Supplementary Data

Total Page:16

File Type:pdf, Size:1020Kb

Supplementary Data Supplementary data Supplementary figure 1. Heatmap of common response, 2 and 6 hours after irradiation. Heatmap of the common response to 4 Gy irradiation in all 32 cell lines, adapted from the BRB-array tools time course plug-in output. Supplementary table 1 gives an overview of cell line characteristics. Supplementary table 2 describes the patient characteristics for the 34 patients in the validation cohort. Supplementary table 3 describes the references used for the definition of miR functions stated in table 1. Supplementary table 4 lists 1226 genes significantly correlated with radioresistance from a BRB time course plug-in analysis. Supplementary table 5 lists the genes in our p53 and DNA repair signatures. Supplementary table 6 lists the HNSCC EMT-scores for the 32 cell lines. Supplementary table 7 lists the 1189 genes in the HNSCC-EMT signature. Supplementary table 8 shows results of testing gene sets for reactive oxygen species, DNA repair, cell cycle phase and several means of cell death against the EMT gene set. Supplementary figure 1. Heatmap of common response, 2 and 6 hours after irradiation. Heatmap of the common response to 4 Gy irradiation in all 32 cell lines, adapted from the BRB-array tools time course plug-in output. Supplementary table 1. Overview of cell line characteristics for the 32 cell lines. Cell line Radiosens- Passage Patient Primary Location T N M Type of Grade Previous itivity (AUC) tested sex Specimen Treatment UT-SCC-1A 1.7 19 F gingiva 2 1 0 rT 2 RT mandibulae UT-SCC-2 1.8 12 M floor of mouth 4 1 0 pT 2 no UT-SCC-4 1.7 9 F supraglottic 3 0 0 rN 2 RT UT-SCC-5 2.3 14 M tongue 1 1 0 ppT 2 RT UT-SCC-6A 2.6 27 F supraglottic 2 1 0 rT 1 RT UT-SCC-7 2 12 M cutis regio 1 0 0 rN 2 RT temporalis UT-SCC-8 1.9 27 M supraglottic 2 0 0 pT 1 no UT-SCC-9 1.4 13 M glottic larynx 2 1 0 N 1 RT UT-SCC-12 2.1 14 F cutis nasi 2 0 0 pT 1 no UT-SCC-15 2.1 15 M tongue 1 0 0 rT 1 RT UT-SCC-16A 1.8 17 F tongue 3 0 0 pT 3 RT UT-SCC-19A 1.7 14 M glottic larynx 4 0 0 pT 2 no UT-SCC-19B 1.7 14 M glottic larynx 4 0 0 ppT 2 RT UT-SCC-20A 2.1 19 F floor of mouth 1 0 0 pT 2 RT UT-SCC-22 1.8 25 M glottic larynx 1 0 0 rT 2 RT UT-SCC-23 1.6 22 M glottic larynx 3 0 0 ppT 1 RT UT-SCC-24A 2.6 24 M tongue 2 0 0 pT 2 no UT-SCC-25 2.2 12 M tongue 2 0 0 pT 1 RT UT-SCC-27 1.9 12 M gingiva 2 0 0 rT 3 RT mandibulae UT-SCC-32 1.7 16 M tongue 3 0 0 ppT 1 RT UT-SCC-36 2.2 8 M floor of mouth 4 1 0 pT 3 no UT-SCC-42A 2.1 7 M supraglottic 4 3 0 pT 3 no UT-SCC-45 2 17 M floor of mouth 3 1 0 pT 3 no UT-SCC-46A 1.6 11 M gingiva maxillae 1 0 0 pT 3 no UT-SCC-47 2 13 M floor of mouth 2 0 0 pT 3 no UT-SCC-48 1.6 15 M parotid gland 3 0 0 pT 2 no UT-SCC-54C 2.3 14 F buccal mucosa 0 0 0 rN 0 RT UT-SCC-60B 2.2 13 M tonsil 4 1 0 ppN 1 RT UT-SCC-76A 2.5 13 M tongue 3 0 0 pT 2 no UT-SCC-77 2.5 23 M tongue 1 0 0 rN 2 no UT-SCC-79A 2.4 14 F parotid gland 2 0 0 rT 2 no UT-SCC-90 2.2 20 M tongue 1 0 0 rT 2 RT Supplementary table 1: Overview of the properties of all 32 cell lines. p= primary tumor, r= recurrent tumor, pp = persistent primary tumor, T=from the primary tumor location, N = from the lymph node. Supplementary table 2: patient characteristics for the 