CLIP2 As Radiation Biomarker in Papillary Thyroid Carcinoma

Total Page:16

File Type:pdf, Size:1020Kb

CLIP2 As Radiation Biomarker in Papillary Thyroid Carcinoma Oncogene (2015) 34, 3917–3925 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc ORIGINAL ARTICLE CLIP2 as radiation biomarker in papillary thyroid carcinoma M Selmansberger1, A Feuchtinger2, L Zurnadzhy3, A Michna1, JC Kaiser4, M Abend5, A Brenner6, T Bogdanova3, A Walch2, K Unger1,7, H Zitzelsberger1,7 and J Hess1,7 A substantial increase in papillary thyroid carcinoma (PTC) among children exposed to the radioiodine fallout has been one of the main consequences of the Chernobyl reactor accident. Recently, the investigation of PTCs from a cohort of young patients exposed to the post-Chernobyl radioiodine fallout at very young age and a matched nonexposed control group revealed a radiation-specific DNA copy number gain on chromosomal band 7q11.23 and the radiation-associated mRNA overexpression of CLIP2. In this study, we investigated the potential role of CLIP2 as a radiation marker to be used for the individual classification of PTCs into CLIP2- positive and -negative cases—a prerequisite for the integration of CLIP2 into epidemiological modelling of the risk of radiation- induced PTC. We were able to validate the radiation-associated CLIP2 overexpression at the protein level by immunohistochemistry (IHC) followed by relative quantification using digital image analysis software (P = 0.0149). Furthermore, we developed a standardized workflow for the determination of CLIP2-positive and -negative cases that combines visual CLIP2 IHC scoring and CLIP2 genomic copy number status. In addition to the discovery cohort (n = 33), two independent validation cohorts of PTCs (n = 115) were investigated. High sensitivity and specificity rates for all three investigated cohorts were obtained, demonstrating robustness of the developed workflow. To analyse the function of CLIP2 in radiation-associated PTC, the CLIP2 gene regulatory network was reconstructed using global mRNA expression data from PTC patient samples. The genes comprising the first neighbourhood of CLIP2 (BAG2, CHST3, KIF3C, NEURL1, PPIL3 and RGS4) suggest the involvement of CLIP2 in the fundamental carcinogenic processes including apoptosis, mitogen-activated protein kinase signalling and genomic instability. In our study, we successfully developed and independently validated a workflow for the typing of PTC clinical samples into CLIP2-positive and CLIP2-negative and provided first insights into the CLIP2 interactome in the context of radiation-associated PTC. Oncogene (2015) 34, 3917–3925; doi:10.1038/onc.2014.311; published online 6 October 2014 INTRODUCTION alterations in PTC are point mutations of the BRAF gene (V600E) One of the major consequences of the Chernobyl nuclear accident and various variants of RET gene rearrangements, all of which lead in 1986 has been a significant increase in the incidence of to a constitutive activation of the mitogen-activated protein 8 papillary thyroid carcinomas (PTCs) among children exposed to kinase (MAPK) pathway. The frequency of RET/PTC rearrange- the radioiodine fallout, particularly to iodine-131.1 To date, PTC ments (that is, RET/PTC1 and RET/PTC3) was associated with 4 radiation exposure levels in a study of the atomic bomb survivors has developed in 4000 individuals who were children or 9–11 adolescents at the time of exposure.2 Thus, young age at exposure and some but not all studies of post-Chernobyl PTC. However, is a significant risk factor for the development of radiation- RET/PTC3 rearrangements have also been observed with similar frequencies in sporadic PTCs from young patients, indicating a induced PTC.3 In order to delineate radiation-associated effects, it relation with young age of PTC onset.11,12 A more recent is crucial that the tumour cohorts of exposed and nonexposed 13 4 publication by Ricarte-Filho et al. reported a higher frequency cases are matched on age and other factors as closely as possible. of fusion oncogenes in radiation-induced PTCs, including rare TRK This approach was enabled by the Chernobyl Tissue Bank and BRAF rearrangements and the recently discovered ETV6– (CTB, www.chernobyltissuebank.com) that systematically collects NTRK3 kinase fusion oncogene. These