, Hans Lehrach2 and Gudrun A. Brockmann1* Abstract Background: We investigated parent-of-origin and allele-specific expression effects on obesity and hepatic gene expression in reciprocal crosses between the Berlin Fat Mouse Inbred line (BFMI) and C57Bl/6NCrl (B6N). Results: We found that F1-males with a BFMI mother developed 1.8 times more fat mass on a high fat diet at 10 weeks than F1-males of a BFMI father. The phenotype was detectable from six weeks on and was preserved after cross-fostering. RNA-seq data of liver provided evidence for higher biosynthesis and elongation of fatty acids (p = 0.00635) in obese male offspring of a BFMI mother versus lean offspring of a BFMI father. Furthermore, fatty acid degradation (p = 0.00198) and the peroxisome pathway were impaired (p = 0.00094). The circadian rhythm was affected as well (p = 0.00087). Among the highest up-regulated protein coding genes in obese males were Acot4 (1.82 fold, p = 0.022), Cyp4a10 (1.35 fold, p = 0.026) and Cyp4a14 (1.32 fold, p = 0.012), which hydroxylize fatty acids and which are known to be increased in liver steatosis. Obese males showed lower expression of the genetically imprinted and paternally expressed 3 (Peg3) gene (0.31 fold, p = 0.046) and higher expression of the androgen receptor (Ar) gene (2.38 fold, p = 0.068).
    [Show full text]
  • Datasheet: VMA00210 Product Details
    Datasheet: VMA00210 Description: MOUSE ANTI NAB1 Specificity: NAB1 Format: Purified Product Type: PrecisionAb™ Monoclonal Clone: OTI1D2 Isotype: IgG1 Quantity: 100 µl Product Details Applications This product has been reported to work in the following applications. This information is derived from testing within our laboratories, peer-reviewed publications or personal communications from the originators. Please refer to references indicated for further information. For general protocol recommendations, please visit www.bio-rad-antibodies.com/protocols. Yes No Not Determined Suggested Dilution Western Blotting 1/1000 PrecisionAb antibodies have been extensively validated for the western blot application. The antibody has been validated at the suggested dilution. Where this product has not been tested for use in a particular technique this does not necessarily exclude its use in such procedures. Further optimization may be required dependant on sample type. Target Species Human Product Form Purified IgG - liquid Preparation Mouse monoclonal antibody purified by affinity chromatography from ascites Buffer Solution Phosphate buffered saline Preservative 0.09% Sodium Azide (NaN3) Stabilisers 1% Bovine Serum Albumin 50% Glycerol Immunogen Recombinant protein corresponding to amino acids 237-487 of human NAB1 (NP_005957) produced in E. coli External Database UniProt: Links Q13506 Related reagents Entrez Gene: 4664 NAB1 Related reagents Specificity Mouse anti Human NAB1 antibody recognizes the NGFI-A-binding protein 1, also known as Page 1 of 2 EGR-1-binding protein 1, EGR1 binding protein 1 and transcriptional regulatory protein p54. Mouse anti Human NAB1 antibody detects a band of 54 kDa. The antibody has been extensively validated for western blotting using whole cell lysates. Western Blotting Anti NAB1 detects a band of approximately 54 kDa in Jurkat cell lysates Instructions For Use Please refer to the PrecisionAb western blotting protocol.
