Supplementary Materials and Methods

Total Page:16

File Type:pdf, Size:1020Kb

Supplementary Materials and Methods Supplementary Materials and Methods Antibodies and reagents The goat polyclonal antibodies against TOX3, the rabbit polyclonal antibody against Ki-67, the mouse monoclonal antibody against BRCA1、DNMT1 and GAPDH, and the secondary goat anti-rabbit biotinylated (IgG) antibody, rabbit anti-goat HRP-conjugated antibody and goat anti-mouse HRP-conjugated antibody were purchased from Abcam (Cambridge, MA, USA). The rabbit polyclonal antibody against TOX3, the mouse monoclonal antibodies against HA and Myc epitopes were products of Sigma-Aldrich (St. Louis, MO, USA). MTT, DMSO, Albumin from bovine serum (BSA), Mayer’s hematoxylin, 3-3’-diaminobenzidine (DAB), Cisplatin (CDDP), 17-β estradiol (E2), trichostatin A (TSA), G418 and puromycin were also products of Sigma-Aldrich. cDNA, shRNA and reporter constructs The full length TNRC9 was PCR amplified using forward 5’- GGCCAATCCGGCCATGGATGTGAGGTTCTACC-3’ and reverse 5’- GGCCTCTAAGGCCTCAGAAAATACTGACCTGCGATAATACTTGAGT-3’ primers and subsequently subcloned into SfiI fragment of pCDEF3 vector (Invitrogen, Carlsbad, CA) with an N-terminal HA tag. The HA-TNRC9mut is a TNRC9 DNA binding domain- deleted truncation of HA-TNRC9 with a deletion from 922bp to 1149bp of NM_001080430. The full length CREB was PCR amplified using forward 5’- GGCCAATCCGGCCATGACCATGGAATCTGGAG-3’ and reverse 5’- GGCCTCTAAGGCCTTAATCTGATTTGTGGCAGT-3’ primers and subcloned into SfiI fragment of pCDEF3 vector with an N-terminal Myc tag. The shRNA construct targeting TNRC9 were established using pSuper RNAi system (Oligoengine, Seattle, WA). The targeting regions were 1931bp-1949bp (sh1) and 974bp-992bp (sh2) of NM_001080430. The non-specific control sequence was 5’-GACGTAAACGGCCACAA GT-3’. A 602 subregion of BRCA1 promoter (-458 to +145) was PCR amplified from human genomic DNA using forward 5’-CTCGAGACCTTTCATTCCGCAACGCATG- 3’ and reverse 5’- AAGCTTTCCCGGGACTCTACTACCTTTAC-3’ primers (1) and subsequently subcloned into XhoI-BglII fragment of pGL4 basic vector (Promega, 1 Madison, WI). It was named as pGL4-promBRCA1. Quantitative real-time PCR All the quantitative real-time PCR were performed on 7500 Real-Time PCR system (Applied Biosystems, Carlsbad, CA) using GoTaq® qPCR Master Mix (Promega, Madison, WI). The genomics region of FUT2 and PTPN4 were used as references to assess the copy number variation of TNRC9 genomic region. Primer sequences were as follows: TNRC9 exon3 forward 5’-CTTGGGGACGAGGAATTCGA-3’, reverse 5’- GTAATGGAAGGGAGGTCCA-3’; TNRC9 exon4 forward 5’-ATCGTCCACATGACC GATG-3’, reverse 5’-CTCTTCATTGATGGAGCTG-3’; FUT2 forward 5’- ATCTTCGTGGTCACCAGTAATGG-3’, reverse 5’-GGTTACACTGTGTGAGTAGAG C-3’; PTPN4 forward 5’-ACGCACGGTCCCATAGTATC-3’, reverse 5’- AAATAGTG GTGCCTCCTTGG-3’. When min (CtFUT2, CtPTPN4) - max (CtTNRC9exon3, CtTNRC9exon4) >1, the sample was considered to have a copy number amplification of TNRC9 genomic DNA. HPRT1 was used as the internal reference to assess the mRNA expression level of TNRC9. The reverse transcription of total RNA was performed by GoTaq® 2-Step RT- qPCR System (Promega, Madison, WI). The primer sequences used to amplify TNRC9 cDNA were 5’-TGCCTCACACATCTCCTTCAC-3’ and 5’- TTGGAGTCTTGGGCTTCTTGC-3’, and HPRT1 cDNA were 5’- TGACACTGGCAAAACAATGCA-3’ and 5’-GGTCCTTTTCACCAGCAAGCT-3’. The bisulfite-converted DNA was analyzed by quantitative PCR using primers for methylated BRCA1 promoter region: forward 5’-TCGTGGTAACGGAAAAGCGC-3’ (-38 to -19) and reverse 5’-AACGAACTCACGCCGCGCAA-3’ (+11 to +30) and unmethylated BRCA1 promoter region: forward 5’-TTGAGAGGTTGTTGTTTAGTGG- 3’ (-74 to -43) and reverse 5’-AACAAACTCACACCACACAA-3’ (+11 to +30) (2). The 3’ end of DHFR was used as a reference: forward 5’- TTGATGTTTAGGAGGAGAAAGG-3’ and reverse 5’- AACCCAACAATATCAAAAACTA-3’. The copies of methylated or unmethylated BRCA1 promoter were calculated using 2-(CtBRCA1-CtDHFR). The combination of methylated BRCA1 promoter and unmethylated BRCA1 promoter was used as the total copies of BRCA1 promoter. The percentage of methylated BRCA1 promoter was 2 calculated by dividing the total copies of BRCA1 promoter. Bisulfite sequencing of BRCA1 promoter To amplify a region of BRCA1 promoter, the bisulfite-converted DNA from TNRC9 abrogated ZR-75-1 cells and from control cells were amplified using a nested PCR (3). The first run was accomplished by the forward primer 5’- GGGGTTGGATGGGAATTGTAG-3’(-686 to -666 of BRCA1 gene) and reverse primer 5’-CTCTACTACCTTTACCCAAAACA-3’ (+130 to +108). One tenth of the first run amplification was re-amplified with forward primer 5’- GTTTATAATTGTTGATAAGTATAAG-3’(-592 to -568) and reverse primer 5’- AAAACCCCACAACCTATCCC-3’(+66 to +47). The second run PCR product was purified and cloned into a T vector-pGEMT (Promega). Twenty clones of each sample were randomly selected and sequenced. Immunohistochemistry Formalin fixed and paraffin embedded sections were cut at 5 micron thick and were deparaffinized and rehydrated through 3 concentrations of alcohol. Endogenous peroxidase activity was blocked with 3% H2O2 for 15 minutes. Antigen retrieval was carried out in 0.01 M citrate buffer, pH 6.0, at 95°C for 20 minutes, followed by 20 minutes cooling at room temperature. Nonspecific binding was blocked using PBST containing 5% BSA for 1 hr. Sections were incubated overnight with goat anti-TNRC9 (1:50) and mouse anti-BRCA1 antibodies (1:50) at 4 °C, followed by HRP-conjugated secondary antibodies (1:500) for 1 hour at room temperature. Antibody binding was revealed with DAB (DAKO, Denmark) as chromogen. In between steps sections were washed with PBST for 3 times, each for 7 min. Sections were counterstained with Mayer’s hematoxylin, dehydrated and mounted. Images were obtained using a bright field microscope (AxioVision software). High-grade and low-grade breast cancer samples were included in this study. Sections incubated with non-immune control sera acted as negative control. Immunoprecipitation and immunoblotting Cells were lysed with RIPA buffer (50mM Tris pH7.4, 100mM Nacl, 0.25% Na- deoxycholate, 1% NP40, 0.1% SDS) containing 1X protease inhibitors cocktail (Sigma- Aldrich, St. Louis, MO). Equal amounts of cell lysates were separated on SDS-PAGE 3 gel, transferred to PVDF membrane and probed with primary antibodies. Bands were visualized using Geliance 600 Imaging System (PerkinElmer, Waltham, MA), in conjugation with HRP-conjugated secondary antibodies and ImmobilonTM western chemiluminescent HRP substrate (Millipore, Billerica, MA). For the detection of interaction between TNRC9 and CREB, 107 transfected HEK293 cells were lysed in RIPA buffer. Lysates were precleared with Protein A/G Plus Agarose beads (Santa Cruz Biotechnology, Santa Cruz, CA) and 2 mg IgG for 2 h at 4 °C. The precleared lysates were immunoprecipitated with anti-TNRC9 or anti-HA for over 4 h at 4 °C. The immunoprecipitated samples were washed three times with lysis buffer, resolved by SDS–PAGE and then subjected to immunoblotting analysis with anti-Myc. Mouse model Cells for injection are agitated slowly on a Vortex mixer in a 50 ml Falcon tube to ensure even suspension. Enough cells are then withdrawn to inoculate 5 mice; that is, ~0.5 ml