Mechanisms of Spontaneous Melanoma Regression in Melim Pigs Florian Rambow

Total Page:16

File Type:pdf, Size:1020Kb

Mechanisms of Spontaneous Melanoma Regression in Melim Pigs Florian Rambow Mechanisms of Spontaneous Melanoma Regression in MeLiM Pigs Florian Rambow To cite this version: Florian Rambow. Mechanisms of Spontaneous Melanoma Regression in MeLiM Pigs. Life Sciences [q-bio]. Université de Versailles-Saint Quentin en Yvelines; Ruprecht-Karls-Universität Heidelberg, 2008. English. tel-00433225 HAL Id: tel-00433225 https://tel.archives-ouvertes.fr/tel-00433225 Submitted on 18 Nov 2009 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. PhD Thesis en Co-tutelle to obtain the degree of DOCTEUR EN SCIENCES DE L’UNIVERSITÉ DE VERSAILLES SAINT-QUENTIN-EN-YVELINES and DOCTOR SCIENTIARUM HUMANARUM of the UNIVERSITY of HEIDELBERG (Medical Faculty) Discipline: Molecular Biology Presented and defended by Florian RAMBOW May 2008 Mechanisms of Spontaneous Melanoma Regression in MeLiM Pigs Thesis Director: P. CHARDON Jury: Referee: Prof Dr M. Groenen Referee: Dr A. Prévost-Blondel Examiner: Prof Dr B. Mignotte Examiner: Prof Dr F. Velten Examiner: Dr P. Chardon “It is not the strongest of the species that survives, nor the most intelligent, but the one most responsive to change.” (Charles Darwin) Dedicated to my parents Abstract Cutaneous melanoma, the most aggressive form of skin cancer, is a tumor originating from melanocytes with rapidly increasing incidence. Patients with advanced disease have a poor prognosis since melanoma is mostly resistant to current therapies. Therefore, the development of novel strategies for preventing and treating melanoma is important. To explore novel therapies we need to find appropriate targets and for that knowledge about the biology of melanoma is important. One elegant way to do so, is to study the natural but rare phenomenon of spontaneous melanoma regression. Unfortunately, complete spontaneous regression of advanced melanoma is extremely rare, and therefore hard to investigate. The Melanoblastoma-bearing Libechov Minipig (MeLiM) is a great opportunity to study spontaneous tumor regression. MeLiM pigs, suffering from hereditary melanoma, develop tumors naturally and regress them completely without external influence. Mechanisms leading to spontaneous melanoma regression are poorly understood at present. Therefore the overall objective of this PhD was to investigate mechanisms of spontaneous melanoma regression in MeLiM pigs. In an initial study we gained a global overview of differentially expressed genes between a growing and regressing tumor by conducting Subtractive Suppression Hybridization (SSH). We also performed SSH analysis of cell cultures isolated from growing and regressing melanoma to be able to distinguish between expressional changes induced by tumor microenvironment. Only a few genes were in common between these two SSH analyses. Thus we focused on the SSH results obtained from tumor tissue. We were able to identify a gene signature for growing melanoma that demonstrated common genes with human progressive melanoma (TYR, MITF, MLANA, SDCBP, SILV, TYRP and ZFP106). Genes overexpressed at the beginning of regression were mainly involved in functional classes such as differentiation, immune system, cell cycle arrest, and tumor suppression. Two genes, CD9 and RARRES1, showed a strong upregulation during early melanoma regression on the mRNA and protein level whereas CD9 is a motility-related protein and RARRES1 a putative tumor suppressor. Since spontaneous regression is a dynamic process, we performed time dependent gene expression profiling using DNA chips in a second study. We identified significant gene signatures at different stages of regression that were able to explain the corresponding phenotype. Identified gene signatures were mainly involved in the immune response, the cell cycle, and melanocyte differentiation/pigmentation processes. Interestingly, we showed an early downregulation of cell cycle related genes that could play a role in regression. Also, we identified different immune gene signatures, suggesting a major role of the host’s immune system in eradicating melanoma cells. By using immunohistology and flow cytometry we characterized tumor infiltrating cells of the innate and adaptive immune system. During regression the infiltration occurred in two phases: an early phase consisting of antigen presenting cells (SWC3+), followed by a later infiltration of mainly cytotoxic T lymphocytes (CD8+). Furthermore the regression process is accompanied by the presence of big highly pigmented cells that we began to characterize. This work allowed us to point out similarities between the pig and human melanoma on the transcriptomic level along with the already observed parallels on the clinical and genetic level. These findings underline the utility for this model to study human melanoma. This work helped also to decipher the process of spontaneous regression on a cellular and