Suppression of Carcinogenesis and Tumor Progression by an Energy Restriction-Mimetic Agent in Murine Models of Prostate Cancer

Total Page:16

File Type:pdf, Size:1020Kb

Suppression of Carcinogenesis and Tumor Progression by an Energy Restriction-Mimetic Agent in Murine Models of Prostate Cancer SUPPRESSION OF CARCINOGENESIS AND TUMOR PROGRESSION BY AN ENERGY RESTRICTION-MIMETIC AGENT IN MURINE MODELS OF PROSTATE CANCER DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Lisa Danielle Berman-Booty, V.M.D Graduate Program in Veterinary Biosciences The Ohio State University 2013 Dissertation Committee: Professor Ching-Shih Chen, Advisor Professor Robert Brueggemeier Professor Steven Clinton Professor Thomas Rosol Copyrighted by Lisa Danielle Berman-Booty 2013 ABSTRACT Cancer cells preferentially utilize glycolysis to generate energy even in the presence of sufficient oxygen for oxidative phosphorylation. This shift in energy metabolism, termed the Warburg effect, is responsible for cancer cells’ high metabolic rate. Therapies that inhibit cancer cell energy metabolism have proved effective in vitro, and dietary caloric restriction is a valuable experimental chemotherapeutic strategy. While animal studies utilizing caloric restriction typically restrict the experimental group’s caloric intake by 20-40%, this degree of caloric restriction is not realistic for human cancer patients. Therefore, agents that can induce a response similar to that of glucose restriction in vitro and caloric restriction in vivo are needed. These agents are termed energy-restriction mimetic agents (ERMAs) and include 2-deoxyglucose, resveratrol, and the thiazolidinedione derivatives OSU-CG12 and OSU-CG5. Here, we characterized OSU- CG5’s mechanism of action in vitro and its chemotherapeutic and chemopreventive activities in vivo. Specifically, we evaluated OSU-CG5’s activity in three different human prostate cancer cell lines that range from androgen-dependent (LNCaP) to castration- resistant (LNCaP-abl and PC3). OSU-CG5 was cytotoxic and induced a response similar to that of glucose deprivation in these cell lines. Additionally, treatment with OSU-CG5 resulted in decreased expression of genes that promote the Warburg effect, namely, those ii for glucose transporter 1 (GLUT1) and a number of metabolic enzymes. Regarding its in vivo chemotherapeutic activity, 100 mg/kg/day of OSU-CG5 administered via daily oral gavage suppressed the growth of LNCaP-abl xenograft tumors by 81%. Tumor growth suppression was associated with decreased tumor cell proliferation, as determined by proliferating cell nuclear antigen (PCNA) levels, and modulation of intratumoral biomarkers associated with cell survival, growth, and metabolism, including insulin like growth factor 1 (IGF-1) and its receptor IGF-1R, Myc, the androgen receptor (AR), and cyclins D1 and E. OSU-CG5’s activity was also investigated using transgenic adenocarcinoma of the mouse prostate (TRAMP) mice. Treatment of 6-week-old TRAMP mice with 100 mg/kg/day of OSU-CG5 via oral gavage for 4 weeks significantly reduced absolute and relative urogenital tract weights, as well as the weights of the individual lobes of the prostate. Reductions in lobe weight were associated with decreased cell proliferation within prostatic intraepithelial neoplasia (PIN) lesions, as determined by immunostaining for Ki67 and western blotting for PCNA. OSU-CG5 also decreased the levels of the AR, phosphorylated Akt (p-Akt), and IGF-1R within the prostates of these mice. When 6-week-old, intact, male TRAMP mice were fed an AIN- 76A diet containing 1286 ppm of OSU-CG5 for approximately 18 weeks, prostate tumor development was not suppressed or significantly delayed. However, the tumors that developed in OSU-CG5-treated mice were 54.6% smaller by volume and 54.1% smaller by mass than the control mouse tumors. Decreased tumor size was associated with decreased tumor cell proliferation, as determined by Ki67 immunostaining, and reductions in the AR and p-Akt. Collectively, these studies demonstrate OSU-CG5’s iii efficacy as a chemotherapeutic agent. OSU-CG5 mediated this effect by reducing tumor cell proliferation and modulating pro-growth and pro-survival biomarkers. iv DEDICATION This document is dedicated to my husband Jordan and parents Marvin and Ronna v ACKNOWLEDGMENTS I thank my advisor Dr. Ching-Shih Chen for his advice, mentorship