of a 3x107 cell/mL suspension. The syringe is then gently inverted (containing the glass bead) to ensure even suspension. A 27 Gauge, ½ inch Precision Glide needle (BD, Franklin Lakes, NJ) is then inserted onto the syringe and primed. NU/NU nude mice (Charles River Laboratories, Wilmington, MA) are anesthetized by placing them in a 5% Isoflurane chamber. Once the mice are deeply under anesthesia, mice are removed individually, placed into the biological-safety cabinet and prepared for injection (positioned prone and upper right flank wiped with a 70% ethanol pad). The syringe is again gently inverted 3 times to ensure even cell suspension. Without tenting the skin, 0.1 ml of the cell suspension (containing approximately 3 million cells) is injected into the upper right flank of the mouse, subcutaneously. Each mouse is then placed into a separate chamber until conscious before being returned to its original cage. Tumors were monitored at 2-3 day intervals for a total of 28 days. Tumor size (width and length) was recorded, the skin overlaying the tumor was inspected and the mice weighed. After 28 days, tumors were surgically excised, measured with digital calipers and placed into 10% neutral buffered formalin. Tumors were then embedded in paraffin, sectioned and stained with hematoxylin and eosin (H&E) or immunostained for Ki-67 (proliferative cells) a proliferative cell marker, using a polyclonal IgG primary antibody (rabbit; abcam) and 4 anti-rabbit biotinylated (IgG) secondary antibody (goat; Abcam) with Vectastain Elite ABC Kit (Vector Laboratories, Burlingame, CA) and methyl green counterstain (Vector Laboratories, Burlingame, CA). All mouse experiments were reviewed and approved by the Institutional Animal Care and Use Committee at Brown University. In silico assays for the expression of TNRC9 and BRCA1 In silico assays were performed on 266 early breast cancer patients. Transcriptional data were retrieved from NCBI GEO database (GSE21653) and further analyzed using BRBArrayTools developed by the Biometric Research Branch, National Cancer Institute (http://linus.nci.nih.gov/BRB-ArrayTools.html), Partek Genomics Suite (PARTEK, St Louis, MO), Stanford Cluster Program and TreeView software (4). Genes were projected using log2 intensity and gene ratios were average corrected across experimental samples and displayed according to uncentered correlation algorithm. Similar to breast
Recommended publications
  • Screening and Identification of Key Biomarkers in Clear Cell Renal Cell Carcinoma Based on Bioinformatics Analysis
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Screening and identification of key biomarkers in clear cell renal cell carcinoma based on bioinformatics analysis Basavaraj Vastrad1, Chanabasayya Vastrad*2 , Iranna Kotturshetti 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. 3. Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka 562209, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract Clear cell renal cell carcinoma (ccRCC) is one of the most common types of malignancy of the urinary system. The pathogenesis and effective diagnosis of ccRCC have become popular topics for research in the previous decade. In the current study, an integrated bioinformatics analysis was performed to identify core genes associated in ccRCC. An expression dataset (GSE105261) was downloaded from the Gene Expression Omnibus database, and included 26 ccRCC and 9 normal kideny samples. Assessment of the microarray dataset led to the recognition of differentially expressed genes (DEGs), which was subsequently used for pathway and gene ontology (GO) enrichment analysis.