molecular level. In this way we observed an early mitotic arrest of tumor cells, linked to regression, that has not been described so far, and an implication of the immune system. If the immune response is the real inducer of regression needs to be verified. Already, this work contributes to human anti- melanoma therapy as it provides potential targets that could be used for developing new strategies. Résumé : Le mélanome est une tumeur originaire des mélanocytes; c’est la forme la plus agressive des cancers cutanés. Son incidence s’accroît régulièrement. De plus sa résistance aux traitements actuels (chimiothérapie, radiothérapie et immunothérapie) ne laisse qu’une faible espérance de survie aux patients présentant un stade avancé de la maladie. Aujourd’hui il est nécessaire de développer de nouveaux traitements ciblés sur la cellule tumorale. Une stratégie originale consisterait à étudier le phénomène naturel de régression spontanée des mélanomes. Malheureusement chez l’homme la régression n’est que partielle et l’extrême rareté des cas de régression totale à un stade avancé de la maladie rend impossible son étude. Cependant le modèle porcin de mélanome cutané (MeLiM) présente une opportunité unique d’étudier ces mécanismes complexes de régression spontanée. En effet, les minis porcs MeLiM developpent des mélanomes héréditaires qui régressent totalement et spontanément, indépendamment de tous facteurs externes. Actuellement, les mécanismes responsables de cette régression spontanée sont peu ou mal connus. C’est pourquoi l’objectif principal de cette thèse a été l’étude des mécanismes de la régression spontanée du mélanome dans ce modèle animal. Dans un premier temps, grace à la technique d’hybridation suppressive soustractive (HSS) nous avons comparé le transcriptome d’une tumeur en croissance et d’une tumeur en tout début de régression. Dans le but de distinguer les signaux provenant du microenvironnement tumoral de ceux propres à la cellule tumorale, nous avons également réalisé une HSS à partir des cellules isolées d’une tumeur en progression et d’une tumeur au début de la régression. Nous avons trouvé très peu de gènes en commun entre les deux HSS. Nous avons donc focalisé notre étude sur les résultats issus de la HSS à partir des tumeurs. Des gènes surexprimés chez le porc pendant la phase de proliferation tumorale sont également retrouvés dans les études d’expression conduites dans le mélanome chez l’homme (TYR, MITF, MLANA, SDCBP, SILV, TYRP and ZFP106). Au tout début du phénomène de régression, on observe une surexpression des gènes impliqués dans les fonctions de la differentiation, le système immunitaire, l’arrêt du cycle cellulaire et la suppression des tumeurs. Deux gènes CD9 et RARRES1 montrent une très forte surexpression pendant la régression tant au niveau transcriptomique que protéique. Le CD9 est lié à la motilité cellulaire et RARRES1 est un gène potentiel suppresseur de tumeur. Dans un second temps, la dynamique du processus de régression a été étudiée grâce à la technologie des puces à ADN qui a permis d’établir un profil cinétique de l’expression génique. Ainsi, pour chacun des stades de la régression une corrélation entre la signature génique et son expression phénotypique a pu être établie. La signature de la régression comprend entre autres, des gènes impliqués dans la réponse immunitaire, le cycle cellulaire et la différentiation ainsi que la pigmentation des mélanocytes. Ce travail révèle que la régression des mélanomes semblerait être lié à une sous régulation précoce du cycle cellulaire. De plus le système immunitaire semble jouer un rôle majeur dans l’éradication des cellules tumorales. Par immunohistologie et cytometrie en flux, nous avons caracterisé le phenotype des cellules infiltrant les tumeurs : les cellules de l’immunité innée et acquise. On observe deux phases pendant le processus de la régression : une phase précoce consistant principalement à une infiltration par des cellules presentatrice d’antigènes (SWC3+) puis une phase d’infiltration tardive par des lymphocytes cytotoxiques (CD8+). La régression s’accompagne également de la présence de grosses cellules hyperpigmentées que nous avons commencés à caracteriser. Ce travail a permis de montrer qu’il existe
Recommended publications
  • Genomic Correlates of Relationship QTL Involved in Fore- Versus Hind Limb Divergence in Mice