34 patients in the validation cohort. Cures Recurrences N 17 17 Sex Male 59% 59% Female 41% 41% Age (years) Average 68 67 Treatment year Average 2007 2007 T-stage T2 65% 59% T3 35% 41% Subsite Glottic 47% 47% Supraglottic 53% 53% Follow up Average 3.9 3.7 (years) Supplementary table 3: references for the miR functions, as stated in table 1. miR miR function miR-203a Inhibit growth, self-renewal, migration, invasion and EMT (1–4) miR-205-5p Promote apoptosis, inhibit growth, migration, invasion and EMT (5–8) miR-452-5p Reduce stem-like traits and tumorigenesis, EMT (9,10) miR-200b-3p Reduced proliferation, migration, invasion and EMT (5,11–16) miR-429 Inhibit proliferation and EMT (5,15,17,18) miR-141-3p Inhibit EMT (5,19) miR-200a-3p Inhibit EMT (5,15,16) miR-7-5p Inhibit invasion, self renewal and EMT (20,21). Promote apoptosis (22) miR-138-5p Inhibit proliferation, invasion, migration, modify DNA damage response (23,24) miR-34a-5p Inhibit proliferation, invasion, metastasis, stemness, EMT (25,26) miR-142-3p Maintenance of dendritic cells (27) Inhibit growth and stemness (28) miR-33b-5p Reduce proliferation, induce G1 arrest (29), cholesterol transport (30) miR-130a-3p enhances cell proliferation and migration (31), reduce trail resistance and migration (32) miR-193b-3p Proliferation, differentiation, apoptosis (33) miR-151a-5p Increased migration, invasion (34–36), also inhibition of EMT (37) miR-10a-5p Apoptosis (33) miR-424-5p Increased proliferation, migration and invasion, inhibit apoptosis (38,39) miR-15a-5p Sensitize cells to apoptosis (40,41) 1. Chen J, Tran UM, Rajarajacholan U, Thalappilly S, Riabowol K. ING1b-inducible microRNA203 inhibits cell proliferation. British journal of cancer. Nature Publishing Group; 2013;108:1143–8. 2. Abella V, Valladares M, Rodriguez T, Haz M, Blanco M, Tarrío N, et al. miR-203 regulates cell proliferation through its influence on Hakai expression. PloS one. 2012;7:e52568. 3. Moes M, Le Béchec A, Crespo I, Laurini C, Halavatyi A, Vetter G, et al. A novel network integrating a miRNA- 203/SNAI1 feedback loop which regulates epithelial to mesenchymal transition. PloS one. 2012;7:e35440. 4. Zhang Z, Zhang B, Li W, Fu L, Fu L, Zhu Z, et al. Epigenetic Silencing of miR-203 Upregulates SNAI2 and Contributes to the Invasiveness of Malignant Breast Cancer Cells. Genes & cancer. 2011;2:782–91. 5. Gregory P a, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nature cell biology. 2008;10:593–601. 6. Lee J-Y, Park MK, Park J-H, Lee HJ, Shin DH, Kang Y, et al. Loss of the polycomb protein Mel-18 enhances the epithelial-mesenchymal transition by ZEB1 and ZEB2 expression through the downregulation of miR-205 in breast cancer. Oncogene. 2013; 7. Xie H, Zhao Y, Caramuta S, Larsson C, Lui W-O. miR-205 expression promotes cell proliferation and migration of human cervical cancer cells. PloS one. 2012;7:e46990. 8. Majid S, Saini S, Dar AA, Hirata H, Shahryari V, Tanaka Y, et al. MicroRNA-205 inhibits Src-mediated oncogenic pathways in renal cancer. Cancer research. 2011;71:2611–21. 9. Liu L, Chen K, Wu J, Shi L, Hu B, Cheng S, et al. Downregulation of miR-452 Promotes Stem-Like Traits and Tumorigenicity of Gliomas. Clinical cancer research. 2013;3429–38. 