    [Show full text]
  • TOX2 Is Differentially Expressed in Non-Small Cell Lung Cancer-PDF 092220
    1 Tox2 is differentially expressed in non-small cell lung cancer and associates with patient 2 survival. 3 Shahan Mamoor1 4 [email protected] East Islip, NY USA 5 6 Non-small cell lung cancer (NSCLC) is the leading cause of cancer death in the United States1. 7 We mined published microarray data2,3,4 to identify differentially expressed genes in NSCLC. 8 We found that the gene encoding the thymyocyte selection-associated high mobility box protein 9 family member 2 transcription factor Tox2 was among the genes whose expression was most quantitatively different in tumors from patients with NSCLC as compared to the lung. Tox2 10 expression was significantly decreased in NSCLC tumors as compared to the lung, and lower 11 expression of Tox2 in patient tumors was significantly associated with worse overall survival. Tox2 may be important for initiation or progression of non-small cell lung cancer in humans. 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 Keywords: Tox2, NSCLC, non-small cell lung cancer, systems biology of NSCLC, targeted 27 therapeutics in NSCLC. 28 1 OF 16 1 In 2016, lung cancer resulted in the death of 158,000 Americans; 81% of all patients 2 diagnosed with lung cancer will expire within 5 years5. Non-small cell lung cancer (NSCLC) is 3 4 the most common type of lung cancer, diagnosed in 84% of patients with lung cancer, and 76% 5 of all patients with NSCLC will expire within 5 years5. The rational development of targeted 6 therapeutics to treat patients with NSCLC can be supported by an enhanced understanding of 7 8 fundamental transcriptional features of NSCLC tumors.
    [Show full text]
  • TOX As a Potential Target for Immunotherapy in Lymphocytic Malignancies Chaofeng Liang1,2, Shuxin Huang1, Yujie Zhao1, Shaohua Chen1* and Yangqiu Li1*
    Liang et al. Biomarker Research (2021) 9:20 https://doi.org/10.1186/s40364-021-00275-y REVIEW Open Access TOX as a potential target for immunotherapy in lymphocytic malignancies Chaofeng Liang1,2, Shuxin Huang1, Yujie Zhao1, Shaohua Chen1* and Yangqiu Li1* Abstract TOX (thymocyte selection-associated HMG BOX) is a member of a family of transcriptional factors that contain the highly conserved high mobility group box (HMG-box) region. Increasing studies have shown that TOX is involved in maintaining tumors and promoting T cell exhaustion. In this review, we summarized the biological functions of TOX and its contribution as related to lymphocytic malignancies. We also discussed the potential role of TOX as an immune biomarker and target in immunotherapy for hematological malignancies. Keywords: Thymocyte selection-associated HMG BOX, Biological function, T cell exhaustion, Lymphocytic malignancies, Immune biomarker, Immunotherapy Background located on different chromosomes and have different TOX (thymocyte selection-associated HMG BOX), a functions (Fig. 1, Table 1)[16]. transcription factor, belongs to the high mobility group box (HMG-box) superfamily. In 2002, Wilkinson et al. TOX demonstrated for the first time that TOX plays an im- TOX, also known as KIAA0808, is located on q12.1 of portant role in the differentiation of CD4 + CD8+ human chromosome 8 and has one transcript. The double-positive thymocytes by gene microarray technol- length of the TOX mRNA is 4076 bp, and it contains ogy. Numerous studies have shown that TOX plays a nine exons that translate into a protein consisting of 526 vital role in the development and formation of various amino acids.
    [Show full text]
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [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]
  • Host-Parasite Interaction of Atlantic Salmon (Salmo Salar) and the Ectoparasite Neoparamoeba Perurans in Amoebic Gill Disease
    ORIGINAL RESEARCH published: 31 May 2021 doi: 10.3389/fimmu.2021.672700 Host-Parasite Interaction of Atlantic salmon (Salmo salar) and the Ectoparasite Neoparamoeba perurans in Amoebic Gill Disease † Natasha A. Botwright 1*, Amin R. Mohamed 1 , Joel Slinger 2, Paula C. Lima 1 and James W. Wynne 3 1 Livestock and Aquaculture, CSIRO Agriculture and Food, St Lucia, QLD, Australia, 2 Livestock and Aquaculture, CSIRO Agriculture and Food, Woorim, QLD, Australia, 3 Livestock and Aquaculture, CSIRO Agriculture and Food, Hobart, TAS, Australia Marine farmed Atlantic salmon (Salmo salar) are susceptible to recurrent amoebic gill disease Edited by: (AGD) caused by the ectoparasite Neoparamoeba perurans over the growout production Samuel A. M. Martin, University of Aberdeen, cycle. The parasite elicits a highly localized response within the gill epithelium resulting in United Kingdom multifocal mucoid patches at the site of parasite attachment. This host-parasite response Reviewed by: drives a complex immune reaction, which remains poorly understood. To generate a model Diego Robledo, for host-parasite interaction during pathogenesis of AGD in Atlantic salmon the local (gill) and University of Edinburgh, United Kingdom systemic transcriptomic response in the host, and the parasite during AGD pathogenesis was Maria K. Dahle, explored. A dual RNA-seq approach together with differential gene expression and system- Norwegian Veterinary Institute (NVI), Norway wide statistical analyses of gene and transcription factor networks was employed. A multi- *Correspondence: tissue transcriptomic data set was generated from the gill (including both lesioned and non- Natasha A. Botwright lesioned tissue), head kidney and spleen tissues naïve and AGD-affected Atlantic salmon [email protected] sourced from an in vivo AGD challenge trial.