promising findings require thyroid tumour tissue samples from residents who lived in the further validation in independent cohorts. We recently reported a contaminated regions of Ukraine and the Russian Federation at radiation-specific DNA copy number gain on the chromosomal the time of the accident. The CTB collection also includes a band 7q11.23 and a radiation-associated mRNA overexpression of substantial number of thyroid tumours from nonexposed patients. the gene CLIP2, located on chromosome 7q11.23.4 Based on these Several studies evidence that radiation exposure can induce copy findings, we aimed to investigate CLIP2 as a radiation biomarker in number alterations and deregulation of gene expressions with the PTC. However, one of the crucial requirements for a biomarker to potential of triggering carcinogenic processes, both of which was be used in epidemiological studies and risk modelling is its validity observed even at low doses of radiation.5–7 Common genetic in terms of sensitivity, specificity, reproducibility and biological 1Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany; 2Research Unit Analytical Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany; 3Institute of Endocrinology and Metabolism, National Academy of Medical Sciences of the Ukraine, Kiev, Ukraine; 4Institute of Radiation Protection, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany; 5Bundeswehr Institute of Radiobiology, Munich, Germany and 6Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, MD, USA. Correspondence: Dr J Hess, Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, Neuherberg 85764, Germany. E-mail: [email protected] 7Clinical Cooperation Group ‘Personalized Radiotherapy of Head and Neck Cancer’, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany. Received 21 March 2014; revised 16 July 2014; accepted 9 August 2014; published online 6 October 2014 CLIP2 as radiation marker in PTC M Selmansberger et al 3918 plausibility. A further aspect for the integration into molecular sections were scanned and the generated images imported into epidemiology is the suitability of both a highly specific marker and the image viewer software Panoramic Viewer (3DHISTECH, an appropriate assay for its detection.14 In addition, a biomarker Budapest, Hungary). The staining intensities were visually scored may provide insights into the mechanisms of the disease it is a as follows: negative staining (score 0), weak staining (score 1), surrogate for. Therefore, this study also aimed to test whether a intermediate staining (score 2) and strong staining (score 3), as CLIP2 radiation-specific mRNA overexpression persists at the demonstrated in Figure 2. Detailed scoring criteria are outlined in protein level and to establish a reproducible workflow that allows the Materials and methods section. The entire classification a classification of PTCs from patients with unknown radiation workflow is illustrated in Figure 3. The resulting visual scores for history. Moreover, we intended to reconstruct the CLIP2 the 33 cases of the Genrisk-T cohort are shown in Table 1. A interactome from global transcriptome data that were derived statistically significant difference (two-sided Fisher’s exact test, from radiation-induced PTCs in order to clarify the role of CLIP2 in P-value = 0.005) between the exposed and nonexposed group was radiation-associated PTC carcinogenesis.15 also revealed using the visual scoring approach. Correlation analysis of the data from both evaluation approaches showed a strong correlation between digital analysis and visual scoring RESULTS − (Spearman’s correlation coefficients: 0.83, P-value: 1.63 × 10 6, CLIP2 protein expression is elevated in PTCs from patients Figure 4). In order to obtain an individual biomarker classification exposed to radioiodine fallout compared with a nonexposed for each case, cases with a visual score of 0 or 1 were considered control group as CLIP2 biomarker negative, whereas cases with visual score 3 In order to evaluate the expression of CLIP2 at the protein level were classified as CLIP2 biomarker positive. For a classification of in exposed and nonexposed cases of the Genrisk-T cohort, an intermediate CLIP2 staining (score 2), the genomic copy immunohistochemical staining with an antibody against CLIP2 number