10. Sheehy NT, Cordes KR, White MP, Ivey KN, Srivastava D. The neural crest-enriched microRNA miR-452 regulates epithelial-mesenchymal signaling in the first pharyngeal arch. Development. 2010;137:4307–16. 11. Li A, Omura N, Hong S-M, Vincent A, Walter K, Griffith M, et al. Pancreatic cancers epigenetically silence SIP1 and hypomethylate and overexpress miR-200a/200b in association with elevated circulating miR-200a and miR-200b levels. Cancer research. 2010;70:5226–37. 12. Sun L, Yao Y, Liu B, Lin Z, Lin L, Yang M, et al. MiR-200b and miR-15b regulate chemotherapy-induced epithelial- mesenchymal transition in human tongue cancer cells by targeting BMI1. Oncogene. 2012;31:432–45. 13. Kurashige J, Kamohara H, Watanabe M, Hiyoshi Y, Iwatsuki M, Tanaka Y, et al. MicroRNA-200b regulates cell proliferation, invasion, and migration by directly targeting ZEB2 in gastric carcinoma. Annals of surgical oncology. 2012;19 Suppl 3:S656–64. 14. Chen Y, Xiao Y, Ge W, Zhou K, Wen J, Yan W, et al. miR-200b inhibits TGF-β1-induced epithelial-mesenchymal transition and promotes growth of intestinal epithelial cells. Cell death & disease. 2013;4:e541. 15. Bracken CP, Gregory PA, Kolesnikoff N, Bert AG, Wang J, Shannon MF, et al. A double-negative feedback loop between ZEB1-SIP1 and the microRNA-200 family regulates epithelial-mesenchymal transition. Cancer research. 2008;68:7846–54. 16. Enkhbaatar Z, Terashima M, Oktyabri D, Tange S, Ishimura A, Yano S, et al. KDM5B histone demethylase controls epithelial-mesenchymal transition of cancer cells by regulating the expression of the microRNA-200 family. Cell cycle. 2013;12. 17. Chen J, Wang L, Matyunina L V, Hill CG, McDonald JF.
Recommended publications
  • Supplemental Figures
    A B Previously-induced Doxycycline-naïve survival (%) survival (%) Recurrence-free Recurrence-free Days following dox withdrawal Age C D Primary Recurrent ** Par-4 mRNA H2B-mCherry DAPI Primary Recurrent Supplemental Figure 1: Recurrent tumors are derived from primary tumors. A. Kaplan-Meier survival plot showing recurrent tumor-free survival in mice previously induced with doxycycline (n=30) or tumor-free survival in doxycycline-naïve mice (n=10). B. Kaplan-Meier survival plot showing recurrence-free survival following doxycycline withdrawal in a cohort of recipient mice with orthotopic tumors (n=5). C. Representative images (40x magnification) of primary and recurrent orthotopic tumors following injection of H2B-mCherry labeled primary tumor cell line #1 into recipient mice. D. qRT-PCR analysis of Par-4 transcripts from primary (n=5) and recurrent (n=5) orthotopic tumors. Significance determined by Student’s t-test. Error bars denote mean ± SEM. **p<0.01. A B 1 TWIST1 TWIST2 0.5 SNAI2 0 VIM -0.5 ZEB1 ZEB2 -1 SNAI1 PAWR Positively correlated Negatively correlated with Par-4 with Par-4 CDH1 CLDN7 CLDN4 CLDN3 KRT18 NES = -2.07335 q-value = 0.001129 KRT8 TWIST1 TWIST2 SNAI2 VIM ZEB1 ZEB2 SNAI1 PAWR CDH1 CLDN7 CLDN4 CLDN3 KRT18 KRT8 Marcotte, et al. C 1 SNAI1 0.5 TWIST1 TWIST2 0 VIM -0.5 SNAI2 -1 ZEB1 ZEB2 PAWR CDH1 KRT18 KRT8 CLDN7 CLDN3 CLDN4 SNAI1 TWIST1 TWIST2 VIM SNAI2 ZEB1 ZEB2 PAWR CDH1 KRT18 KRT8 CLDN7 CLDN3 CLDN4 TCGA, Cell 2015 Supplemental Figure 2: Par-4 expression is negatively correlated with EMT in human breast cancer. A.
