Isatin Inhibits the Invasion of Human Neuroblastoma SH‑SY5Y Cells, Based on Microarray Analysis

Total Page:16

File Type:pdf, Size:1020Kb

Isatin Inhibits the Invasion of Human Neuroblastoma SH‑SY5Y Cells, Based on Microarray Analysis 1700 MOLECULAR MEDICINE REPORTS 20: 1700-1706, 2019 Isatin inhibits the invasion of human neuroblastoma SH‑SY5Y cells, based on microarray analysis LI ZHANG1,2*, WENYAN SUN1,3*, YI CAO1, LIN HOU1, CHUANXIA JU2 and XUEFENG WANG3 1Department of Biochemistry, Medical College; 2Experimental Center for Undergraduates of Pharmacy, School of Pharmacy; 3Institute of Metabolic Diseases, Qingdao University, Qingdao, Shandong 266021, P.R. China Received March 22, 2018; Accepted April 24, 2019 DOI: 10.3892/mmr.2019.10378 Abstract. Neuroblastoma is the fourth most common type of treatment for high-risk NB. New therapeutic approaches need extracranial malignant solid tumor in children. Isatin had been to be developed (2). demonstrated to have inhibitory effects on neuroblastoma Clinical trial results have demonstrated that multi-target tumors in vivo and in vitro. The aim of the present study was to inhibitor drugs are more effective compared with single-target investigate the molecular mechanism related to the anti-inva- drugs during cancer treatment (5). 1H-indole-2, 3-dione sion effect of isatin on SH-SY5Y cells using microarray (isatin) is a derivative of the anti-cancer drug indirubin, which analysis. The microarray data identified a number of genes exhibits beneficial biological activities, including antibacterial, to be differentially upregulated or downregulated between antifungal and antitumor properties (6-8). Isatin was initially isatin-treated cells and untreated controls. A large number of considered to be an inhibitor of monoamine oxidase-b and has these genes were associated with the mTOR signaling pathway. been used to treat Parkinson's disease (9,10). Recent studies The differentially expressed genes involved in the mTOR have reported that isatin inhibits cell proliferation and invasion signaling pathway were verified further, as well as their down- in human neuroblastoma SH-SY5Y cells (11,12). stream genes associated with autophagy. The results of the mTOR has been identified as a key molecule in tumori- present study provided an insight into the potential inhibitory genesis and cancer progression (13). Increasing evidence mechanism of isatin on neuroblastoma metastasis. has identified the mTOR pathway as a relevant target for the suppression of tumorigenesis; thus, inhibition of mTOR Introduction may be a promising method for targeting human malignan- cies (14). The aim of the present study was to investigate the Neuroblastoma (NB) is the most common extracranial solid underlying molecular mechanisms of the inhibitory effect of cancer in childhood and infancy, with a mortality rate of 15%, isatin on migration and invasion in SH-SY5Y cells, which are and an incidence between 5.9 and 10.5 per million children associated with the mTOR pathway. under 15 years of age (1). The majority of NB cases are meta- static, and thus are associated with a poor prognosis and a Materials and methods high mortality rate (2). A total of 20-50% of high-risk cases do not respond adequately to high-dose chemotherapy and Cell culture. Human neuroblastoma SH-SY5Y cells (human are progressive or refractory (3). Novel treatments using new origin) were purchased from Peking Union Medical agents and combinations against NB are available in phase I College. The cell line had been STR authenticated (ATCC or II clinical trials, but the outcomes remain poor (4). Single no. CRL‑2266). Cells were cultured in DMEM (HyClone; GE targeted agents are unlikely to be sufficient for long-term Healthcare Life Sciences) supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) at 37˚C in a humidified atmosphere with 5% CO2. Isatin was dissolved in 0.1% dimethyl sulfoxide and added to 6-well plates at concen- trations of 50, 100, 200 and 300 µM, which were determined based on our previous study (11), when the cells had reached Correspondence to: Professor Lin Hou, Department of ~70% confluence. The cells (~2x106/well) were harvested Biochemistry, Medical College, Qingdao University, 38 Dengzhou following isatin treatment at 37˚C for 48 h for further analysis. Road, Qingdao, Shandong 266021, P.R. China E‑mail: [email protected] Cell migration and invasion assay. Cells (1x106) were seeded *Contributed equally in 6-well culture plates and exposed to isatin (200 µM) with or without rapamycin (10 µM) at 37˚C for 48 h. The concentration Key words: isatin, neuroblastoma, invasion, mTOR signaling of isatin was determined according to previous results, which pathway demonstrated that 200 µM isatin did not induce necrosis of SH-SY5Y cells (11). A wound healing assay was performed to monitor cell invasion, as previously described (11). For the ZHANG et al: IsATIN INHIBIts THE INVASION OF HUMAN NEUROBLAstOMA SH-SY5Y CELLS 1701 Transwell invasion assay, isatin-treated cells were seeded in expressed as fold change compared with control. The 2-ΔΔCq Boyden chambers (EMD Millipore) with and without Matrigel, method was employed to analyze the relative expression of and the assay was performed as previously described (11). genes (22). Cells were counted in at least five fields under an inverted light microscope (magnification, x10/x20). All experiments were Protein extraction and western blot analysis. Total protein performed in triplicate. was extracted from SH-SY5Y cells using RIPA lysis buffer (Beyotime Institute of Biotechnology) following isatin treat- Microarray analysis. Microarray expression analysis was ment for 48 h , a n d t h e p r o t e i n c o n c e n t r a t i o n w a s q u a n t i fi e d u s i ng performed using total RNA from SH-SY5Y cells that were a bicinchoninic acid protein assay. Proteins (20-40 µg/lane) incubated at 37˚C with or without 200 µM isatin for 48 h. were separated on SDS-polyacrylamide gels (8-12%) and Total RNA was extracted using TRIzol® (Beyotime Institute transferred onto PVDF membranes. Membranes were blocked of Biotechnology). An Affymetrix Microarray kit (Thermo with 5% bovine serum albumin for 2 h at room temperature, Fisher Scientific, Inc.) was used for the gene expression and then incubated with the following primary antibodies analysis. Raw data intensities were quantile-normalized, and diluted in 5% BSA: β-actin (1:3,000; cat. no. ab8227; Abcam), genes exhibiting significantly higher intensities compared AMPK (1:500; cat. no. ab131512; Abcam), phosphorylated with the background and a fold-change (FC)>1.5 following (p)‑AMPK (1:500; cat. no. ab131357; Abcam), mTOR (1:300; isatin treatment, compared with the control, were selected. cat. no. ab32028; Abcam), p-mTOR (1:5,000; cat. no. ab109268; This selection uncovered a total of 429 genes. Expression Abcam), microtubule associated protein 1 light chain 3α values of the genes were rescaled to mean 0 and SD 1, and (LC3; 1:1,000; cat. no. 3868; Cell Signaling Technology), hierarchical clustering was performed using Ward's algo- Beclin-1 (1:1,000; cat. no. 3738; Cell Signaling Technology) rithm (15). The number of clusters was determined using the and p62 (1:1,000; cat. no. 5114; Cell Signaling Technology) Akaike Information Criterion (16). A Mann-Whitney test was at 4˚C overnight, washed with TBS with Tween‑20, and subse- performed to discriminate clusters of genes with differential quently incubated with horseradish peroxidase-conjugated expression levels between the two groups. Differentially goat anti-rabbit immunoglobulin G secondary antibodies expressed genes were selected based on FC in average gene (1:1,000; cat. no. HS101-01; Beijing Transgen Biotech Co., expression with P<0.05, as determined by Student's t-test. Ltd.) for 1 h at room temperature. Protein expression levels Gene Ontology (GO) enrichment (‘biological process’, were detected using the Enhanced Chemiluminescence Plus ‘cellular component’ and ‘molecular function’) (17,18) and kit (Wuhan Boster Biological Technology, Ltd.) and densito- Kyoto Encyclopedia of Genes and Genomes (KEGG) path- metric analysis was performed using Quantity One analysis ways (19-21) of differentially expressed genes were analyzed software (version 4.52; Bio-Rad Laboratories, Inc.). using the Database