and unwavering support. I thank the members of my advisory committee for their guidance. I thank Dr. Samuel Kulp for his advice and help with experimental design, animal studies, and manuscript preparation. I appreciate all the help and encouragement provided by the other members of the Chen lab. In particular, I acknowledge Dr. Po-Chen Chu for his technical expertise and assistance and Dr. Dasheng Wang for synthesizing OSU-CG5. I thank Dr. Jennifer Thomas-Ahner of the Clinton lab for her assistance with mouse necropsies and urogenital tract microdissection. I thank Drs. Brad Bolon and Krista La Perle of the Comparative Pathology and Mouse Phenotyping Shared Resource (CPMPSR) for their guidance and mentorship in mouse and toxicologic pathology. I appreciate Alan Fletchner, Anne Saulsbery, Bill Kimble, and Florinda Jaynes of the CPMPSR for their help with tissue and slide processing and immunohistochemistry. I thank Jody Sneddon of the CPMPSR for performing serum chemistries and complete blood counts. I acknowledge the National Institute of Health for my research support and funding. From June 2009 through April 2012, my research was supported by the T-32 Institutional National Research Service Award (NRSA) in Mouse Models of Human vi Disease (Ruth L. Kirschstein award). From April 2012 through the present time, my research has been supported by a K01 SERCA. Lastly, I thank my husband and parents for their love and support throughout this entire process. vii VITA March 7, 1982 ................................................Born—Brookline, MA, USA 2004 ...............................................................Bachelors of Arts, Biology School of Arts and Sciences University of Pennsylvania Philadelphia, PA 2008 ...............................................................Veterinariae Medicinae Doctoris School of Veterinary Medicine University of Pennsylvania Philadelphia, PA 2008 to present ..............................................Residency in Veterinary Anatomic Pathology Department of Veterinary Biosciences The Ohio State University Columbus, OH 2008 to present ..............................................Postdoctoral Fellow Department of Veterinary Biosciences The Ohio State University Columbus, OH 2012................................................................Diplomate, American College of Veterinary Pathologists (anatomic pathology) viii PUBLICATIONS 1. Berman-Booty LD, Chu PC, Thomas-Ahner JM, Bolon B, Wang D, Yang T, Clinton SK, Kulp SK, Chen CS. Suppression of prostate epithelial proliferation and pro-growth signaling in transgenic mice by a new energy-restriction mimetic agent. Cancer Prev Res in press (PMID: 23275006) 2.Omar HA, Berman-Booty L, Weng JR. Energy restriction: stepping stones towards cancer therapy. Future Oncol 2012; 8 (12): 1503-6 3.Berman-Booty LD, Garzel LM, Bergdall V, La Perle KM. A prostate fibromyxoid sarcoma with smooth muscle differentiation in a F344xBNF1 rat. Vet Pathol 2012; 49(4): 642-647 4.Berman-Booty LD, Sargeant AM, Rosol TJ, Rengel RC, Clinton SK, Chen CS, Kulp SK. A review of the existing grading schemes and a proposal for a modified grading scheme for prostatic lesions in TRAMP mice. Toxicol Pathol 2012; 40(1):5-17 5.Omar HA, Chou CC, Berman-Booty LD, Ma Y, Hung JH, Wang D, Kogure T, Patel T, Terracciano L, Muthusamy N, Byrd JC, Kulp SK, Chen CS. Antitumor effects of OSU- 2S, a non-immunosuppressive analogue of FTY720, in hepatocellular carcinoma. Hepatology 2011; 53(6):1943-58 6.Omar HA, Berman-Booty LD, Kulp SK, Chen CS. Energy restriction as an antitumor target. Future Oncol 2010; 6(11): 1675-9 7.Berman-Booty LD, Cui J, Horvath SJ, Premanandan C. Pathology in Practice. Tularemia. J Am Vet Med Assoc 2010; 237(2): 163-5 8.Zaph C, Troy AE, Taylor BC, Berman-Booty LD, Guild KJ, Du Y, Yost EA, Gruber AD, May MJ, Greten FR, Eckmann L, Karin M, Artis D. Epithelial-cell-intrinsic IKK- beta expression regulates intestinal immune homeostasis. Nature 2007; 446(7135): 552-6 9.Blais MC, Berman L, Oakley D, and Giger U. The canine DAL blood type: a red cell antigen lacking in some Dalmatians. J Vet Intern Med 2007; 21(2): 281-6. ix FIELDS OF STUDY Major Field: Veterinary Biosciences x TABLE OF CONTENTS Abstract ............................................................................................................................... ii Dedication ........................................................................................................................... v Acknowledgments.............................................................................................................. vi Vita .................................................................................................................................. viiii List of Tables ................................................................................................................... xiv List of Figures .................................................................................................................. xvi Chapter 1: A review of the existing grading schemes and a proposal for a modified grading scheme for prostatic lesions in TRAMP mice ....................................................... 1 Abstract ........................................................................................................................... 1 Introduction
Recommended publications
  • Mir-379 Deletion Ameliorates Features of Diabetic Kidney Disease By
    ARTICLE https://doi.org/10.1038/s42003-020-01516-w OPEN miR-379 deletion ameliorates features of diabetic kidney disease by enhancing adaptive mitophagy via FIS1 ✉ Mitsuo Kato 1,8 , Maryam Abdollahi 1,8, Ragadeepthi Tunduguru1, Walter Tsark2, Zhuo Chen 1, 1234567890():,; Xiwei Wu3, Jinhui Wang3, Zhen Bouman Chen 1,4, Feng-Mao Lin1, Linda Lanting1, Mei Wang1, Janice Huss5, ✉ Patrick T Fueger 5,6, David Chan7 & Rama Natarajan 1,4 Diabetic kidney disease (DKD) is a major complication of diabetes. Expression of members of the microRNA (miRNA) miR-379 cluster is increased in DKD. miR-379, the most upstream 5′-miRNA in the cluster, functions in endoplasmic reticulum (ER) stress by targeting EDEM3. However, the in vivo functions of miR-379 remain unclear. We created miR-379 knockout (KO) mice using CRISPR-Cas9 nickase and dual guide RNA technique and characterized their phenotype in diabetes. We screened for miR-379 targets in renal mesangial cells from WT vs. miR-379KO mice using AGO2-immunopreciptation and CLASH (cross-linking, ligation, sequencing hybrids) and identified the redox protein thioredoxin and mitochondrial fission-1 protein. miR-379KO mice were protected from features of DKD as well as body weight loss associated with mitochondrial dysfunction, ER- and oxidative stress. These results reveal a role for miR-379 in DKD and metabolic processes via reducing adaptive mitophagy. Strate- gies targeting miR-379 could offer therapeutic options for DKD. 1 Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA.
    [Show full text]
  • Analysis of Trans Esnps Infers Regulatory Network Architecture
    Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2014 © 2014 Anat Kreimer All rights reserved ABSTRACT Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer eSNPs are genetic variants associated with transcript expression levels. The characteristics of such variants highlight their importance and present a unique opportunity for studying gene regulation. eSNPs affect most genes and their cell type specificity can shed light on different processes that are activated in each cell. They can identify functional variants by connecting SNPs that are implicated in disease to a molecular mechanism. Examining eSNPs that are associated with distal genes can provide insights regarding the inference of regulatory networks but also presents challenges due to the high statistical burden of multiple testing. Such association studies allow: simultaneous investigation of many gene expression phenotypes without assuming any prior knowledge and identification of unknown regulators of gene expression while uncovering directionality. This thesis will focus on such distal eSNPs to map regulatory interactions between different loci and expose the architecture of the regulatory network defined by such interactions. We develop novel computational approaches and apply them to genetics-genomics data in human. We go beyond pairwise interactions to define network motifs, including regulatory modules and bi-fan structures, showing them to be prevalent in real data and exposing distinct attributes of such arrangements. We project eSNP associations onto a protein-protein interaction network to expose topological properties of eSNPs and their targets and highlight different modes of distal regulation.