    [Show full text]
  • Novel Tumor Subgroups of Urothelial Carcinoma of the Bladder Defined by Integrated Genomic Analysis
    Published OnlineFirst August 29, 2012; DOI: 10.1158/1078-0432.CCR-12-1807 Clinical Cancer Human Cancer Biology Research Novel Tumor Subgroups of Urothelial Carcinoma of the Bladder Defined by Integrated Genomic Analysis Carolyn D. Hurst, Fiona M. Platt, Claire F. Taylor, and Margaret A. Knowles Abstract Purpose: There is a need for improved subclassification of urothelial carcinoma (UC) at diagnosis. A major aim of this study was to search for novel genomic subgroups. Experimental design: We assessed 160 tumors for genome-wide copy number alterations and mutation in genes implicated in UC. These comprised all tumor grades and stages and included 49 high-grade stage T1 (T1G3) tumors. Results: Our findings point to the existence of genomic subclasses of the "gold-standard" grade/stage groups. The T1G3 tumors separated into 3 major subgroups that differed with respect to the type and number of copy number events and to FGFR3 and TP53 mutation status. We also identified novel regions of copy number alteration, uncovered relationships between molecular events, and elucidated relationships between molecular events and clinico-pathologic features. FGFR3 mutant tumors were more chromosom- ally stable than their wild-type counterparts and a mutually exclusive relationship between FGFR3 mutation and overrepresentation of 8q was observed in non-muscle-invasive tumors. In muscle-invasive (MI) tumors, metastasis was positively associated with losses of regions on 10q (including PTEN), 16q and 22q, and gains on 10p, 11q, 12p, 19p, and 19q. Concomitant copy number alterations positively associated with TP53 mutation in MI tumors were losses on 16p, 2q, 4q, 11p, 10q, 13q, 14q, 16q, and 19p, and gains on 1p, 8q, 10q, and 12q.
    [Show full text]
  • Regulation of Cdc42 and Its Effectors in Epithelial Morphogenesis Franck Pichaud1,2,*, Rhian F
    © 2019. Published by The Company of Biologists Ltd | Journal of Cell Science (2019) 132, jcs217869. doi:10.1242/jcs.217869 REVIEW SUBJECT COLLECTION: ADHESION Regulation of Cdc42 and its effectors in epithelial morphogenesis Franck Pichaud1,2,*, Rhian F. Walther1 and Francisca Nunes de Almeida1 ABSTRACT An overview of Cdc42 Cdc42 – a member of the small Rho GTPase family – regulates cell Cdc42 was discovered in yeast and belongs to a large family of small – polarity across organisms from yeast to humans. It is an essential (20 30 kDa) GTP-binding proteins (Adams et al., 1990; Johnson regulator of polarized morphogenesis in epithelial cells, through and Pringle, 1990). It is part of the Ras-homologous Rho subfamily coordination of apical membrane morphogenesis, lumen formation and of GTPases, of which there are 20 members in humans, including junction maturation. In parallel, work in yeast and Caenorhabditis elegans the RhoA and Rac GTPases, (Hall, 2012). Rho, Rac and Cdc42 has provided important clues as to how this molecular switch can homologues are found in all eukaryotes, except for plants, which do generate and regulate polarity through localized activation or inhibition, not have a clear homologue for Cdc42. Together, the function of and cytoskeleton regulation. Recent studies have revealed how Rho GTPases influences most, if not all, cellular processes. important and complex these regulations can be during epithelial In the early 1990s, seminal work from Alan Hall and his morphogenesis. This complexity is mirrored by the fact that Cdc42 can collaborators identified Rho, Rac and Cdc42 as main regulators of exert its function through many effector proteins.