    [Show full text]
  • Large Meta-Analysis of Genome-Wide Association Studies
    medRxiv preprint doi: https://doi.org/10.1101/2020.10.01.20200659; this version posted October 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. It is made available under a CC-BY-NC-ND 4.0 International license . Large meta-analysis of genome-wide association studies expands knowledge of the genetic etiology of Alzheimer’s disease and highlights potential translational opportunities Céline Bellenguez1,*,#, Fahri Küçükali2,3,4*, Iris Jansen5,6*, Victor Andrade7,8*, Sonia Morenau- Grau9,10,*, Najaf Amin11,12, Benjamin Grenier-Boley1, Anne Boland13, Luca Kleineidam7,8, Peter Holmans14, Pablo Garcia9,10, Rafael Campos Martin7, Adam Naj15,16, Yang Qiong17, Joshua C. Bis18, Vincent Damotte1, Sven Van der Lee5,6,19, Marcos Costa1, Julien Chapuis1, Vilmentas Giedraitis20, María Jesús Bullido10,21, Adolfo López de Munáin10,22, Jordi Pérez- Tur10,23, Pascual Sánchez-Juan10,24, Raquel Sánchez-Valle25, Victoria Álvarez26, Pau Pastor27, Miguel Medina10,28, Jasper Van Dongen2,3,4, Christine Van Broeckhoven2,3,4, Rik Vandenberghe29,30, Sebastiaan Engelborghs31,32, Gael Nicolas33, Florence Pasquier34, Olivier Hanon35, Carole Dufouil36, Claudine Berr37, Stéphanie Debette36, Jean-François Dartigues36, Gianfranco Spalletta38, Benedetta Nacmias39,40, Vincenzo Solfrezzi41, Barbara Borroni42, Lucio Tremolizzo43, Davide Seripa44, Paolo Caffarra45, Antonio Daniele46,47, Daniela Galimberti48,49, Innocenzo Rainero50, Luisa Benussi51, Alesio Squassina52, Patrizia Mecoci53, Lucilla Parnetti54, Carlo Masullo55, Beatrice Arosio56, John Hardy57, Simon Mead58, Kevin Morgan59, Clive Holmes60, Patrick Kehoe61, Bob Woods62, EADB, Charge, ADGC, Jin Sha15,16, Yi Zhao15,63, Chien-Yueh Lee15,63, Pavel P.