    [Show full text]
  • View of HER2: Human Epidermal Growth Factor Receptor 2; TNBC: Triple-Negative Breast Resistance to Systemic Therapy in Patients with Breast Cancer
    Wen et al. Cancer Cell Int (2018) 18:128 https://doi.org/10.1186/s12935-018-0625-9 Cancer Cell International PRIMARY RESEARCH Open Access Sulbactam‑enhanced cytotoxicity of doxorubicin in breast cancer cells Shao‑hsuan Wen1†, Shey‑chiang Su2†, Bo‑huang Liou3, Cheng‑hao Lin1 and Kuan‑rong Lee1* Abstract Background: Multidrug resistance (MDR) is a major obstacle in breast cancer treatment. The predominant mecha‑ nism underlying MDR is an increase in the activity of adenosine triphosphate (ATP)-dependent drug efux trans‑ porters. Sulbactam, a β-lactamase inhibitor, is generally combined with β-lactam antibiotics for treating bacterial infections. However, sulbactam alone can be used to treat Acinetobacter baumannii infections because it inhibits the expression of ATP-binding cassette (ABC) transporter proteins. This is the frst study to report the efects of sulbactam on mammalian cells. Methods: We used the breast cancer cell lines as a model system to determine whether sulbactam afects cancer cells. The cell viabilities in the present of doxorubicin with or without sulbactam were measured by MTT assay. Protein identities and the changes in protein expression levels in the cells after sulbactam and doxorubicin treatment were determined using LC–MS/MS. Real-time reverse transcription polymerase chain reaction (real-time RT-PCR) was used to analyze the change in mRNA expression levels of ABC transporters after treatment of doxorubicin with or without sulbactam. The efux of doxorubicin was measures by the doxorubicin efux assay. Results: MTT assay revealed that sulbactam enhanced the cytotoxicity of doxorubicin in breast cancer cells. The results of proteomics showed that ABC transporter proteins and proteins associated with the process of transcription and initiation of translation were reduced.
    [Show full text]
  • Crystal Structure of Human Gadd45 Reveals an Active Dimer
    Protein Cell 2011, 2(10): 814–826 Protein & Cell DOI 10.1007/s13238-011-1090-6 RESEARCH ARTICLE Crystal structure of human Gadd45 reveals an active dimer 1,2,3* 1* 4 4 1 1 3 Wenzheng Zhang , Sheng Fu , Xuefeng Liu , Xuelian Zhao✉ , Wenchi Zhang✉ , Wei Peng , Congying Wu , Yuanyuan Li3, Xuemei Li1, Mark Bartlam1,2, Zong-Hao Zeng1 , Qimin Zhan4 , Zihe Rao1,2,3 1 National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China 2 Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin 300071, China 3 Laboratory of Structural Biology, Tsinghua University, Beijing 100084, China 4 State Key Laboratory of Molecular Oncology, Cancer Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China ✉ Correspondence: [email protected] (Z.-H. Zeng), [email protected] (Q. Zhan) Received August 23, 2011 Accepted September 19, 2011 ABSTRACT INTRODUCTION The human Gadd45 protein family plays critical roles in The human Gadd45 family of growth arrest and DNA DNA repair, negative growth control, genomic stability, damage-inducible proteins has three members, Gadd45α, cell cycle checkpoints and apoptosis. Here we report the -β, and -γ, grouped together on the basis of their amino acid crystal structure of human Gadd45, revealing a unique sequences and functional similarities (Vairapandi et al., 1996; dimer formed via a bundle of four parallel helices, Takekawa and Saito, 1998; Nakayama et al., 1999). The involving the most conserved residues among the genes encoding the α, β and γ isoforms of Gadd45 are Gadd45 isoforms.