for Annotation, Visualization and Integrated Discovery Bioinformatics Resources, version 6.7 Statistical analysis. Each experiment was performed at least (http://david.abcc.ncifcrf.gov). Subsequent selection of three times. Data are expressed as the mean ± SD. Multiple enriched GO terms was performed based on the calculated comparisons were performed by one-way ANOVA followed q-values using the threshold q<0.05. by a least significant difference or Tamhane's T2 post hoc test in SPSS 22.0 (IBM Corp.). P<0.05 was considered to indicate Reverse transcription‑qPCR (RT‑qPCR). Total RNA a statistically significant difference. of SH-SY5Y cells was extracted using TRIzol and was reverse transcribed to cDNA using TransScript™ Reverse Results Transcriptase (Beijing Transgen Biotech Co., Ltd.) at 42˚C for 15 min and 85˚C for 5 sec. Then, qPCR was performed Isatin treatment affects gene expression in SH‑SY5Y cells. To independently three times using SYBR Green mix (Takara investigate the underlying molecular events of the anti-inva- Bio, Inc.). Gene expression was analyzed using the following sive activity of isatin in SH-SY5Y cells, RNA was extracted primers: β-actin forward, 5'-GCG TGA​CAT TAA​GGA​ from SH-SY5Y cells (untreated or treated with 200 µM isatin CAA​GC‑3' and reverse, 5'‑CCA​CGT​CAC​ACT TCA​TGA​ for 48 h) for Affymetrix cDNA microarray analysis. The TGG-3'; mTOR forward, 5'‑CTG GGA​CTC​AAA​TGT GTG GeneChip results revealed 284 differentially upregulated and CAG TTC‑3' and reverse, 5'-GAA​CAA​TAG GGT GAA​TGA​ 145 downregulated genes between cells treated with isatin and TCC​GGG-3'; ribosomal protein S6 kinase B1 (RPS6KB1) controls (FC>1.5; Fig. 1A). GO term analysis demonstrated forward, 5'-GGT GGA​GTT TGG GAG​CAT TA‑3' and reverse, that the differentially expressed genes were involved in redox 5'-TGT GAG GTA​GGG AGG​CAA​AT-3'; eukaryotic transla- activity, binding and transcription function, cell metabolism tion initiation factor 4E‑binding protein 1 (EIF4EBP1) and transport (data not shown).
Recommended publications
  • EGFR and Mtor As Therapeutic Targets in Glioblastoma
    www.oncotarget.com Oncotarget, 2019, Vol. 10, (No. 46), pp: 4721-4723 Editorial EGFR and mTOR as therapeutic targets in glioblastoma Michael W. Ronellenfitsch, Anna-Luisa Luger and Joachim P. Steinbach The quest for new and improved therapies for mammalian target of rapamycin complex 1 (mTORC1) glioblastoma (GB) has been mostly unsuccessful in signaling were found in the majority of GBs [3]. more than a decade despite significant efforts. The few Therefore, many hopes have rested on targeted therapies. exceptions include the optimization of classical alkylating However, the results from clinical trials have been largely chemotherapy by including lomustine in the first line disappointing [4]. Nevertheless, unplanned retrospective regimen for GB with a methylated MGMT promoter and subgroup analyses of the patient cohorts of negative tumor treating fields [1, 2]. The GB signaling network has clinical trials indicated that dysregulation or activation been well-characterized and genetic alterations resulting of signaling could be a predictive factor for susceptibility in activation of receptor tyrosine kinases and especially to pathway inhibition: Tumors with enhanced levels of epidermal growth factor receptor (EGFR) and downstream mTORC1 activation markers, including phosphorylated Figure 1: Scheme of EGFR signaling and DDIT4-mediated adaptive processes. Conditions of the glioblastoma microenvironment including hypoxia, alkylating therapy or irradiation trigger induction of DDIT4 which activates TSC1/2 to inhibit mTORC1 and can counteract epidermal growth factor receptor (EGFR)-mediated TSC1/2 inhibition. Inhibition of mTORC1 ultimately induces adaptive processes to cope with external stressors. www.oncotarget.com 4721 Oncotarget ribosomal protein S6 and phosphorylated mTOR itself, of effects. Depending on the half life and pharmacokinetics appeared to respond to pathway inhibition by the EGFR of the drugs, stepwise treatment algorithms could be an antibody nimotuzumab or the mTORC1 inhibitor option to prevent antagonistic effects.