    [Show full text]
  • TOX3 Is Expressed in Mammary ER+ Epithelial Cells and Regulates ER
    Seksenyan et al. BMC Cancer (2015) 15:22 DOI 10.1186/s12885-015-1018-2 RESEARCH ARTICLE Open Access TOX3 is expressed in mammary ER+ epithelial cells and regulates ER target genes in luminal breast cancer Akop Seksenyan1, Asha Kadavallore1, Ann E Walts2, Brian de la Torre1, Dror Berel3,4, Samuel P Strom5,6, Parinaz Aliahmad1, Vincent A Funari5 and Jonathan Kaye1,3,7* Abstract Background: A breast cancer susceptibility locus has been mapped to the gene encoding TOX3. Little is known regarding the expression pattern or biological role of TOX3 in breast cancer or in the mammary gland. Here we analyzed TOX3 expression in murine and human mammary glands and in molecular subtypes of breast cancer, and assessed its ability to alter the biology of breast cancer cells. Methods: We used a cell sorting strategy, followed by quantitative real-time PCR, to study TOX3 gene expression in the mouse mammary gland. To study the expression of this nuclear protein in human mammary glands and breast tumors, we generated a rabbit monoclonal antibody specific for human TOX3. In vitro studies were performed on MCF7, BT474 and MDA-MB-231 cell lines to study the effects of TOX3 modulation on gene expression in the context of breast cancer cells. Results: We found TOX3 expression in estrogen receptor-positive mammary epithelial cells, including progenitor cells. A subset of breast tumors also highly expresses TOX3, with poor outcome associated with high expression of TOX3 in luminal B breast cancers. We also demonstrate the ability of TOX3 to alter gene expression in MCF7 luminal breast cancer cells, including cancer relevant genes TFF1 and CXCR4.
    [Show full text]
  • NATURAL KILLER CELLS, HYPOXIA, and EPIGENETIC REGULATION of HEMOCHORIAL PLACENTATION by Damayanti Chakraborty Submitted to the G
    NATURAL KILLER CELLS, HYPOXIA, AND EPIGENETIC REGULATION OF HEMOCHORIAL PLACENTATION BY Damayanti Chakraborty Submitted to the graduate degree program in Pathology and Laboratory Medicine and the Graduate Faculty of the University of Kansas in partial fulfillment ofthe requirements for the degree of Doctor of Philosophy. ________________________________ Chair: Michael J. Soares, Ph.D. ________________________________ Jay Vivian, Ph.D. ________________________________ Patrick Fields, Ph.D. ________________________________ Soumen Paul, Ph.D. ________________________________ Michael Wolfe, Ph.D. ________________________________ Adam J. Krieg, Ph.D. Date Defended: 04/01/2013 The Dissertation Committee for Damayanti Chakraborty certifies that this is the approved version of the following dissertation: NATURAL KILLER CELLS, HYPOXIA, AND EPIGENETIC REGULATION OF HEMOCHORIAL PLACENTATION ________________________________ Chair: Michael J. Soares, Ph.D. Date approved: 04/01/2013 ii ABSTRACT During the establishment of pregnancy, uterine stromal cells differentiate into decidual cells and recruit natural killer (NK) cells. These NK cells are characterized by low cytotoxicity and distinct cytokine production. In rodent as well as in human pregnancy, the uterine NK cells peak in number around mid-gestation after which they decline. NK cells associate with uterine spiral arteries and are implicated in pregnancy associated vascular remodeling processes and potentially in modulating trophoblast invasion. Failure of trophoblast invasion and vascular remodeling has been shown to be associated with pathological conditions like preeclampsia syndrome, hypertension in mother and/or fetal growth restriction. We hypothesize that NK cells fundamentally contribute to the organization of the placentation site. In order to study the in vivo role of NK cells during pregnancy, gestation stage- specific NK cell depletion was performed in rats using anti asialo GM1 antibodies.
    [Show full text]
  • Associated 16P11.2 Deletion in Drosophila Melanogaster
    ARTICLE DOI: 10.1038/s41467-018-04882-6 OPEN Pervasive genetic interactions modulate neurodevelopmental defects of the autism- associated 16p11.2 deletion in Drosophila melanogaster Janani Iyer1, Mayanglambam Dhruba Singh1, Matthew Jensen1,2, Payal Patel 1, Lucilla Pizzo1, Emily Huber1, Haley Koerselman3, Alexis T. Weiner 1, Paola Lepanto4, Komal Vadodaria1, Alexis Kubina1, Qingyu Wang 1,2, Abigail Talbert1, Sneha Yennawar1, Jose Badano 4, J. Robert Manak3,5, Melissa M. Rolls1, Arjun Krishnan6,7 & 1234567890():,; Santhosh Girirajan 1,2,8 As opposed to syndromic CNVs caused by single genes, extensive phenotypic heterogeneity in variably-expressive CNVs complicates disease gene discovery and functional evaluation. Here, we propose a complex interaction model for pathogenicity of the autism-associated 16p11.2 deletion, where CNV genes interact with each other in conserved pathways to modulate expression of the phenotype. Using multiple quantitative methods in Drosophila RNAi lines, we identify a range of neurodevelopmental phenotypes for knockdown of indi- vidual 16p11.2 homologs in different tissues. We test 565 pairwise knockdowns in the developing eye, and identify 24 interactions between pairs of 16p11.2 homologs and 46 interactions between 16p11.2 homologs and neurodevelopmental genes that suppress or enhance cell proliferation phenotypes compared to one-hit knockdowns. These interac- tions within cell proliferation pathways are also enriched in a human brain-specific network, providing translational relevance in humans. Our study indicates a role for pervasive genetic interactions within CNVs towards cellular and developmental phenotypes. 1 Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA. 2 Bioinformatics and Genomics Program, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA.