status (gained or not gained) of chromosomal band was performed in a highly standardized manner, followed by 7q11.23 (localization of CLIP2) was taken into account. Preferably, digital image analysis using the Definiens software (Definiens AG, data from array comparative genomic hybridization (array CGH) Munich, Germany). The obtained average marker staining were used. However, if array CGH data were not available, intensities within the analysed tumour regions (regions of interest) interphase fluorescence in situ hybridization analysis with a probe are listed in Supplementary Table 1. Statistical testing revealed specific for 7q11.23 was performed. Cases with a visual score of 2 significantly increased staining intensities (Mann–Whitney
Recommended publications
  • 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]
  • A Cell Line P53 Mutation Type UM
    A Cell line p53 mutation Type UM-SCC 1 wt UM-SCC5 Exon 5, 157 GTC --> TTC Missense mutation by transversion (Valine --> Phenylalanine UM-SCC6 wt UM-SCC9 wt UM-SCC11A wt UM-SCC11B Exon 7, 242 TGC --> TCC Missense mutation by transversion (Cysteine --> Serine) UM-SCC22A Exon 6, 220 TAT --> TGT Missense mutation by transition (Tyrosine --> Cysteine) UM-SCC22B Exon 6, 220 TAT --> TGT Missense mutation by transition (Tyrosine --> Cysteine) UM-SCC38 Exon 5, 132 AAG --> AAT Missense mutation by transversion (Lysine --> Asparagine) UM-SCC46 Exon 8, 278 CCT --> CGT Missense mutation by transversion (Proline --> Alanine) B 1 Supplementary Methods Cell Lines and Cell Culture A panel of ten established HNSCC cell lines from the University of Michigan series (UM-SCC) was obtained from Dr. T. E. Carey at the University of Michigan, Ann Arbor, MI. The UM-SCC cell lines were derived from eight patients with SCC of the upper aerodigestive tract (supplemental Table 1). Patient age at tumor diagnosis ranged from 37 to 72 years. The cell lines selected were obtained from patients with stage I-IV tumors, distributed among oral, pharyngeal and laryngeal sites. All the patients had aggressive disease, with early recurrence and death within two years of therapy. Cell lines established from single isolates of a patient specimen are designated by a numeric designation, and where isolates from two time points or anatomical sites were obtained, the designation includes an alphabetical suffix (i.e., "A" or "B"). The cell lines were maintained in Eagle's minimal essential media supplemented with 10% fetal bovine serum and penicillin/streptomycin.
    [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]
  • 1 Supporting Information for a Microrna Network Regulates
    Supporting Information for A microRNA Network Regulates Expression and Biosynthesis of CFTR and CFTR-ΔF508 Shyam Ramachandrana,b, Philip H. Karpc, Peng Jiangc, Lynda S. Ostedgaardc, Amy E. Walza, John T. Fishere, Shaf Keshavjeeh, Kim A. Lennoxi, Ashley M. Jacobii, Scott D. Rosei, Mark A. Behlkei, Michael J. Welshb,c,d,g, Yi Xingb,c,f, Paul B. McCray Jr.a,b,c Author Affiliations: Department of Pediatricsa, Interdisciplinary Program in Geneticsb, Departments of Internal Medicinec, Molecular Physiology and Biophysicsd, Anatomy and Cell Biologye, Biomedical Engineeringf, Howard Hughes Medical Instituteg, Carver College of Medicine, University of Iowa, Iowa City, IA-52242 Division of Thoracic Surgeryh, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada-M5G 2C4 Integrated DNA Technologiesi, Coralville, IA-52241 To whom correspondence should be addressed: Email: [email protected] (M.J.W.); yi- [email protected] (Y.X.); Email: [email protected] (P.B.M.) This PDF file includes: Materials and Methods References Fig. S1. miR-138 regulates SIN3A in a dose-dependent and site-specific manner. Fig. S2. miR-138 regulates endogenous SIN3A protein expression. Fig. S3. miR-138 regulates endogenous CFTR protein expression in Calu-3 cells. Fig. S4. miR-138 regulates endogenous CFTR protein expression in primary human airway epithelia. Fig. S5. miR-138 regulates CFTR expression in HeLa cells. Fig. S6. miR-138 regulates CFTR expression in HEK293T cells. Fig. S7. HeLa cells exhibit CFTR channel activity. Fig. S8. miR-138 improves CFTR processing. Fig. S9. miR-138 improves CFTR-ΔF508 processing. Fig. S10. SIN3A inhibition yields partial rescue of Cl- transport in CF epithelia.