    Loyola University Chicago Loyola eCommons Biology: Faculty Publications and Other Works Faculty Publications 2013 Genomic Correlates of Relationship QTL Involved in Fore- Versus Hind Limb Divergence in Mice Mihaela Palicev Gunter P. Wagner James P. Noonan Benedikt Hallgrimsson James M. Cheverud Loyola University Chicago, [email protected] Follow this and additional works at: https://ecommons.luc.edu/biology_facpubs Part of the Biology Commons Recommended Citation Palicev, M, GP Wagner, JP Noonan, B Hallgrimsson, and JM Cheverud. "Genomic Correlates of Relationship QTL Involved in Fore- Versus Hind Limb Divergence in Mice." Genome Biology and Evolution 5(10), 2013. This Article is brought to you for free and open access by the Faculty Publications at Loyola eCommons. It has been accepted for inclusion in Biology: Faculty Publications and Other Works by an authorized administrator of Loyola eCommons. For more information, please contact [email protected]. This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 3.0 License. © Palicev et al., 2013. GBE Genomic Correlates of Relationship QTL Involved in Fore- versus Hind Limb Divergence in Mice Mihaela Pavlicev1,2,*, Gu¨ nter P. Wagner3, James P. Noonan4, Benedikt Hallgrı´msson5,and James M. Cheverud6 1Konrad Lorenz Institute for Evolution and Cognition Research, Altenberg, Austria 2Department of Pediatrics, Cincinnati Children‘s Hospital Medical Center, Cincinnati, Ohio 3Yale Systems Biology Institute and Department of Ecology and Evolutionary Biology, Yale University 4Department of Genetics, Yale University School of Medicine 5Department of Cell Biology and Anatomy, The McCaig Institute for Bone and Joint Health and the Alberta Children’s Hospital Research Institute for Child and Maternal Health, University of Calgary, Calgary, Canada 6Department of Anatomy and Neurobiology, Washington University *Corresponding author: E-mail: [email protected].
    [Show full text]
  • Identification of Key Pathways and Genes in Endometrial Cancer Using Bioinformatics Analyses
    ONCOLOGY LETTERS 17: 897-906, 2019 Identification of key pathways and genes in endometrial cancer using bioinformatics analyses YAN LIU, TENG HUA, SHUQI CHI and HONGBO WANG Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China Received March 16, 2018; Accepted October 12, 2018 DOI: 10.3892/ol.2018.9667 Abstract. Endometrial cancer (EC) is one of the most Introduction common gynecological cancer types worldwide. However, to the best of our knowledge, its underlying mechanisms Endometrial carcinoma (EC) is one of the most common remain unknown. The current study downloaded three mRNA gynecological cancer types, with increasing global incidence and microRNA (miRNA) datasets of EC and normal tissue in recent years (1). A total of 60,050 cases of EC and 10,470 samples, GSE17025, GSE63678 and GSE35794, from the EC-associated cases of mortality were reported in the USA in Gene Expression Omnibus to identify differentially expressed 2016 (1), which was markedly higher than the 2012 statistics genes (DEGs) and miRNAs (DEMs) in EC tumor tissues. of 47,130 cases and 8,010 mortalities (2). Although numerous The DEGs and DEMs were then validated using data from studies have been conducted to investigate the mechanisms of The Cancer Genome Atlas and subjected to gene ontology endometrial tumorigenesis and development, to the best of our and Kyoto Encyclopedia of Genes and Genomes pathway knowledge, the exact etiology remains unknown. Understanding analysis. STRING and Cytoscape were used to construct a the potential molecular mechanisms underlying EC initiation protein-protein interaction network and the prognostic effects and progression is of great clinical significance.
    [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]
  • Open Full Page
    Research Article Distinct Genomic Profiles in Hereditary Breast Tumors Identified by Array-Based Comparative Genomic Hybridization Go¨ran Jo¨nsson,1 Tara L. Naylor,5 Johan Vallon-Christersson,1 Johan Staaf,1 Jia Huang,5 M. Renee Ward,5 Joel D. Greshock,5 Lena Luts,4 Ha˚kan Olsson,1 Nazneen Rahman,6 Michael Stratton,6 Markus Ringne´r,3 A˚ke Borg,1,2 and Barbara L. Weber5 1Department of Oncology, University Hospital; 2Lund Strategic Research Center for Stem Cell Biology and Cell Therapy and 3Department of Theoretical Physics, Lund University; and 4Department of Pathology, University Hospital, Lund, Sweden; 5Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania; and 6Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, United Kingdom Abstract of additional predisposing genes, although technical limitations Mutations in BRCA1 and BRCA2 account for a significant and the complexity of BRCA gene regulation and mutation proportion of hereditary breast cancers. Earlier studies have spectrum can probably explain why some disease-causing muta- shown that inherited and sporadic tumors progress along tions are missed (2). BRCA1 and BRCA2 function as classic tumor different somatic genetic pathways and that global gene suppressor genes with frequent loss of the wild-type allele in expression profiles distinguish between these groups. To tumors of mutation carriers. The BRCA1 protein has been determine whether genomic profiles similarly discriminate implicated in a broad range of cellular functions, including repair among BRCA1, BRCA2, and sporadic tumors, we established of double-strand breaks by homologous recombination, cell cycle DNA copy number profiles using comparative genomic checkpoint control, chromatin remodeling, and transcriptional hybridization to BAC-clone microarrays providing <1 Mb regulation.