    [Show full text]
  • ASPH-Notch Axis Guided Exosomal Delivery of Prometastatic Secretome
    Lin et al. Molecular Cancer (2019) 18:156 https://doi.org/10.1186/s12943-019-1077-0 RESEARCH Open Access ASPH-notch Axis guided Exosomal delivery of Prometastatic Secretome renders breast Cancer multi-organ metastasis Qiushi Lin1†, Xuesong Chen2†, Fanzheng Meng3†, Kosuke Ogawa4†, Min Li5, Ruipeng Song3, Shugeng Zhang3, Ziran Zhang3, Xianglu Kong3, Qinggang Xu1,6, Fuliang He1,7, Xuewei Bai8, Bei Sun8, Mien-Chie Hung9,10, Lianxin Liu3,11*, Jack Wands4* and Xiaoqun Dong12,1* Abstract Background: Aspartate β-hydroxylase (ASPH) is silent in normal adult tissues only to re-emerge during oncogenesis where its function is required for generation and maintenance of malignant phenotypes. Exosomes enable prooncogenic secretome delivering and trafficking for long-distance cell-to-cell communication. This study aims to explore molecular mechanisms underlying how ASPH network regulates designated exosomes to program development and progression of breast cancer. Methods: Stable cell lines overexpressing or knocking-out of ASPH were established using lentivirus transfection or CRISPR-CAS9 systems. Western blot, MTT, immunofluorescence, luciferase reporter, co-immunoprecipitation, 2D/3-D invasion, tube formation, mammosphere formation, immunohistochemistry and newly developed in vitro metastasis were applied. Results: Through physical interactions with Notch receptors, ligands (JAGs) and regulators (ADAM10/17), ASPH activates Notch cascade to provide raw materials (especially MMPs/ADAMs) for synthesis/release of pro-metastatic exosomes. Exosomes orchestrate EMT, 2-D/3-D invasion, stemness, angiogenesis, and premetastatic niche formation. Small molecule inhibitors (SMIs) of ASPH’s β-hydroxylase specifically/efficiently abrogated in vitro metastasis, which mimics basement membrane invasion at primary site, intravasation/extravasation (transendothelial migration), and colonization/outgrowth at distant sites.
    [Show full text]
  • Propranolol-Mediated Attenuation of MMP-9 Excretion in Infants with Hemangiomas
    Supplementary Online Content Thaivalappil S, Bauman N, Saieg A, Movius E, Brown KJ, Preciado D. Propranolol-mediated attenuation of MMP-9 excretion in infants with hemangiomas. JAMA Otolaryngol Head Neck Surg. doi:10.1001/jamaoto.2013.4773 eTable. List of All of the Proteins Identified by Proteomics This supplementary material has been provided by the authors to give readers additional information about their work. © 2013 American Medical Association. All rights reserved. Downloaded From: https://jamanetwork.com/ on 10/01/2021 eTable. List of All of the Proteins Identified by Proteomics Protein Name Prop 12 mo/4 Pred 12 mo/4 Δ Prop to Pred mo mo Myeloperoxidase OS=Homo sapiens GN=MPO 26.00 143.00 ‐117.00 Lactotransferrin OS=Homo sapiens GN=LTF 114.00 205.50 ‐91.50 Matrix metalloproteinase‐9 OS=Homo sapiens GN=MMP9 5.00 36.00 ‐31.00 Neutrophil elastase OS=Homo sapiens GN=ELANE 24.00 48.00 ‐24.00 Bleomycin hydrolase OS=Homo sapiens GN=BLMH 3.00 25.00 ‐22.00 CAP7_HUMAN Azurocidin OS=Homo sapiens GN=AZU1 PE=1 SV=3 4.00 26.00 ‐22.00 S10A8_HUMAN Protein S100‐A8 OS=Homo sapiens GN=S100A8 PE=1 14.67 30.50 ‐15.83 SV=1 IL1F9_HUMAN Interleukin‐1 family member 9 OS=Homo sapiens 1.00 15.00 ‐14.00 GN=IL1F9 PE=1 SV=1 MUC5B_HUMAN Mucin‐5B OS=Homo sapiens GN=MUC5B PE=1 SV=3 2.00 14.00 ‐12.00 MUC4_HUMAN Mucin‐4 OS=Homo sapiens GN=MUC4 PE=1 SV=3 1.00 12.00 ‐11.00 HRG_HUMAN Histidine‐rich glycoprotein OS=Homo sapiens GN=HRG 1.00 12.00 ‐11.00 PE=1 SV=1 TKT_HUMAN Transketolase OS=Homo sapiens GN=TKT PE=1 SV=3 17.00 28.00 ‐11.00 CATG_HUMAN Cathepsin G OS=Homo