    [Show full text]
  • Supplemental Information
    Supplemental information Dissection of the genomic structure of the miR-183/96/182 gene. Previously, we showed that the miR-183/96/182 cluster is an intergenic miRNA cluster, located in a ~60-kb interval between the genes encoding nuclear respiratory factor-1 (Nrf1) and ubiquitin-conjugating enzyme E2H (Ube2h) on mouse chr6qA3.3 (1). To start to uncover the genomic structure of the miR- 183/96/182 gene, we first studied genomic features around miR-183/96/182 in the UCSC genome browser (http://genome.UCSC.edu/), and identified two CpG islands 3.4-6.5 kb 5’ of pre-miR-183, the most 5’ miRNA of the cluster (Fig. 1A; Fig. S1 and Seq. S1). A cDNA clone, AK044220, located at 3.2-4.6 kb 5’ to pre-miR-183, encompasses the second CpG island (Fig. 1A; Fig. S1). We hypothesized that this cDNA clone was derived from 5’ exon(s) of the primary transcript of the miR-183/96/182 gene, as CpG islands are often associated with promoters (2). Supporting this hypothesis, multiple expressed sequences detected by gene-trap clones, including clone D016D06 (3, 4), were co-localized with the cDNA clone AK044220 (Fig. 1A; Fig. S1). Clone D016D06, deposited by the German GeneTrap Consortium (GGTC) (http://tikus.gsf.de) (3, 4), was derived from insertion of a retroviral construct, rFlpROSAβgeo in 129S2 ES cells (Fig. 1A and C). The rFlpROSAβgeo construct carries a promoterless reporter gene, the β−geo cassette - an in-frame fusion of the β-galactosidase and neomycin resistance (Neor) gene (5), with a splicing acceptor (SA) immediately upstream, and a polyA signal downstream of the β−geo cassette (Fig.
    [Show full text]
  • A Mode of Cell Adhesion and Migration Facilitated by CD44-Dependent Microtentacles
    A mode of cell adhesion and migration facilitated by CD44-dependent microtentacles Kayla J. Wolfa,b, Poojan Shuklab, Kelsey Springerb, Stacey Leea,b, Jason D. Coombesb,c, Caleb J. Choyd, Samuel J. Kennye,KeXue,f, and Sanjay Kumara,b,g,1 aUniversity of California, Berkeley–University of California San Francisco Graduate Program in Bioengineering, Department of Bioengineering, University of California, Berkeley, CA 94720; bDepartment of Bioengineering, University of California, Berkeley, CA, 94720; cInflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom, SE5 9NU; dDepartment of Molecular and Cell Biology, University of California, Berkeley, CA 94720; eDepartment of Chemistry, University of California, Berkeley, CA 94720; fDivision of Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and gDepartment of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720 Edited by David A. Weitz, Harvard University, Cambridge, MA, and approved March 25, 2020 (received for review August 19, 2019) The structure and mechanics of many connective tissues are grafted C6 gliomas (14). Furthermore, knockdown (KD) of dictated by a collagen-rich extracellular matrix (ECM), where col- CD44 in human GBM tumors slows tumor growth and sensitizes lagen fibers provide topological cues that direct cell migration. tumors to cytotoxic agents (15). CD44 is also a marker of glioma However, comparatively little is known about how cells navigate stem cells (GSCs) (also known as tumor-initiating cells) and the hyaluronic acid (HA)-rich, nanoporous ECM of the brain, a contributes to maintaining stemness (16). Our laboratory has problem with fundamental implications for development, inflam- shown that CD44 is necessary for adhesion and migration on mation, and tumor invasion.