    [Show full text]
  • Datasheet A11267-1 Anti-ZNF318 Antibody
    Product datasheet Anti-ZNF318 Antibody Catalog Number: A11267-1 BOSTER BIOLOGICAL TECHNOLOGY Special NO.1, International Enterprise Center, 2nd Guanshan Road, Wuhan, China Web: www.boster.com.cn Phone: +86 27 67845390 Fax: +86 27 67845390 Email: [email protected] Basic Information Product Name Anti-ZNF318 Antibody Gene Name ZNF318 Source Rabbit IgG Species Reactivity human, rat Tested Application WB, FCM, Direct ELISA Contents 500 ug/ml antibody with PBS ,0.02% NaN3 , 1 mg BSA and 50% glycerol. Immunogen E.coli-derived human ZNF318 recombinant protein (Position: E680-Q1197). Purification Immunogen affinity purified. Observed MW 290KD Dilution Ratios Western blot: 1:500-2000 Flow cytometry (FCM): 1-3μg/1x106 cells Direct ELISA: 1:100-1000 Storage 12 months from date of receipt,-20℃ as supplied.6 months 2 to 8℃ after reconstitution. Avoid repeated freezing and thawing Background Information Zinc finger protein 318 is a protein that in humans is encoded by the ZNF318 gene. ZNF318 encodes a nuclear protein with a zinc finger motif of the Cys2-His2 type that is a novel corepressor of androgen receptor (AR). Reference Anti-ZNF318 Antibody被引用在0文献中。 暂无引用 FOR RESEARCH USE ONLY. NOT FOR DIAGNOSTIC AND CLINICAL USE. 1 Product datasheet Anti-ZNF318 Antibody Catalog Number: A11267-1 BOSTER BIOLOGICAL TECHNOLOGY Special NO.1, International Enterprise Center, 2nd Guanshan Road, Wuhan, China Web: www.boster.com.cn Phone: +86 27 67845390 Fax: +86 27 67845390 Email: [email protected] Selected Validation Data Figure 1. Western blot Figure 2. Flow cytometry analysis of anti- ZNF318 analysis of A431 cell Antibody (A11267-1).
    [Show full text]
  • Enzyme DHRS7
    Toward the identification of a function of the “orphan” enzyme DHRS7 Inauguraldissertation zur Erlangung der Würde eines Doktors der Philosophie vorgelegt der Philosophisch-Naturwissenschaftlichen Fakultät der Universität Basel von Selene Araya, aus Lugano, Tessin Basel, 2018 Originaldokument gespeichert auf dem Dokumentenserver der Universität Basel edoc.unibas.ch Genehmigt von der Philosophisch-Naturwissenschaftlichen Fakultät auf Antrag von Prof. Dr. Alex Odermatt (Fakultätsverantwortlicher) und Prof. Dr. Michael Arand (Korreferent) Basel, den 26.6.2018 ________________________ Dekan Prof. Dr. Martin Spiess I. List of Abbreviations 3α/βAdiol 3α/β-Androstanediol (5α-Androstane-3α/β,17β-diol) 3α/βHSD 3α/β-hydroxysteroid dehydrogenase 17β-HSD 17β-Hydroxysteroid Dehydrogenase 17αOHProg 17α-Hydroxyprogesterone 20α/βOHProg 20α/β-Hydroxyprogesterone 17α,20α/βdiOHProg 20α/βdihydroxyprogesterone ADT Androgen deprivation therapy ANOVA Analysis of variance AR Androgen Receptor AKR Aldo-Keto Reductase ATCC American Type Culture Collection CAM Cell Adhesion Molecule CYP Cytochrome P450 CBR1 Carbonyl reductase 1 CRPC Castration resistant prostate cancer Ct-value Cycle threshold-value DHRS7 (B/C) Dehydrogenase/Reductase Short Chain Dehydrogenase Family Member 7 (B/C) DHEA Dehydroepiandrosterone DHP Dehydroprogesterone DHT 5α-Dihydrotestosterone DMEM Dulbecco's Modified Eagle's Medium DMSO Dimethyl Sulfoxide DTT Dithiothreitol E1 Estrone E2 Estradiol ECM Extracellular Membrane EDTA Ethylenediaminetetraacetic acid EMT Epithelial-mesenchymal transition ER Endoplasmic Reticulum ERα/β Estrogen Receptor α/β FBS Fetal Bovine Serum 3 FDR False discovery rate FGF Fibroblast growth factor HEPES 4-(2-Hydroxyethyl)-1-Piperazineethanesulfonic Acid HMDB Human Metabolome Database HPLC High Performance Liquid Chromatography HSD Hydroxysteroid Dehydrogenase IC50 Half-Maximal Inhibitory Concentration LNCaP Lymph node carcinoma of the prostate mRNA Messenger Ribonucleic Acid n.d.