    [Show full text]
  • Cellular and Molecular Signatures in the Disease Tissue of Early
    Cellular and Molecular Signatures in the Disease Tissue of Early Rheumatoid Arthritis Stratify Clinical Response to csDMARD-Therapy and Predict Radiographic Progression Frances Humby1,* Myles Lewis1,* Nandhini Ramamoorthi2, Jason Hackney3, Michael Barnes1, Michele Bombardieri1, Francesca Setiadi2, Stephen Kelly1, Fabiola Bene1, Maria di Cicco1, Sudeh Riahi1, Vidalba Rocher-Ros1, Nora Ng1, Ilias Lazorou1, Rebecca E. Hands1, Desiree van der Heijde4, Robert Landewé5, Annette van der Helm-van Mil4, Alberto Cauli6, Iain B. McInnes7, Christopher D. Buckley8, Ernest Choy9, Peter Taylor10, Michael J. Townsend2 & Costantino Pitzalis1 1Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Departments of 2Biomarker Discovery OMNI, 3Bioinformatics and Computational Biology, Genentech Research and Early Development, South San Francisco, California 94080 USA 4Department of Rheumatology, Leiden University Medical Center, The Netherlands 5Department of Clinical Immunology & Rheumatology, Amsterdam Rheumatology & Immunology Center, Amsterdam, The Netherlands 6Rheumatology Unit, Department of Medical Sciences, Policlinico of the University of Cagliari, Cagliari, Italy 7Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK 8Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham B15 2WB, UK 9Institute of
    [Show full text]
  • Decorin Causes Autophagy in Endothelial Cells Via Peg3 PNAS PLUS
    Decorin causes autophagy in endothelial cells via Peg3 PNAS PLUS Simone Buraschia,b,1, Thomas Neilla,b,1, Atul Goyala,b, Chiara Poluzzia,b, James Smythiesa,b, Rick T. Owensc, Liliana Schaeferd, Annabel Torresa,b, and Renato V. Iozzoa,b,2 aDepartment of Pathology, Anatomy and Cell Biology, and the bCell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107; cLifeCell Corporation, Branchburg, NJ 08876; and dPharmazentrum Frankfurt, Goethe University, 60590 Frankfurt, Germany Edited by Carlo M. Croce, The Ohio State University, Columbus, Ohio, and approved May 29, 2013 (received for review March 27, 2013) Soluble decorin affects the biology of several receptor tyrosine capable of significantly inhibiting neovascularization of triple- kinases by triggering receptor internalization and degradation. We negative basal cell breast carcinomas (23). found that decorin induced paternally expressed gene 3 (Peg3), an During preclinical studies focusing on identifying decorin- imprinted tumor suppressor gene, and that Peg3 relocated into induced genes in vivo, we found that systemic delivery of decorin autophagosomes labeled by Beclin 1 and microtubule-associated protein core to mice carrying orthotopic mammary carcinoma light chain 3. Decorin evoked Peg3-dependent autophagy in both xenografts induced expression of a small subset of genes (24). microvascular and macrovascular endothelial cells leading to sup- Notably, these genes were induced exclusively in the tumor stroma pression of angiogenesis. Peg3 coimmunoprecipitated with Beclin 1 of mouse origin but not in the mammary carcinomas of human and LC3 and was required for maintaining basal levels of Beclin 1. origin (24). One of the most up-regulated genes was Paternally Decorin, via Peg3, induced transcription of Beclin 1 and microtu- expressed gene 3 (Peg3), an imprinted putative tumor-suppressor bule-associated protein 1 light chain 3 alpha genes, thereby leading gene frequently silenced by promoter hypermethylation and/or to a protracted autophagic program.
    [Show full text]
  • Early Growth Response 1 Acts As a Tumor Suppressor in Vivo and in Vitro Via Regulation of P53
    Research Article Early Growth Response 1 Acts as a Tumor Suppressor In vivo and In vitro via Regulation of p53 Anja Krones-Herzig,1 Shalu Mittal,1 Kelly Yule,1 Hongyan Liang,2 Chris English,1 Rafael Urcis,1 Tarun Soni,1 Eileen D. Adamson,2 and Dan Mercola1,3 1Sidney Kimmel Cancer Center, San Diego, California and 2The Burnham Institute; 3Cancer Center, University of California at San Diego, La Jolla, California Abstract human tumor cell lines express little or no Egr1 in contrast to their normal counterparts (9–12). Furthermore, Egr1 is decreased or The early growth response 1 (Egr1) gene is a transcription factor that acts as both a tumor suppressor and a tumor undetectable in small cell lung tumors, human breast tumors promoter. Egr1-null mouse embryo fibroblasts bypass repli- (11, 13), and human gliomas (12). Reexpression of Egr1 in human tumor cells inhibits transformation. The mechanism of suppression cative senescence and exhibit a loss of DNA damage response h and an apparent immortal growth, suggesting loss of p53 involves the direct induction of TGF- 1 leading to an autocrine- functions. Stringent expression analysis revealed 266 tran- mediated suppression of transformation (8), increased fibronectin, scripts with >2-fold differential expression in Egr1-null mouse and plasminogen activator inhibitor (9). Egr1 also has been embryo fibroblasts, including 143 known genes. Of the 143 implicated in the regulation of p53 in human melanoma cells genes, program-assisted searching revealed 66 informative leading to apoptosis (14–16), and the proapoptotic tumor genes linked to Egr1. All 66 genes could be placed on a single suppressor gene PTEN also is directly regulated by Egr1 (17).