    [Show full text]
  • The Contribution of CLIP2 Haploinsufficiency to the Clinical
    REPORT The Contribution of CLIP2 Haploinsufficiency to the Clinical Manifestations of the Williams-Beuren Syndrome Geert Vandeweyer,1 Nathalie Van der Aa,1 Edwin Reyniers,1 and R. Frank Kooy1,* Williams-Beuren syndrome is a rare contiguous gene syndrome, characterized by intellectual disability, facial dysmorphisms, connec- tive-tissue abnormalities, cardiac defects, structural brain abnormalities, and transient infantile hypercalcemia. Genes lying telomeric to RFC2, including CLIP2, GTF2I and GTF2IRD1, are currently thought to be the most likely major contributors to the typical Williams syndrome cognitive profile, characterized by a better-than-expected auditory rote-memory ability, a relative sparing of language capa- bilities, and a severe visual-spatial constructive impairment. Atypical deletions in the region have helped to establish genotype-pheno- type correlations. So far, however, hardly any deletions affecting only a single gene in the disease region have been described. We present here two healthy siblings with a pure, hemizygous deletion of CLIP2. A putative role in the cognitive and behavioral abnormalities seen in Williams-Beuren patients has been suggested for this gene on the basis of observations in a knock-out mouse model. The presented siblings did not show any of the clinical features associated with the syndrome. Cognitive testing showed an average IQ for both and no indication of the Williams syndrome cognitive profile. This shows that CLIP2 haploinsufficiency by itself does not lead to the physical or cognitive characteristics of the Williams-Beuren syndrome, nor does it lead to the Williams syndrome cognitive profile. Although contri- bution of CLIP2 to the phenotype cannot be excluded when it is deleted in combination with other genes, our results support the hypothesis that GTF2IRD1 and GTF2I are the main genes causing the cognitive defects associated with Williams-Beuren syndrome.
    [Show full text]
  • In This Table Protein Name, Uniprot Code, Gene Name P-Value
    Supplementary Table S1: In this table protein name, uniprot code, gene name p-value and Fold change (FC) for each comparison are shown, for 299 of the 301 significantly regulated proteins found in both comparisons (p-value<0.01, fold change (FC) >+/-0.37) ALS versus control and FTLD-U versus control. Two uncharacterized proteins have been excluded from this list Protein name Uniprot Gene name p value FC FTLD-U p value FC ALS FTLD-U ALS Cytochrome b-c1 complex P14927 UQCRB 1.534E-03 -1.591E+00 6.005E-04 -1.639E+00 subunit 7 NADH dehydrogenase O95182 NDUFA7 4.127E-04 -9.471E-01 3.467E-05 -1.643E+00 [ubiquinone] 1 alpha subcomplex subunit 7 NADH dehydrogenase O43678 NDUFA2 3.230E-04 -9.145E-01 2.113E-04 -1.450E+00 [ubiquinone] 1 alpha subcomplex subunit 2 NADH dehydrogenase O43920 NDUFS5 1.769E-04 -8.829E-01 3.235E-05 -1.007E+00 [ubiquinone] iron-sulfur protein 5 ARF GTPase-activating A0A0C4DGN6 GIT1 1.306E-03 -8.810E-01 1.115E-03 -7.228E-01 protein GIT1 Methylglutaconyl-CoA Q13825 AUH 6.097E-04 -7.666E-01 5.619E-06 -1.178E+00 hydratase, mitochondrial ADP/ATP translocase 1 P12235 SLC25A4 6.068E-03 -6.095E-01 3.595E-04 -1.011E+00 MIC J3QTA6 CHCHD6 1.090E-04 -5.913E-01 2.124E-03 -5.948E-01 MIC J3QTA6 CHCHD6 1.090E-04 -5.913E-01 2.124E-03 -5.948E-01 Protein kinase C and casein Q9BY11 PACSIN1 3.837E-03 -5.863E-01 3.680E-06 -1.824E+00 kinase substrate in neurons protein 1 Tubulin polymerization- O94811 TPPP 6.466E-03 -5.755E-01 6.943E-06 -1.169E+00 promoting protein MIC C9JRZ6 CHCHD3 2.912E-02 -6.187E-01 2.195E-03 -9.781E-01 Mitochondrial 2-
    [Show full text]