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • S41467-020-18249-3.Pdf
    ARTICLE https://doi.org/10.1038/s41467-020-18249-3 OPEN Pharmacologically reversible zonation-dependent endothelial cell transcriptomic changes with neurodegenerative disease associations in the aged brain Lei Zhao1,2,17, Zhongqi Li 1,2,17, Joaquim S. L. Vong2,3,17, Xinyi Chen1,2, Hei-Ming Lai1,2,4,5,6, Leo Y. C. Yan1,2, Junzhe Huang1,2, Samuel K. H. Sy1,2,7, Xiaoyu Tian 8, Yu Huang 8, Ho Yin Edwin Chan5,9, Hon-Cheong So6,8, ✉ ✉ Wai-Lung Ng 10, Yamei Tang11, Wei-Jye Lin12,13, Vincent C. T. Mok1,5,6,14,15 &HoKo 1,2,4,5,6,8,14,16 1234567890():,; The molecular signatures of cells in the brain have been revealed in unprecedented detail, yet the ageing-associated genome-wide expression changes that may contribute to neurovas- cular dysfunction in neurodegenerative diseases remain elusive. Here, we report zonation- dependent transcriptomic changes in aged mouse brain endothelial cells (ECs), which pro- minently implicate altered immune/cytokine signaling in ECs of all vascular segments, and functional changes impacting the blood–brain barrier (BBB) and glucose/energy metabolism especially in capillary ECs (capECs). An overrepresentation of Alzheimer disease (AD) GWAS genes is evident among the human orthologs of the differentially expressed genes of aged capECs, while comparative analysis revealed a subset of concordantly downregulated, functionally important genes in human AD brains. Treatment with exenatide, a glucagon-like peptide-1 receptor agonist, strongly reverses aged mouse brain EC transcriptomic changes and BBB leakage, with associated attenuation of microglial priming. We thus revealed tran- scriptomic alterations underlying brain EC ageing that are complex yet pharmacologically reversible.
    [Show full text]
  • Supplementary Data
    SUPPLEMENTARY DATA A cyclin D1-dependent transcriptional program predicts clinical outcome in mantle cell lymphoma Santiago Demajo et al. 1 SUPPLEMENTARY DATA INDEX Supplementary Methods p. 3 Supplementary References p. 8 Supplementary Tables (S1 to S5) p. 9 Supplementary Figures (S1 to S15) p. 17 2 SUPPLEMENTARY METHODS Western blot, immunoprecipitation, and qRT-PCR Western blot (WB) analysis was performed as previously described (1), using cyclin D1 (Santa Cruz Biotechnology, sc-753, RRID:AB_2070433) and tubulin (Sigma-Aldrich, T5168, RRID:AB_477579) antibodies. Co-immunoprecipitation assays were performed as described before (2), using cyclin D1 antibody (Santa Cruz Biotechnology, sc-8396, RRID:AB_627344) or control IgG (Santa Cruz Biotechnology, sc-2025, RRID:AB_737182) followed by protein G- magnetic beads (Invitrogen) incubation and elution with Glycine 100mM pH=2.5. Co-IP experiments were performed within five weeks after cell thawing. Cyclin D1 (Santa Cruz Biotechnology, sc-753), E2F4 (Bethyl, A302-134A, RRID:AB_1720353), FOXM1 (Santa Cruz Biotechnology, sc-502, RRID:AB_631523), and CBP (Santa Cruz Biotechnology, sc-7300, RRID:AB_626817) antibodies were used for WB detection. In figure 1A and supplementary figure S2A, the same blot was probed with cyclin D1 and tubulin antibodies by cutting the membrane. In figure 2H, cyclin D1 and CBP blots correspond to the same membrane while E2F4 and FOXM1 blots correspond to an independent membrane. Image acquisition was performed with ImageQuant LAS 4000 mini (GE Healthcare). Image processing and quantification were performed with Multi Gauge software (Fujifilm). For qRT-PCR analysis, cDNA was generated from 1 µg RNA with qScript cDNA Synthesis kit (Quantabio). qRT–PCR reaction was performed using SYBR green (Roche).