    [Show full text]
  • Amwands 1.Pdf
    CHARACTERIZATION OF THE DYNAMIC INTERACTIONS OF TRANSCRIPTIONAL ACTIVATORS by Amberlyn M. Wands A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Chemistry) in The University of Michigan 2010 Doctoral Committee: Associate Professor Anna K. Mapp, Chair Professor Hashim M. Al-Hashimi Professor E Neil G. Marsh Associate Professor Jorge A. Iñiguez-Lluhí Amberlyn M. Wands All rights reserved 2010 Acknowledgements I have so many people to thank for helping me throughout my graduate school career. First, I would like to thank my advisor Dr. Anna Mapp for all of the guidance you have given me, as well as allowing me the freedom to express myself as a scientist. Your patience and confidence in my abilities means a lot to me, and I promise to keep working on presenting myself to others in a positive yet assertive manner. I would also like to thank you for taking the time to instill in your students the importance of thinking and writing critically about scientific concepts, which I know we will carry with us into our future careers. Next I would like to thank my committee members for their time, and for always asking me challenging questions that made me look at my projects from a different perspective. I would also like to give a special thanks to Dr. Carol Fierke and Dr. John Hsieh for their willingness to work on a collaboration with people starting with a minimal background in the field of transient kinetics. Their love of solving kinetic problems is inspiring, and I appreciate being given the opportunity to work with them.
    [Show full text]
  • Mouse Letmd1 Conditional Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Letmd1 Conditional Knockout Project (CRISPR/Cas9) Objective: To create a Letmd1 conditional knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Letmd1 gene (NCBI Reference Sequence: NM_134093 ; Ensembl: ENSMUSG00000037353 ) is located on Mouse chromosome 15. 9 exons are identified, with the ATG start codon in exon 1 and the TGA stop codon in exon 9 (Transcript: ENSMUST00000037001). Exon 5~7 will be selected as conditional knockout region (cKO region). Deletion of this region should result in the loss of function of the Mouse Letmd1 gene. To engineer the targeting vector, homologous arms and cKO region will be generated by PCR using BAC clone RP23-81M8 as template. Cas9, gRNA and targeting vector will be co-injected into fertilized eggs for cKO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Exon 5 starts from about 43.89% of the coding region. The knockout of Exon 5~7 will result in frameshift of the gene. The size of intron 4 for 5'-loxP site insertion: 2391 bp, and the size of intron 7 for 3'-loxP site insertion: 1135 bp. The size of effective cKO region: ~1132 bp. The cKO region does not have any other known gene. Page 1 of 8 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele 5' gRNA region gRNA region 3' 1 5 6 7 8 9 Targeting vector Targeted allele Constitutive KO allele (After Cre recombination) Legends Exon of mouse Letmd1 Homology arm cKO region loxP site Page 2 of 8 https://www.alphaknockout.com Overview of the Dot Plot Window size: 10 bp Forward Reverse Complement Sequence 12 Note: The sequence of homologous arms and cKO region is aligned with itself to determine if there are tandem repeats.
    [Show full text]
  • Environmental Influences on Endothelial Gene Expression
    ENDOTHELIAL CELL GENE EXPRESSION John Matthew Jeff Herbert Supervisors: Prof. Roy Bicknell and Dr. Victoria Heath PhD thesis University of Birmingham August 2012 University of Birmingham Research Archive e-theses repository This unpublished thesis/dissertation is copyright of the author and/or third parties. The intellectual property rights of the author or third parties in respect of this work are as defined by The Copyright Designs and Patents Act 1988 or as modified by any successor legislation. Any use made of information contained in this thesis/dissertation must be in accordance with that legislation and must be properly acknowledged. Further distribution or reproduction in any format is prohibited without the permission of the copyright holder. ABSTRACT Tumour angiogenesis is a vital process in the pathology of tumour development and metastasis. Targeting markers of tumour endothelium provide a means of targeted destruction of a tumours oxygen and nutrient supply via destruction of tumour vasculature, which in turn ultimately leads to beneficial consequences to patients. Although current anti -angiogenic and vascular targeting strategies help patients, more potently in combination with chemo therapy, there is still a need for more tumour endothelial marker discoveries as current treatments have cardiovascular and other side effects. For the first time, the analyses of in-vivo biotinylation of an embryonic system is performed to obtain putative vascular targets. Also for the first time, deep sequencing is applied to freshly isolated tumour and normal endothelial cells from lung, colon and bladder tissues for the identification of pan-vascular-targets. Integration of the proteomic, deep sequencing, public cDNA libraries and microarrays, delivers 5,892 putative vascular targets to the science community.