    [Show full text]
  • A Genome-Wide Association Study Lystra P
    Hayden et al. Respiratory Research (2018) 19:209 https://doi.org/10.1186/s12931-018-0890-0 RESEARCH Open Access Childhood asthma is associated with COPD and known asthma variants in COPDGene: a genome-wide association study Lystra P. Hayden1,2* , Michael H. Cho2,3, Benjamin A. Raby1,2,3, Terri H. Beaty4, Edwin K. Silverman2,3, Craig P. Hersh2,3 and on behalf of the COPDGene Investigators Abstract Background: Childhood asthma is strongly influenced by genetics and is a risk factor for reduced lung function and chronic obstructive pulmonary disease (COPD) in adults. This study investigates self-reported childhood asthma in adult smokers from the COPDGene Study. We hypothesize that childhood asthma is associated with decreased lung function, increased risk for COPD, and that a genome-wide association study (GWAS) will show association with established asthma variants. Methods: We evaluated current and former smokers ages 45–80 of non-Hispanic white (NHW) or African American (AA) race. Childhood asthma was defined by self-report of asthma, diagnosed by a medical professional, with onset at < 16 years or during childhood. Subjects with a history of childhood asthma were compared to those who never had asthma based on lung function, development of COPD, and genetic variation. GWAS was performed in NHW and AA populations, and combined in meta-analysis. Two sets of established asthma SNPs from published literature were examined for association with childhood asthma. Results: Among 10,199 adult smokers, 730 (7%) reported childhood asthma and 7493 (73%) reported no history of asthma. Childhood asthmatics had reduced lung function and increased risk for COPD (OR 3.42, 95% CI 2.81–4.18).
    [Show full text]
  • Table S1 the Four Gene Sets Derived from Gene Expression Profiles of Escs and Differentiated Cells
    Table S1 The four gene sets derived from gene expression profiles of ESCs and differentiated cells Uniform High Uniform Low ES Up ES Down EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol 269261 Rpl12 11354 Abpa 68239 Krt42 15132 Hbb-bh1 67891 Rpl4 11537 Cfd 26380 Esrrb 15126 Hba-x 55949 Eef1b2 11698 Ambn 73703 Dppa2 15111 Hand2 18148 Npm1 11730 Ang3 67374 Jam2 65255 Asb4 67427 Rps20 11731 Ang2 22702 Zfp42 17292 Mesp1 15481 Hspa8 11807 Apoa2 58865 Tdh 19737 Rgs5 100041686 LOC100041686 11814 Apoc3 26388 Ifi202b 225518 Prdm6 11983 Atpif1 11945 Atp4b 11614 Nr0b1 20378 Frzb 19241 Tmsb4x 12007 Azgp1 76815 Calcoco2 12767 Cxcr4 20116 Rps8 12044 Bcl2a1a 219132 D14Ertd668e 103889 Hoxb2 20103 Rps5 12047 Bcl2a1d 381411 Gm1967 17701 Msx1 14694 Gnb2l1 12049 Bcl2l10 20899 Stra8 23796 Aplnr 19941 Rpl26 12096 Bglap1 78625 1700061G19Rik 12627 Cfc1 12070 Ngfrap1 12097 Bglap2 21816 Tgm1 12622 Cer1 19989 Rpl7 12267 C3ar1 67405 Nts 21385 Tbx2 19896 Rpl10a 12279 C9 435337 EG435337 56720 Tdo2 20044 Rps14 12391 Cav3 545913 Zscan4d 16869 Lhx1 19175 Psmb6 12409 Cbr2 244448 Triml1 22253 Unc5c 22627 Ywhae 12477 Ctla4 69134 2200001I15Rik 14174 Fgf3 19951 Rpl32 12523 Cd84 66065 Hsd17b14 16542 Kdr 66152 1110020P15Rik 12524 Cd86 81879 Tcfcp2l1 15122 Hba-a1 66489 Rpl35 12640 Cga 17907 Mylpf 15414 Hoxb6 15519 Hsp90aa1 12642 Ch25h 26424 Nr5a2 210530 Leprel1 66483 Rpl36al 12655 Chi3l3 83560 Tex14 12338 Capn6 27370 Rps26 12796 Camp 17450 Morc1 20671 Sox17 66576 Uqcrh 12869 Cox8b 79455 Pdcl2 20613 Snai1 22154 Tubb5 12959 Cryba4 231821 Centa1 17897
    [Show full text]
  • Mutant IDH, (R)-2-Hydroxyglutarate, and Cancer
    Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press REVIEW What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer Julie-Aurore Losman1 and William G. Kaelin Jr.1,2,3 1Department of Medical Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA; 2Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA Mutations in metabolic enzymes, including isocitrate whether altered cellular metabolism is a cause of cancer dehydrogenase 1 (IDH1) and IDH2, in cancer strongly or merely an adaptive response of cancer cells in the face implicate altered metabolism in tumorigenesis. IDH1 of accelerated cell proliferation is still a topic of some and IDH2 catalyze the interconversion of isocitrate and debate. 2-oxoglutarate (2OG). 2OG is a TCA cycle intermediate The recent identification of cancer-associated muta- and an essential cofactor for many enzymes, including tions in three metabolic enzymes suggests that altered JmjC domain-containing histone demethylases, TET cellular metabolism can indeed be a cause of some 5-methylcytosine hydroxylases, and EglN prolyl-4-hydrox- cancers (Pollard et al. 2003; King et al. 2006; Raimundo ylases. Cancer-associated IDH mutations alter the enzymes et al. 2011). Two of these enzymes, fumarate hydratase such that they reduce 2OG to the structurally similar (FH) and succinate dehydrogenase (SDH), are bone fide metabolite (R)-2-hydroxyglutarate [(R)-2HG]. Here we tumor suppressors, and loss-of-function mutations in FH review what is known about the molecular mechanisms and SDH have been identified in various cancers, in- of transformation by mutant IDH and discuss their im- cluding renal cell carcinomas and paragangliomas.
    [Show full text]
  • FAM158A Polyclonal Antibody Catalog Number:23919-1-AP
    For Research Use Only FAM158A Polyclonal antibody www.ptgcn.com Catalog Number:23919-1-AP Catalog Number: GenBank Accession Number: Recommended Dilutions: Basic Information 23919-1-AP BC002491 WB 1:200-1:1000 Size: GeneID (NCBI): IHC 1:20-1:200 500 μg/ml 51016 IF 1:20-1:200 Source: Full Name: Rabbit family with sequence similarity 158, Isotype: member A IgG Calculated MW: Purification Method: 208 aa, 23 kDa Antigen Affinity purified Observed MW: Immunogen Catalog Number: 23 kDa AG20744 Applications Tested Applications: Positive Controls: IF, IHC, WB,ELISA WB : HEK-293 cells; Species Specificity: IHC : human pancreas tissue; human Note-IHC: suggested antigen retrieval with IF : HEK-293 cells; TE buffer pH 9.0; (*) Alternatively, antigen retrieval may be performed with citrate buffer pH 6.0 UPF0172 protein FAM158A, also known as c14orf122 or CGI112, is a protein that in humans is encoded by the Background Information FAM158A gene located on chromosome 14q11.2. Human FAM158A and its paralogs in other species are part of the uncharacterized protein family UPF0172 family, which is a subset of the JAB1/Mov34/MPN/PAD-1 ubiquitin protease protein family. The MPN superfamily contributes to ubiquitination and de-ubiquitination activity within the cell. The UPF0172 subset no longer has a functional ubiquitination domain and the function is uncharacterized. Fam158a is nearly ubiquitously expressed throughout the human body. The homolog in mice also shows expression throughout the entire body. Several micro-arrays demonstrate the variable expression of Fam158a in response to other factors and in various cancer types. None of this information gives any indication of a specific function but the wide expression of the gene and its high conservation indicate that Fam158a plays an important role in cellular function.