    [Show full text]
  • Effects of Rapamycin on Social Interaction Deficits and Gene
    Kotajima-Murakami et al. Molecular Brain (2019) 12:3 https://doi.org/10.1186/s13041-018-0423-2 RESEARCH Open Access Effects of rapamycin on social interaction deficits and gene expression in mice exposed to valproic acid in utero Hiroko Kotajima-Murakami1,2, Toshiyuki Kobayashi3, Hirofumi Kashii1,4, Atsushi Sato1,5, Yoko Hagino1, Miho Tanaka1,6, Yasumasa Nishito7, Yukio Takamatsu7, Shigeo Uchino1,2 and Kazutaka Ikeda1* Abstract The mammalian target of rapamycin (mTOR) signaling pathway plays a crucial role in cell metabolism, growth, and proliferation. The overactivation of mTOR has been implicated in the pathogenesis of syndromic autism spectrum disorder (ASD), such as tuberous sclerosis complex (TSC). Treatment with the mTOR inhibitor rapamycin improved social interaction deficits in mouse models of TSC. Prenatal exposure to valproic acid (VPA) increases the incidence of ASD. Rodent pups that are exposed to VPA in utero have been used as an animal model of ASD. Activation of the mTOR signaling pathway was recently observed in rodents that were exposed to VPA in utero, and rapamycin ameliorated social interaction deficits. The present study investigated the effect of rapamycin on social interaction deficits in both adolescence and adulthood, and gene expressions in mice that were exposed to VPA in utero. We subcutaneously injected 600 mg/kg VPA in pregnant mice on gestational day 12.5 and used the pups as a model of ASD. The pups were intraperitoneally injected with rapamycin or an equal volume of vehicle once daily for 2 consecutive days. The social interaction test was conducted in the offspring after the last rapamycin administration at 5–6 weeks of ages (adolescence) or 10–11 weeks of age (adulthood).
    [Show full text]
  • Accumulation of Dephosphorylated 4EBP After Mtor Inhibition with Rapamycin Is Sufficient to Disrupt Paracrine Transformation By
    Oncogene (2014) 33, 2405–2412 & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc SHORT COMMUNICATION Accumulation of dephosphorylated 4EBP after mTOR inhibition with rapamycin is sufficient to disrupt paracrine transformation by the KSHV vGPCR oncogene D Martin, Q Nguyen, A Molinolo and JS Gutkind Dysregulation of the PI3K/Akt/mTOR pathway is one of the most frequent events in human cancer. However, the clinical benefits of PI3K/Akt/mTOR inhibitors have not yet achieved their predicted potential in many of the most prevalent human cancers. Of interest, treatment of Kaposi’s sarcoma (KS) patients with rapamycin provided the first evidence of the antineoplastic activity of mTOR inhibitors in humans, becoming the standard of care for KS arising in renal transplant patients. Thus, the study of KS may provide a unique opportunity to dissect the contribution of specific mTOR downstream targets to cancer development. The KS- associated herpesvirus (KSHV) is the etiological agent for KS, and the KSHV-encoded oncogene viral-G protein-coupled receptor (vGPCR) promotes the potent activation of the PI3K-Akt-mTOR pathway by both direct and paracrine mechanisms. We focused on a direct target of mTOR, EIF4EBP1/2/3 (4EBP), which inhibits the translation of eukaryotic initiation factor 4E (eiF4E)-bound mRNAs. 4EBP phosphorylation by mTOR relieves its inhibitory activity, hence resulting in increased eiF4E-dependent mRNA translation. We developed a paracrine transformation model, recapitulating the cellular composition of KS lesions, in which vGPCR-expressing cells promote the rapid proliferation of endothelial cells, thus expressing KSHV-latent genes by the release of growth factors.