    [Show full text]
  • Supplementary Materials
    Supplementary materials Supplementary Table S1: MGNC compound library Ingredien Molecule Caco- Mol ID MW AlogP OB (%) BBB DL FASA- HL t Name Name 2 shengdi MOL012254 campesterol 400.8 7.63 37.58 1.34 0.98 0.7 0.21 20.2 shengdi MOL000519 coniferin 314.4 3.16 31.11 0.42 -0.2 0.3 0.27 74.6 beta- shengdi MOL000359 414.8 8.08 36.91 1.32 0.99 0.8 0.23 20.2 sitosterol pachymic shengdi MOL000289 528.9 6.54 33.63 0.1 -0.6 0.8 0 9.27 acid Poricoic acid shengdi MOL000291 484.7 5.64 30.52 -0.08 -0.9 0.8 0 8.67 B Chrysanthem shengdi MOL004492 585 8.24 38.72 0.51 -1 0.6 0.3 17.5 axanthin 20- shengdi MOL011455 Hexadecano 418.6 1.91 32.7 -0.24 -0.4 0.7 0.29 104 ylingenol huanglian MOL001454 berberine 336.4 3.45 36.86 1.24 0.57 0.8 0.19 6.57 huanglian MOL013352 Obacunone 454.6 2.68 43.29 0.01 -0.4 0.8 0.31 -13 huanglian MOL002894 berberrubine 322.4 3.2 35.74 1.07 0.17 0.7 0.24 6.46 huanglian MOL002897 epiberberine 336.4 3.45 43.09 1.17 0.4 0.8 0.19 6.1 huanglian MOL002903 (R)-Canadine 339.4 3.4 55.37 1.04 0.57 0.8 0.2 6.41 huanglian MOL002904 Berlambine 351.4 2.49 36.68 0.97 0.17 0.8 0.28 7.33 Corchorosid huanglian MOL002907 404.6 1.34 105 -0.91 -1.3 0.8 0.29 6.68 e A_qt Magnogrand huanglian MOL000622 266.4 1.18 63.71 0.02 -0.2 0.2 0.3 3.17 iolide huanglian MOL000762 Palmidin A 510.5 4.52 35.36 -0.38 -1.5 0.7 0.39 33.2 huanglian MOL000785 palmatine 352.4 3.65 64.6 1.33 0.37 0.7 0.13 2.25 huanglian MOL000098 quercetin 302.3 1.5 46.43 0.05 -0.8 0.3 0.38 14.4 huanglian MOL001458 coptisine 320.3 3.25 30.67 1.21 0.32 0.9 0.26 9.33 huanglian MOL002668 Worenine
    [Show full text]
  • Quantitative SUMO Proteomics Reveals the Modulation of Several
    www.nature.com/scientificreports OPEN Quantitative SUMO proteomics reveals the modulation of several PML nuclear body associated Received: 10 October 2017 Accepted: 28 March 2018 proteins and an anti-senescence Published: xx xx xxxx function of UBC9 Francis P. McManus1, Véronique Bourdeau2, Mariana Acevedo2, Stéphane Lopes-Paciencia2, Lian Mignacca2, Frédéric Lamoliatte1,3, John W. Rojas Pino2, Gerardo Ferbeyre2 & Pierre Thibault1,3 Several regulators of SUMOylation have been previously linked to senescence but most targets of this modifcation in senescent cells remain unidentifed. Using a two-step purifcation of a modifed SUMO3, we profled the SUMO proteome of senescent cells in a site-specifc manner. We identifed 25 SUMO sites on 23 proteins that were signifcantly regulated during senescence. Of note, most of these proteins were PML nuclear body (PML-NB) associated, which correlates with the increased number and size of PML-NBs observed in senescent cells. Interestingly, the sole SUMO E2 enzyme, UBC9, was more SUMOylated during senescence on its Lys-49. Functional studies of a UBC9 mutant at Lys-49 showed a decreased association to PML-NBs and the loss of UBC9’s ability to delay senescence. We thus propose both pro- and anti-senescence functions of protein SUMOylation. Many cellular mechanisms of defense have evolved to reduce the onset of tumors and potential cancer develop- ment. One such mechanism is cellular senescence where cells undergo cell cycle arrest in response to various stressors1,2. Multiple triggers for the onset of senescence have been documented. While replicative senescence is primarily caused in response to telomere shortening3,4, senescence can also be triggered early by a number of exogenous factors including DNA damage, elevated levels of reactive oxygen species (ROS), high cytokine signa- ling, and constitutively-active oncogenes (such as H-RAS-G12V)5,6.
    [Show full text]