    [Show full text]
  • Dual Proteome-Scale Networks Reveal Cell-Specific Remodeling of the Human Interactome
    bioRxiv preprint doi: https://doi.org/10.1101/2020.01.19.905109; this version posted January 19, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Dual Proteome-scale Networks Reveal Cell-specific Remodeling of the Human Interactome Edward L. Huttlin1*, Raphael J. Bruckner1,3, Jose Navarrete-Perea1, Joe R. Cannon1,4, Kurt Baltier1,5, Fana Gebreab1, Melanie P. Gygi1, Alexandra Thornock1, Gabriela Zarraga1,6, Stanley Tam1,7, John Szpyt1, Alexandra Panov1, Hannah Parzen1,8, Sipei Fu1, Arvene Golbazi1, Eila Maenpaa1, Keegan Stricker1, Sanjukta Guha Thakurta1, Ramin Rad1, Joshua Pan2, David P. Nusinow1, Joao A. Paulo1, Devin K. Schweppe1, Laura Pontano Vaites1, J. Wade Harper1*, Steven P. Gygi1*# 1Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA. 2Broad Institute, Cambridge, MA, 02142, USA. 3Present address: ICCB-Longwood Screening Facility, Harvard Medical School, Boston, MA, 02115, USA. 4Present address: Merck, West Point, PA, 19486, USA. 5Present address: IQ Proteomics, Cambridge, MA, 02139, USA. 6Present address: Vor Biopharma, Cambridge, MA, 02142, USA. 7Present address: Rubius Therapeutics, Cambridge, MA, 02139, USA. 8Present address: RPS North America, South Kingstown, RI, 02879, USA. *Correspondence: [email protected] (E.L.H.), [email protected] (J.W.H.), [email protected] (S.P.G.) #Lead Contact: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/2020.01.19.905109; this version posted January 19, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.
    [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]
  • Supplementary Table 3 Complete List of RNA-Sequencing Analysis of Gene Expression Changed by ≥ Tenfold Between Xenograft and Cells Cultured in 10%O2
    Supplementary Table 3 Complete list of RNA-Sequencing analysis of gene expression changed by ≥ tenfold between xenograft and cells cultured in 10%O2 Expr Log2 Ratio Symbol Entrez Gene Name (culture/xenograft) -7.182 PGM5 phosphoglucomutase 5 -6.883 GPBAR1 G protein-coupled bile acid receptor 1 -6.683 CPVL carboxypeptidase, vitellogenic like -6.398 MTMR9LP myotubularin related protein 9-like, pseudogene -6.131 SCN7A sodium voltage-gated channel alpha subunit 7 -6.115 POPDC2 popeye domain containing 2 -6.014 LGI1 leucine rich glioma inactivated 1 -5.86 SCN1A sodium voltage-gated channel alpha subunit 1 -5.713 C6 complement C6 -5.365 ANGPTL1 angiopoietin like 1 -5.327 TNN tenascin N -5.228 DHRS2 dehydrogenase/reductase 2 leucine rich repeat and fibronectin type III domain -5.115 LRFN2 containing 2 -5.076 FOXO6 forkhead box O6 -5.035 ETNPPL ethanolamine-phosphate phospho-lyase -4.993 MYO15A myosin XVA -4.972 IGF1 insulin like growth factor 1 -4.956 DLG2 discs large MAGUK scaffold protein 2 -4.86 SCML4 sex comb on midleg like 4 (Drosophila) Src homology 2 domain containing transforming -4.816 SHD protein D -4.764 PLP1 proteolipid protein 1 -4.764 TSPAN32 tetraspanin 32 -4.713 N4BP3 NEDD4 binding protein 3 -4.705 MYOC myocilin -4.646 CLEC3B C-type lectin domain family 3 member B -4.646 C7 complement C7 -4.62 TGM2 transglutaminase 2 -4.562 COL9A1 collagen type IX alpha 1 chain -4.55 SOSTDC1 sclerostin domain containing 1 -4.55 OGN osteoglycin -4.505 DAPL1 death associated protein like 1 -4.491 C10orf105 chromosome 10 open reading frame 105 -4.491
    [Show full text]