    [Show full text]
  • Adaptive Capabilities of the Pi3k/Akt/Mtor Pathway in Acute Myeloid Leukemia Revealed by the Use of Selective Inhibitors
    Alma Mater Studiorum – Università di Bologna SCIENZE BIOMEDICHE: PROGETTO 5 “SCIENZE MORFOLOGICHE UMANE E MOLECOLARI” Ciclo XXV SSD : BIO 16, SETTORE CONCORSUALE :05H1 ADAPTIVE CAPABILITIES OF THE PI3K/AKT/MTOR PATHWAY IN ACUTE MYELOID LEUKEMIA REVEALED BY THE USE OF SELECTIVE INHIBITORS Presentata da Jessika Bertacchini Relatori Prof.ssa Lucia Manzoli Prof.ssa Sandra Marmiroli Coordinatore Chiar. mo Prof. Lucio Cocco TABLE OF CONTENTS o ABSTRACT o INTRODUCTION o ACUTE MYELOID LEUKEMIA o PROGNOSIS AND GENETICS o DEREGULATED SIGNAL TRANSDUCTION PATHWAYS IN ACUTE MYELOID LEUKEMIA o PI3K/AKT/MTOR SIGNAL TRANSDUCTION PATHWAY IN ACUTE MYELOID LEUKEMIA o PI3K o AKT/PKB o Mtor o NEGATIVE REGULATION OF PI3K/AKT/mTOR PATHWAY o PI3K/AKT/mTOR PATHWAY AND SURVIVAL o PI3K/AKT/mTOR PATHWAY AND CELL CYCLE o PI3K/AKT/mTOR PATHWAY AND METABOLISM o PI3K/AKT/mTOR INHIBITORS o PI3K INHIBITORS o AKT INHIBITORS o mTOR INHIBITORS o TYROSINE KINASE RECEPTOR o TYROSINE KINASE RECEPTOR IN ACUTE MYELOID LEUKEMIA o AIMS o MATERIALS AND METHODS o PATIENTS DEMOGRAPHICS o CELL CULTURE AND DRUG TREATMENTS o ARRAY ASSEMBLY o RESULTS o RESULTS o DISCUSSION o BIBLIOGRA 1.ABSTRACT The objective of the study was to investigate the sensitivity of primary blasts from AML patients to PI3K/Akt/mTor inhibitors through reverse-phase protein microarray. Reverse-phase microarray assays using phosphospecific antibodies (RPPA) can directly measure levels of phosphorylated protein isoforms. Mapping of deregulated kinases and protein signaling networks within tumors can provide a means to stratify patients with shared biological characteristics to the most optimal treatment, and identify drug targets. In particular, the PI3K/AKT/mTOR signaling pathways are frequently activated in blast cells from patients with acute myelogenous leukemia (AML), a neoplastic disorder characterized by the accumulation of genetically altered myelogenous cells displaying deregulated intracellular signalling pathways and aggressive clinical behavior with poor prognosis.
    [Show full text]
  • Transcriptional Changes in Huntington Disease Identified Using Genome
    Human Molecular Genetics, 2010, Vol. 19, No. 8 1438–1452 doi:10.1093/hmg/ddq018 Advance Access published on January 20, 2010 Transcriptional changes in Huntington disease identified using genome-wide expression profiling and cross-platform analysis Kristina Becanovic1, Mahmoud A. Pouladi1, Raymond S. Lim2, Alexandre Kuhn3, Paul Pavlidis2, Ruth Luthi-Carter3, Michael R. Hayden1 and Blair R. Leavitt1,Ã 1Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada V5Z 4H4 2Centre for High-throughput Biology and Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada V6T 1Z4 and 3Brain Mind Institute, E´ cole Polytechnique Fe´de´rale de Lausanne (EPFL), Station 15, CH1015 Lausanne, Switzerland Received September 10, 2009; Revised and Accepted January 18, 2010 Evaluation of transcriptional changes in the striatum may be an effective approach to understanding the natural history of changes in expression contributing to the pathogenesis of Huntington disease (HD). We have performed genome-wide expression profiling of the YAC128 transgenic mouse model of HD at 12 and 24 months of age using two platforms in parallel: Affymetrix and Illumina. The data from these two powerful platforms were integrated to create a combined rank list, thereby revealing the identity of additional genes that proved to be differentially expressed between YAC128 and control mice. Using this approach, we ident- ified 13 genes to be differentially expressed between YAC128 and controls which were validated by quanti- tative real-time PCR in independent cohorts of animals. In addition, we analyzed additional time points relevant to disease pathology: 3, 6 and 9 months of age.