  • Bag6 Complex Contains a Minimal Tail-Anchor–Targeting Module and a Mock BAG Domain
    Bag6 complex contains a minimal tail-anchor–targeting module and a mock BAG domain Jee-Young Mocka, Justin William Chartrona,Ma’ayan Zaslavera,YueXub,YihongYeb, and William Melvon Clemons Jr.a,1 aDivision of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125; and bLaboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 Edited by Gregory A. Petsko, Weill Cornell Medical College, New York, NY, and approved December 1, 2014 (received for review February 12, 2014) BCL2-associated athanogene cochaperone 6 (Bag6) plays a central analogous yeast complex contains two proteins, Get4 and Get5/ role in cellular homeostasis in a diverse array of processes and is Mdy2, which are homologs of the mammalian proteins TRC35 part of the heterotrimeric Bag6 complex, which also includes and Ubl4A, respectively. In yeast, these two proteins form ubiquitin-like 4A (Ubl4A) and transmembrane domain recognition a heterotetramer that regulates the handoff of the TA protein complex 35 (TRC35). This complex recently has been shown to be from the cochaperone small, glutamine-rich, tetratricopeptide important in the TRC pathway, the mislocalized protein degrada- repeat protein 2 (Sgt2) [small glutamine-rich tetratricopeptide tion pathway, and the endoplasmic reticulum-associated degrada- repeat-containing protein (SGTA) in mammals] to the delivery tion pathway. Here we define the architecture of the Bag6 factor Get3 (TRC40 in mammals) (19–22). It is expected that the complex, demonstrating that both TRC35 and Ubl4A have distinct mammalian homologs, along with Bag6, play a similar role (23– C-terminal binding sites on Bag6 defining a minimal Bag6 complex.
    [Show full text]
  • Transcriptome Network of the Papillary Thyroid Carcinoma Radiation Marker
    Selmansberger et al. Radiation Oncology (2020) 15:182 https://doi.org/10.1186/s13014-020-01620-5 RESEARCH Open Access Transcriptome network of the papillary thyroid carcinoma radiation marker CLIP2 Martin Selmansberger1, Agata Michna1, Herbert Braselmann1, Ines Höfig2, Kenji Schorpp3, Peter Weber1, Natasa Anastasov2, Horst Zitzelsberger1,4,5, Julia Hess1,4,5 and Kristian Unger1,4,5* Abstract Background: We present a functional gene association network of the CLIP2 gene, generated by de-novo reconstruction from transcriptomic microarray data. CLIP2 was previously identified as a potential marker for radiation induced papillary thyroid carcinoma (PTC) of young patients in the aftermath of the Chernobyl reactor accident. Considering the rising thyroid cancer incidence rates in western societies, potentially related to medical radiation exposure, the functional characterization of CLIP2 is of relevance and contributes to the knowledge about radiation-induced thyroid malignancies. Methods: We generated a transcriptomic mRNA expression data set from a CLIP2-perturbed thyroid cancer cell line (TPC-1) with induced CLIP2 mRNA overexpression and siRNA knockdown, respectively, followed by gene-association network reconstruction using the partial correlation-based approach GeneNet. Furthermore, we investigated different approaches for prioritizing differentially expressed genes for network reconstruction and compared the resulting networks with existing functional interaction networks from the Reactome, Biogrid and STRING databases. The derived CLIP2 interaction partners were validated on transcript and protein level. Results: The best reconstructed network with regard to selection parameters contained a set of 20 genes in the 1st neighborhood of CLIP2 and suggests involvement of CLIP2 in the biological processes DNA repair/maintenance, chromosomal instability, promotion of proliferation and metastasis.