    [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]
  • RNF2 Antibody Order 021-34695924 [email protected] Support 400-6123-828 50Ul [email protected] 100 Ul √ √ Web
    TD7403 RNF2 Antibody Order 021-34695924 [email protected] Support 400-6123-828 50ul [email protected] 100 uL √ √ Web www.ab-mart.com.cn Description: E3 ubiquitin-protein ligase that mediates monoubiquitination of 'Lys-119' of histone H2A (H2AK119Ub), thereby playing a central role in histone code and gene regulation. H2AK119Ub gives a specific tag for epigenetic transcriptional repression and participates in X chromosome inactivation of female mammals. May be involved in the initiation of both imprinted and random X inactivation (By similarity). Essential component of a Polycomb group (PcG) multiprotein PRC1-like complex, a complex class required to maintain the transcriptionally repressive state of many genes, including Hox genes, throughout development. PcG PRC1 complex acts via chromatin remodeling and modification of histones, rendering chromatin heritably changed in its expressibility. E3 ubiquitin-protein ligase activity is enhanced by BMI1/PCGF4. Acts as the main E3 ubiquitin ligase on histone H2A of the PRC1 complex, while RING1 may rather act as a modulator of RNF2/RING2 activity (Probable). Association with the chromosomal DNA is cell-cycle dependent. In resting B- and T-lymphocytes, interaction with AURKB leads to block its activity, thereby maintaining transcription in resting lymphocytes (By similarity). Uniprot:Q99496 Alternative Names: BAP 1; BAP1; DING; DinG protein; E3 ubiquitin protein ligase RING 2; E3 ubiquitin protein ligase RING2; E3 ubiquitin-protein ligase RING2; HIP2 interacting protein 3; HIP2- interacting protein 3; HIPI 3; HIPI3; Huntingtin interacting protein 2 interacting protein 3; Huntingtin-interacting protein 2-interacting protein 3; OTTHUMP00000060668; Polycomb M33 interacting protein Ring 1B; Polycomb M33 interacting protein Ring1B; Protein DinG; RING 1B; RING 2; RING finger protein 1B; RING finger protein 2; RING finger protein BAP 1; RING finger protein BAP-1; RING finger protein BAP1; RING1b; RING2_HUMAN; RNF 2; Rnf2; Specificity: RNF2 Antibody detects endogenous levels of total RNF2.
    [Show full text]
  • Product Data Sheet
    For research purposes only, not for human use Product Data Sheet RNF2 siRNA (Human) Catalog # Source Reactivity Applications CRH4092 Synthetic H RNAi Description siRNA to inhibit RNF2 expression using RNA interference Specificity RNF2 siRNA (Human) is a target-specific 19-23 nt siRNA oligo duplexes designed to knock down gene expression. Form Lyophilized powder Gene Symbol RNF2 Alternative Names BAP1; DING; HIPI3; RING1B; E3 ubiquitin-protein ligase RING2; Huntingtin-interacting protein 2-interacting protein 3; HIP2-interacting protein 3; Protein DinG; RING finger protein 1B; RING1b; RING finger protein 2; RING finger protein BAP-1 Entrez Gene 6045 (Human) SwissProt Q99496 (Human) Purity > 97% Quality Control Oligonucleotide synthesis is monitored base by base through trityl analysis to ensure appropriate coupling efficiency. The oligo is subsequently purified by affinity-solid phase extraction. The annealed RNA duplex is further analyzed by mass spectrometry to verify the exact composition of the duplex. Each lot is compared to the previous lot by mass spectrometry to ensure maximum lot-to-lot consistency. Components We offers pre-designed sets of 3 different target-specific siRNA oligo duplexes of human RNF2 gene. Each vial contains 5 nmol of lyophilized siRNA. The duplexes can be transfected individually or pooled together to achieve knockdown of the target gene, which is most commonly assessed by qPCR or western blot. Our siRNA oligos Application key: E- ELISA, WB- Western blot, IH- Immunohistochemistry, IF- Immunofluorescence, FC-