    [Show full text]
  • Comparative Mapping of Bovine Chromosome 27 with Human Chromosome 8 Near a Dairy Form QTL in Cattle
    University of Nebraska - Lincoln DigitalCommons@University of Nebraska - Lincoln U.S. Department of Agriculture: Agricultural Publications from USDA-ARS / UNL Faculty Research Service, Lincoln, Nebraska 2006 Comparative mapping of bovine chromosome 27 with human chromosome 8 near a dairy form QTL in cattle E. E. Connor USDA-ARS M. S. Ashwell North Carolina State University at Raleigh R. Schnabel University of Missouri-Columbia J. L. Williams Roslin Institute (Edinburgh) Follow this and additional works at: https://digitalcommons.unl.edu/usdaarsfacpub Part of the Agricultural Science Commons Connor, E. E.; Ashwell, M. S.; Schnabel, R.; and Williams, J. L., "Comparative mapping of bovine chromosome 27 with human chromosome 8 near a dairy form QTL in cattle" (2006). Publications from USDA-ARS / UNL Faculty. 698. https://digitalcommons.unl.edu/usdaarsfacpub/698 This Article is brought to you for free and open access by the U.S. Department of Agriculture: Agricultural Research Service, Lincoln, Nebraska at DigitalCommons@University of Nebraska - Lincoln. It has been accepted for inclusion in Publications from USDA-ARS / UNL Faculty by an authorized administrator of DigitalCommons@University of Nebraska - Lincoln. Original Article Cytogenet Genome Res 112:98–102 (2006) DOI: 10.1159/000087519 Comparative mapping of bovine chromosome 27 with human chromosome 8 near a dairy form QTL in cattle E.E. Connor,a M.S. Ashwell,a,b R. Schnabel,c J.L. Williamsd a Beltsville Agricultural Research Center, ARS, USDA, Beltsville, MD; b North Carolina State University, Department of Animal Science, Raleigh, NC; c University of Missouri-Columbia, Animal Sciences Unit, Columbia, MO (USA); d Roslin Institute (Edinburgh), Roslin, Midlothian, Scotland (UK) Manuscript received 30 December 2004; accepted in revised form for publication by T.
    [Show full text]
  • An Integrative Transcriptome Study Reveals Ddit4/Redd1 As a Key Regulator of Cancer Cachexia in Rodent Models
    www.nature.com/cddis ARTICLE OPEN An integrative transcriptome study reveals Ddit4/Redd1 as a key regulator of cancer cachexia in rodent models 1,2,4 1,2,4 3 1,2 1,2 1,2 2 1,2 Mengyuan Niu ,LiLi , Zhonglan✉ Su , Lulu Wei✉ , Wenyuan Pu , Chen Zhao✉ , Yibing Ding , Junaid Wazir , Wangsen Cao 2, Shiyu Song 1,2 , Qian Gao 2 and Hongwei Wang 1,2 © The Author(s) 2021 Cancer cachexia is a multifactorial metabolic syndrome that causes up to 20% of cancer-related deaths. Muscle atrophy, the hallmark of cancer cachexia, strongly impairs the quality of life of cancer patients; however, the underlying pathological process is still poorly understood. Investigation of the disease pathogenesis largely relies on cachectic mouse models. In our study, the transcriptome of the cachectic gastrocnemius muscle in the C26 xenograft model was integrated and compared with that of 5 more different datasets. The bioinformatic analysis revealed pivotal gene ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of the disease, and the key genes were validated. Construction of the protein-protein interaction network and the comparison of pathways enriched in cancer cachexia with 5 other muscle atrophy models revealed Ddit4 (DNA damage-inducible transcript 4), as a key protein in cancer cachexia. The higher expression of Ddit4 in cachectic muscle was further validated in animal models and cachectic cancer patients. Further study revealed that p38 induced the expression of Ddit4, which in turn inhibited the mTOR pathway in atrophic cells. Cell Death and Disease (2021)12:652 ; https://doi.org/10.1038/s41419-021-03932-0 INTRODUCTION the overall transcription profile between cancer or noncancer- Cancer cachexia is a multifactorial metabolic syndrome that affects induced muscle atrophy will be greatly helpful in revealing the approximately 50–80% of cancer patients, especially in the uniqueness of the disease.