    [Show full text]
  • Mir-17-92 Fine-Tunes MYC Expression and Function to Ensure
    ARTICLE Received 31 Mar 2015 | Accepted 22 Sep 2015 | Published 10 Nov 2015 DOI: 10.1038/ncomms9725 OPEN miR-17-92 fine-tunes MYC expression and function to ensure optimal B cell lymphoma growth Marija Mihailovich1, Michael Bremang1, Valeria Spadotto1, Daniele Musiani1, Elena Vitale1, Gabriele Varano2,w, Federico Zambelli3, Francesco M. Mancuso1,w, David A. Cairns1,w, Giulio Pavesi3, Stefano Casola2 & Tiziana Bonaldi1 The synergism between c-MYC and miR-17-19b, a truncated version of the miR-17-92 cluster, is well-documented during tumor initiation. However, little is known about miR-17-19b function in established cancers. Here we investigate the role of miR-17-19b in c-MYC-driven lymphomas by integrating SILAC-based quantitative proteomics, transcriptomics and 30 untranslated region (UTR) analysis upon miR-17-19b overexpression. We identify over one hundred miR-17-19b targets, of which 40% are co-regulated by c-MYC. Downregulation of a new miR-17/20 target, checkpoint kinase 2 (Chek2), increases the recruitment of HuR to c- MYC transcripts, resulting in the inhibition of c-MYC translation and thus interfering with in vivo tumor growth. Hence, in established lymphomas, miR-17-19b fine-tunes c-MYC activity through a tight control of its function and expression, ultimately ensuring cancer cell homeostasis. Our data highlight the plasticity of miRNA function, reflecting changes in the mRNA landscape and 30 UTR shortening at different stages of tumorigenesis. 1 Department of Experimental Oncology, European Institute of Oncology, Via Adamello 16, Milan 20139, Italy. 2 Units of Genetics of B cells and lymphomas, IFOM, FIRC Institute of Molecular Oncology Foundation, Milan 20139, Italy.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Monoclonal Antibodies
    NOTE Clinical Pathology Establishment of rat anti-canine DEP domain containing 1B (DEPDC1B) monoclonal antibodies Masaya IGASE1), Yuki MORINAGA1), Masahiro KATO2), Toshihiro TSUKUI2), Yusuke SAKAI3), Masaru OKUDA4) and Takuya MIZUNO1)* 1)Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi-shi, Yamaguchi 753-8511, Japan 2)Nippon Zenyaku Kogyo Co., Ltd., 1-1 Tairanoue, Sasagawa, Asaka-machi, Koriyama, Fukushima 963-0196, Japan 3)Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi-shi, Yamaguchi 753-8511, Japan 4)Laboratory of Veterinary Internal Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi-shi, Yamaguchi 753-8511, Japan ABSTRACT. DEP domain-containing 1B (DEPDC1B) is involved in the regulation of cell de- J. Vet. Med. Sci. adhesion and actin cytoskeleton activity during the G2/M transition of the cell cycle, and its 82(4): 483–487, 2020 overexpression has been proven to be associated with cancer progression in several human cancers. Canine DEPDC1B was identified as a gene that was overexpressed in canine lymphoma doi: 10.1292/jvms.19-0667 tissues in our previous study. However, in dogs, the protein expression of DEPDC1B remains to be determined due to the lack of a specific monoclonal antibody. Here, we developed rat monoclonal antibodies against canine DEPDC1B and characterized their applicability for immunodetection Received: 12 December 2019 assays. Our findings demonstrated that these antibodies are functional and can be important tools Accepted: 20 February 2020 to investigate the precise role of DEPDC1B in canine tumors. Advanced Epub: 6 March 2020 KEY WORDS: antibody, cancer, DEP domain-containing 1B, dog The DEP domain-containing 1B (DEPDC1B) gene was recently identified and localized at human chromosome 5 (5q12.1).
    [Show full text]