    [Show full text]
  • Snps) Distant from Xenobiotic Response Elements Can Modulate Aryl Hydrocarbon Receptor Function: SNP-Dependent CYP1A1 Induction S
    Supplemental material to this article can be found at: http://dmd.aspetjournals.org/content/suppl/2018/07/06/dmd.118.082164.DC1 1521-009X/46/9/1372–1381$35.00 https://doi.org/10.1124/dmd.118.082164 DRUG METABOLISM AND DISPOSITION Drug Metab Dispos 46:1372–1381, September 2018 Copyright ª 2018 by The American Society for Pharmacology and Experimental Therapeutics Single Nucleotide Polymorphisms (SNPs) Distant from Xenobiotic Response Elements Can Modulate Aryl Hydrocarbon Receptor Function: SNP-Dependent CYP1A1 Induction s Duan Liu, Sisi Qin, Balmiki Ray,1 Krishna R. Kalari, Liewei Wang, and Richard M. Weinshilboum Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (D.L., S.Q., B.R., L.W., R.M.W.) and Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.K.), Mayo Clinic, Rochester, Minnesota Received April 22, 2018; accepted June 28, 2018 ABSTRACT Downloaded from CYP1A1 expression can be upregulated by the ligand-activated aryl fashion. LCLs with the AA genotype displayed significantly higher hydrocarbon receptor (AHR). Based on prior observations with AHR-XRE binding and CYP1A1 mRNA expression after 3MC estrogen receptors and estrogen response elements, we tested treatment than did those with the GG genotype. Electrophoretic the hypothesis that single-nucleotide polymorphisms (SNPs) map- mobility shift assay (EMSA) showed that oligonucleotides with the ping hundreds of base pairs (bp) from xenobiotic response elements AA genotype displayed higher LCL nuclear extract binding after (XREs) might influence AHR binding and subsequent gene expres- 3MC treatment than did those with the GG genotype, and mass dmd.aspetjournals.org sion.
    [Show full text]
  • Title: Therapeutic Potential of HSP90 Inhibition for Neurofibromatosis Type 2
    Author Manuscript Published OnlineFirst on May 28, 2013; DOI: 10.1158/1078-0432.CCR-12-3167 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Title: Therapeutic Potential of HSP90 Inhibition for Neurofibromatosis type 2 Karo Tanaka1, Ascia Eskin3, Fabrice Chareyre1, Walter J. Jessen4, Jan Manent5, Michiko Niwa-Kawakita6, Ruihong Chen7, Cory H. White2, Jeremie Vitte1, Zahara M. Jaffer1, Stanley F. Nelson3, Allan E. Rubenstein8, Marco Giovannini1,9§. Authors’ affiliations: House Research Institute, 1Center for Neural Tumor Research and 2Section on Genetics of Hereditary Ear Disorders, Los Angeles, CA; 3Department of Human Genetics, University of California, Los Angeles, CA; 4Informatics, Covance Inc., Princeton, NJ; 5Peter MacCallum Cancer Institute, Melbourne, Australia; 6Inserm U944, CNRS U7212, Université Paris, Institut Universitaire d'Hématologie, Paris, France; 7NexGenix Pharmaceuticals, Burlingame, CA; and 8New York University Langone Medical Center, New York, NY; and Department of Cell and Neurobiology, University of Southern California, Keck School of Medicine, Los Angeles, CA Running title: HSP90 Inhibition for NF2 Keywords: NF2, HSP90 inhibitors, Transcriptome Financial support: This work was supported by a Drug Discovery Initiative Award, Children’s Tumor Foundation, to M.G., and by the House Research Institute. Corresponding author: Marco Giovannini, House Research Institute, Center for Neural Tumor Research, 2100 West 3rd street, Los Angeles, CA90057. Phone: +1-213-989-6708; Fax: +1-213-989-6778; E-mail: [email protected] 1 Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on May 28, 2013; DOI: 10.1158/1078-0432.CCR-12-3167 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
    [Show full text]