    [Show full text]
  • DDIT4 Antibody (N-Term) Purified Rabbit Polyclonal Antibody (Pab) Catalog # AP6268A
    10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 DDIT4 Antibody (N-term) Purified Rabbit Polyclonal Antibody (Pab) Catalog # AP6268A Specification DDIT4 Antibody (N-term) - Product Information Application WB, IHC-P,E Primary Accession Q9NX09 Reactivity Human Host Rabbit Clonality Polyclonal Isotype Rabbit Ig Antigen Region 20-49 DDIT4 Antibody (N-term) - Additional Information Gene ID 54541 Other Names DNA damage-inducible transcript 4 protein, HIF-1 responsive protein RTP801, Protein regulated in development and DNA damage Anti-DDIT4 Antibody (N-term) at 1:2000 response 1, REDD-1, DDIT4, REDD1, dilution + Hela whole cell lysate RTP801 Lysates/proteins at 20 µg per lane. Secondary Goat Anti-Rabbit IgG, (H+L), Target/Specificity Peroxidase conjugated at 1/10000 dilution. This DDIT4 antibody is generated from Predicted band size : 25 kDa rabbits immunized with a KLH conjugated Blocking/Dilution buffer: 5% NFDM/TBST. synthetic peptide between 20-49 amino acids from the N-terminal region of human DDIT4. Dilution WB~~1:2000 IHC-P~~1:25 Format Purified polyclonal antibody supplied in PBS with 0.09% (W/V) sodium azide. This antibody is purified through a protein A column, followed by peptide affinity purification. Storage Maintain refrigerated at 2-8°C for up to 2 weeks. For long term storage store at -20°C in small aliquots to prevent freeze-thaw cycles. AP6268a staining DDIT4 in human kidney tissue sections by Immunohistochemistry Precautions (IHC-P - paraformaldehyde-fixed, DDIT4 Antibody (N-term) is for research use paraffin-embedded sections). Tissue was Page 1/3 10320 Camino Santa Fe, Suite G San Diego, CA 92121 Tel: 858.875.1900 Fax: 858.622.0609 only and not for use in diagnostic or fixed with formaldehyde and blocked with 3% therapeutic procedures.
    [Show full text]
  • Downloaded from the UCSC PRO-Seq Library Preparation and Sequencing
    ARTICLE https://doi.org/10.1038/s41467-021-21687-2 OPEN Multi-omics analysis reveals contextual tumor suppressive and oncogenic gene modules within the acute hypoxic response ✉ ✉ Zdenek Andrysik1,2, Heather Bender1,2, Matthew D. Galbraith 1,2 & Joaquin M. Espinosa 1,2,3 Cellular adaptation to hypoxia is a hallmark of cancer, but the relative contribution of hypoxia- inducible factors (HIFs) versus other oxygen sensors to tumorigenesis is unclear. We employ 1234567890():,; a multi-omics pipeline including measurements of nascent RNA to characterize transcrip- tional changes upon acute hypoxia. We identify an immediate early transcriptional response that is strongly dependent on HIF1A and the kinase activity of its cofactor CDK8, includes indirect repression of MYC targets, and is highly conserved across cancer types. HIF1A drives this acute response via conserved high-occupancy enhancers. Genetic screen data indicates that, in normoxia, HIF1A displays strong cell-autonomous tumor suppressive effects through a gene module mediating mTOR inhibition. Conversely, in advanced malignancies, expression of a module of HIF1A targets involved in collagen remodeling is associated with poor prog- nosis across diverse cancer types. In this work, we provide a valuable resource for investi- gating context-dependent roles of HIF1A and its targets in cancer biology. 1 Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA. 2 Linda Crnic Institute for Down Syndrome, School of Medicine, University
    [Show full text]
  • Human 4E-BP1 / EIF4EBP1 Protein (His Tag)
    Human 4E-BP1 / EIF4EBP1 Protein (His Tag) Catalog Number: 10022-H07E General Information SDS-PAGE: Gene Name Synonym: 4E-BP1; 4EBP1; BP-1; PHAS-I Protein Construction: A DNA sequence encoding the human 4EBP1 (NP_004086.1) (Ser 2-Ile 118) with a N-terminal polyhistidine tag was expressed. Source: Human Expression Host: E. coli QC Testing Purity: > 90 % as determined by SDS-PAGE Endotoxin: Protein Description Please contact us for more information. The translational suppressor eIF4E binding protein-1, 4E-BP1 functions as Stability: a key regulator in cellular growth, differentiation, apoptosis and survival. The Eif4ebp1 gene, encoding 4E-BP1, is a direct target of a transcription Samples are stable for up to twelve months from date of receipt at -70 ℃ factor activating transcription factor-4 (ATF4), a master regulator of gene expression in stress responses. 4E-BP1 is characterized by its capacity to Predicted N terminal: Met bind specifically to eIF4E and inhibit its interaction with eIF4G. Molecular Mass: Phosphorylation of 4E-BP1 regulates eIF4E availability, and therefore, cap- dependent translation, in cell stress. Binding of eIF4E to eIF4G is inhibited The secreted recombinant human 4EBP1 comprises 124 amino acids with in a competitive manner by 4E-BP1. Phosphorylation of 4E-BP1 decreases a predicted molecular mass of 13.4 kDa. It migrates as an approximately the affinity of this protein for eIF4E, thus favouring the binding of eIF4G 19 kDa band in SDS-PAGE under reducing conditions. and enhancing translation. 4E-BP1 is important for beta-cell survival under endoplasmic reticulum (ER) stress. 4E-BP1 mediates the regulation of Formulation: protein translation by hormones, growth factors and other stimuli that signal through the MAP kinase and mTORC1 pathways.