    [Show full text]
  • TOX3 Is a Favorable Prognostic Indicator and Potential Immunomodulatory Factor in Lung Adenocarcinoma
    4144 ONCOLOGY LETTERS 18: 4144-4152, 2019 TOX3 is a favorable prognostic indicator and potential immunomodulatory factor in lung adenocarcinoma DE ZENG1, HAOYU LIN2, JIANXIONG CUI2 and WEIQUAN LIANG2 1Department of Medical Oncology, Cancer Hospital of Shantou University Medical College; 2Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China Received February 13, 2018; Accepted January 14, 2019 DOI: 10.3892/ol.2019.10748 Abstract. Thymocyte selection-associated high mobility and cytotoxic T lymphocyte-associated protein 4 (CTLA4), as group box (TOX) genes represent a novel family of genes. well as the development of novel immunotherapy agents, lung Deregulated expression of TOXs has been reported in a cancer treatments have improved notably in terms of disease variety of cancer types, including lung cancer. It has also control and survival outcomes (3). However, the success rate been reported that TOXs are crucial regulators of the immune of immunotherapy remains low, and a large number of cancer system. The present study systematically evaluated the prog- types, including lung cancer, remain largely insensitive or nostic values of TOX family members using a set of publicly refractory to PD-1, PD-L1 and CTLA4 blockade (2,4-6). accessible databases, including Oncomine, Kaplan-Meier Therefore, research has focused on identifying novel immu- plotter and cBioPortal. It was revealed that TOX expression notherapy targets or immunomodulatory factors that can profiles differed between lung cancer and normal tissues, be exploited to enhance the response to current immuno- and high expression of TOX mRNAs generally predicted therapeutic agents and address immunotherapy resistance in improved survival outcomes.
    [Show full text]
  • A Dissertation Entitled the Androgen Receptor
    A Dissertation entitled The Androgen Receptor as a Transcriptional Co-activator: Implications in the Growth and Progression of Prostate Cancer By Mesfin Gonit Submitted to the Graduate Faculty as partial fulfillment of the requirements for the PhD Degree in Biomedical science Dr. Manohar Ratnam, Committee Chair Dr. Lirim Shemshedini, Committee Member Dr. Robert Trumbly, Committee Member Dr. Edwin Sanchez, Committee Member Dr. Beata Lecka -Czernik, Committee Member Dr. Patricia R. Komuniecki, Dean College of Graduate Studies The University of Toledo August 2011 Copyright 2011, Mesfin Gonit This document is copyrighted material. Under copyright law, no parts of this document may be reproduced without the expressed permission of the author. An Abstract of The Androgen Receptor as a Transcriptional Co-activator: Implications in the Growth and Progression of Prostate Cancer By Mesfin Gonit As partial fulfillment of the requirements for the PhD Degree in Biomedical science The University of Toledo August 2011 Prostate cancer depends on the androgen receptor (AR) for growth and survival even in the absence of androgen. In the classical models of gene activation by AR, ligand activated AR signals through binding to the androgen response elements (AREs) in the target gene promoter/enhancer. In the present study the role of AREs in the androgen- independent transcriptional signaling was investigated using LP50 cells, derived from parental LNCaP cells through extended passage in vitro. LP50 cells reflected the signature gene overexpression profile of advanced clinical prostate tumors. The growth of LP50 cells was profoundly dependent on nuclear localized AR but was independent of androgen. Nevertheless, in these cells AR was unable to bind to AREs in the absence of androgen.
    [Show full text]