    [Show full text]
  • Senescence Inhibits the Chaperone Response to Thermal Stress
    SUPPLEMENTAL INFORMATION Senescence inhibits the chaperone response to thermal stress Jack Llewellyn1, 2, Venkatesh Mallikarjun1, 2, 3, Ellen Appleton1, 2, Maria Osipova1, 2, Hamish TJ Gilbert1, 2, Stephen M Richardson2, Simon J Hubbard4, 5 and Joe Swift1, 2, 5 (1) Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester, M13 9PT, UK. (2) Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK. (3) Current address: Department of Biomedical Engineering, University of Virginia, Box 800759, Health System, Charlottesville, VA, 22903, USA. (4) Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PL, UK. (5) Correspondence to SJH ([email protected]) or JS ([email protected]). Page 1 of 11 Supplemental Information: Llewellyn et al. Chaperone stress response in senescence CONTENTS Supplemental figures S1 – S5 … … … … … … … … 3 Supplemental table S6 … … … … … … … … 10 Supplemental references … … … … … … … … 11 Page 2 of 11 Supplemental Information: Llewellyn et al. Chaperone stress response in senescence SUPPLEMENTAL FIGURES Figure S1. A EP (passage 3) LP (passage 16) 200 µm 200 µm 1.5 3 B Mass spectrometry proteomics (n = 4) C mRNA (n = 4) D 100k EP 1.0 2 p < 0.0001 p < 0.0001 LP p < 0.0001 p < 0.0001 ) 0.5 1 2 p < 0.0001 p < 0.0001 10k 0.0 0 -0.5 -1 Cell area (µm Cell area fold change vs. EP fold change vs.
    [Show full text]
  • LOXL1 Confers Antiapoptosis and Promotes Gliomagenesis Through Stabilizing BAG2
    Cell Death & Differentiation (2020) 27:3021–3036 https://doi.org/10.1038/s41418-020-0558-4 ARTICLE LOXL1 confers antiapoptosis and promotes gliomagenesis through stabilizing BAG2 1,2 3 4 3 4 3 1 1 Hua Yu ● Jun Ding ● Hongwen Zhu ● Yao Jing ● Hu Zhou ● Hengli Tian ● Ke Tang ● Gang Wang ● Xiongjun Wang1,2 Received: 10 January 2020 / Revised: 30 April 2020 / Accepted: 5 May 2020 / Published online: 18 May 2020 © The Author(s) 2020. This article is published with open access Abstract The lysyl oxidase (LOX) family is closely related to the progression of glioma. To ensure the clinical significance of LOX family in glioma, The Cancer Genome Atlas (TCGA) database was mined and the analysis indicated that higher LOXL1 expression was correlated with more malignant glioma progression. The functions of LOXL1 in promoting glioma cell survival and inhibiting apoptosis were studied by gain- and loss-of-function experiments in cells and animals. LOXL1 was found to exhibit antiapoptotic activity by interacting with multiple antiapoptosis modulators, especially BAG family molecular chaperone regulator 2 (BAG2). LOXL1-D515 interacted with BAG2-K186 through a hydrogen bond, and its lysyl 1234567890();,: 1234567890();,: oxidase activity prevented BAG2 degradation by competing with K186 ubiquitylation. Then, we discovered that LOXL1 expression was specifically upregulated through the VEGFR-Src-CEBPA axis. Clinically, the patients with higher LOXL1 levels in their blood had much more abundant BAG2 protein levels in glioma tissues. Conclusively, LOXL1 functions as an important mediator that increases the antiapoptotic capacity of tumor cells, and approaches targeting LOXL1 represent a potential strategy for treating glioma.
    [Show full text]