    [Show full text]
  • Relevance of Translation Initiation in Diffuse Glioma Biology and Its
    cells Review Relevance of Translation Initiation in Diffuse Glioma Biology and its Therapeutic Potential Digregorio Marina 1, Lombard Arnaud 1,2, Lumapat Paul Noel 1, Scholtes Felix 1,2, Rogister Bernard 1,3 and Coppieters Natacha 1,* 1 Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences Research Centre, University of Liège, 4000 Liège, Belgium; [email protected] (D.M.); [email protected] (L.A.); [email protected] (L.P.N.); [email protected] (S.F.); [email protected] (R.B.) 2 Department of Neurosurgery, CHU of Liège, 4000 Liège, Belgium 3 Department of Neurology, CHU of Liège, 4000 Liège, Belgium * Correspondence: [email protected] Received: 18 October 2019; Accepted: 26 November 2019; Published: 29 November 2019 Abstract: Cancer cells are continually exposed to environmental stressors forcing them to adapt their protein production to survive. The translational machinery can be recruited by malignant cells to synthesize proteins required to promote their survival, even in times of high physiological and pathological stress. This phenomenon has been described in several cancers including in gliomas. Abnormal regulation of translation has encouraged the development of new therapeutics targeting the protein synthesis pathway. This approach could be meaningful for glioma given the fact that the median survival following diagnosis of the highest grade of glioma remains short despite current therapy. The identification of new targets for the development of novel therapeutics is therefore needed in order to improve this devastating overall survival rate. This review discusses current literature on translation in gliomas with a focus on the initiation step covering both the cap-dependent and cap-independent modes of initiation.
    [Show full text]
  • DATASHEET Eukaryotic Translation Initiation Factor 4E-Binding
    DATASHEET Abbexa Ltd, Innovation Centre, Cambridge Science Park, Cambridge, CB4 0EY, UK Telephone: +44 (0) 1223 755950 - Fax: +44 (0) 1223 755951 - E-Mail: [email protected] Eukaryotic Translation Initiation Factor 4E-Binding Protein 1 (4EBP1) Antibody Catalogue No.:abx015717 Western blot analysis using 4E-BP1 antibody against truncated 4E-BP1 recombinant protein (1) and A431 cell lysate (2). Immunohistochemical analysis of paraffin-embedded human pancreas carcinoma (A), esophagus carcinoma tissue (B) and ovary tumor tissue (C), showing cytoplasmic and membrane localization using 4E-BP1 antibody with DAB staining. 4E-BP1 (eukaryotic translation Initiation Factor 4E Binding Protein 1),also called ELF4EBP1/BP-1/PHAS-I,which is located on chromosome 8p12, with 118-amino acid protein (about 13kDa).Binding of eIF4EBP1 to eIF4E is reversible and is dependent on the phosphorylation status of eIF4EBP1.Non phosphorylated eIF4EBP1 will bind strongly to eIF4E while (24kDa), the phosphorylated form will not.Akt, TOR, MAP kinase, S6 kinase, and Cdc2 are known kinases capable of inactivating eIF4EBP1 binding to eIF4E by phosphorylating either threonines 35, 45, 69 or serine 64.Although, not all phosphorylation events equally block the eIF4EBP1-eIF4E interaction. Target: 4EBP1 Reactivity: Human Host: Mouse Clonality: Monoclonal Tested Applications: ELISA, WB, IHC Recommended dilutions: ELISA: 1/10000, WB: 1/500 - 1/2000, IHC: 1/200 - 1/1000. Optimal dilutions/concentrations should be determined by the end user. Immunogen: Purified recombinant fragment of 4E-BP1 expressed in E. coli. Purification: Unpurified Ascites. Registered in England: 8475531 - VAT Reg No: GB 167 9240 79 Page 1 DATASHEET Abbexa Ltd, Innovation Centre, Cambridge Science Park, Cambridge, CB4 0EY, UK Telephone: +44 (0) 1223 755950 - Fax: +44 (0) 1223 755951 - E-Mail: [email protected] Isotype: IgG1 Conjugation: Unconjugated Storage: Aliquot and store at -20 °C.
    [Show full text]