PC) of Surface Area Measurements in 34 Cortical Regions: PC1 (Panel A) and PC2 (Panel B)

Total Page:16

File Type:pdf, Size:1020Kb

PC) of Surface Area Measurements in 34 Cortical Regions: PC1 (Panel A) and PC2 (Panel B) Supplementary Figures Figure E1. Loadings for the first two leading principal components (PC) of surface area measurements in 34 cortical regions: PC1 (panel A) and PC2 (panel B). Figure E2. Loadings for the first two leading principal components (PC) of thickness measurements in 34 cortical regions: PC1 (panel A) and PC2 (panel B). Figure E3. Manhattan plots for the meta-analyses of GWAS for surface area PC1 (panel A) and PC2 (panel B) scores in CHARGE and UKBB cohorts. Figure E4. Forest plot of the top PC2 SNP rs73313052. Figure E5. Manhattan plots for the meta-analyses of GWAS thickness PC1 (panel A) and PC2 (panel B) scores in CHARGE and UKBB cohorts. Figure E6. DAAM1 co-localizes with mitochondrial marker ATP5A in the primary visual cortical plate. A. Figure E7. Associations between lateral geniculate nucleus (LGN) volume and surface area PC2 scores in 694 SYS major-homozygotes (GG) (panel A) and in the 206 carriers of minor allele of rs73313052 (GA or AA) (panel B). Figure E8. Genetic correlation between schizophrenia vs. surface area PC1 and PC2. Figure E9. Genetic correlation between ADHD vs. surface area PC1 and PC2. Figure E10. Genetic correlation between autism spectrum disease (ASD) vs. surface area PC1 and PC2. Figure E11. Genetic correlation between bipolar disorder (BD) vs. surface area PC1 and PC2. Figure E12. Genetic correlation between major depressive disorder (MDD) vs. surface area PC1 and PC2. Figure E13. Genetic correlation between neuroticism vs. surface area PC1 and PC2. Figure E14. Genetic correlation between extraversion vs. surface area PC1 and PC2. Figure E15. Genetic correlation between neo-agreeableness vs. surface area PC1 and PC2. Figure E16. Genetic correlation between neo-agreeableness vs. surface area PC1 and PC2. Figure E17. Genetic correlation between neo-openness vs. surface area PC1 and PC2. Figure E18. Genetic correlation between global cognitive function vs. surface area PC1 and PC2. Figure E19. Genetic correlation between educational attainment vs. surface area PC1 and PC2. Figure E20. Genetic correlation between anorexia nervosa vs. surface area PC1 and PC2. Figure E1. Loadings for the first two leading principal components (PC) of surface area measurements in 34 cortical regions: PC1 (panel A) and PC2 (panel B). Each cohort estimated the surface area of the 34 cortical regions (left and right hemispheres summed), using FreeSurfer and carried out unrotated principal component analyses to obtain PC loading values for the first two leading PCs, which are indicated by colored lines: 0-indigo (ARIC), 1-light-blue (ASPSFam), 2-green (BIL&GIN), 3-yellow (CHS), 4-red (FHS), 5-purple (LIFE-Adult), 6-grey-blue (RS), 7-olive-green (SYS-Adult), and 8-mint-green (SYS- Adolescent). Black thicker lines indicate the median loading values across all the cohorts. The median loading values were then used to derive the ‘general’ PC score for each individual and later used as the response variable in the meta-GWAS analyses. 0 ARIC 2 BIL&GIN 4 FHS 6 RS 8 SYS cohort 1 ASPSFam 3 CHS 5 LIFE−Adult 7 SPS A 1.00 4 4 4 4 4 231● 4 3● 4 5 4 83 4 28 2 4 4 4 24 760 1 1 4 25● 6 2 ● 3 0 4 3 ● 4 31 3● 4 2 2 16 61 2 2 51 3 3 2● 5 4 031 4 4 3● 73 ● 8 70 ● 1 56 7 4 ● 6 7 4 2 8 1 0 0 4 2 41 3 3● 05 27 2 3● 1 60 8 5 60 4 580 2578 756 7 ● 1 32● 0 1 7 2 ● 15 6 16 2● 01 5 4 5 2 0.75 15 67 76 1 2 7 3 8 87 6 8 5 ● 0 1 2 170 12 1 0 32 8 50 2 506 85 6 6 05● 86 4 281● 2 8● 1 7 30 1 8 743 8 ● 5 5 ● 7 3 3 03 5 3 8 7 87 1 06 7 2 4 8 0 8 6 3● 2 3 8 7 1 1 7● 5 3 8 465 7 4 8 0817 34 ● 3 5 ● ● 0 8 1 6 ● 7 0.50 067 5 3 4 0● 3 80 1 08 0 5 752 6 6 7265 5 PC1 PC1 13 0 6 0 2 6 2 3 ● ● 7 ● 5 8 75 8 25 6034 0 ● 7 7 8 167 68 6 8 0.25 6 5 8 6 0 0 ARIC 2 BIL&GIN 4 FHS 6 RS 8 SYS cohort 1 ASPSFam 3 CHS 5 LIFE−Adult 7 SPS 0.00 0 ARIC 2 BIL&GIN 4 FHS 6 RS 8 SYS frontalpole rostralmiddlefrontal parsorbitalis medialorbitofrontal rostralanteriorcingulate cohortlateralorbitofrontal parstriangularis superiorfrontal caudalanteriorcingulate parsopercularis caudalmiddlefrontal temporalpole insula entorhinal precentral superiortemporal posteriorcingulate postcentral transversetemporal middletemporal paracentral parahippocampal inferiortemporal supramarginal fusiform bankssts isthmuscingulate precuneus superiorparietal lingual inferiorparietal cuneus pericalcarine lateraloccipital 1.0 1 ASPSFam 3 CHS 5 LIFE−Adult 7 SPS 6 B 6 6 07 7 3● 0 85 650 1.00 ● 0.5 1 1 2 3● 60 825 7● 8 4 3 305 71 056 1 ● 4 1 2 5 8● 8 5 6 0 68● 2 82 7352 5 8 5 1 ● 3 ● 0 241 8 5 3 50 8● 7 ● 5 0 2 1 7 ● 6 6 8 ● 8 5 8 ● ● 2 3 26 8● 305 6 2 ● 4 4 0 7● 7 7 ● 6 2 ● 8 8 3 4 4 ● ● 4 4 35 0 7 66 17 742 60 45 6 4 1 7 ● 78 38 4 3 1273 5 0 27 3 802 21 6 ● 2 6 508 1 8 7 75 1 0 6 7 57 7● ● 75 0 4 ● 4 ● 20 4 8 0 4 44 4 2 7 ● ● 3 1 1 06 3 0 4 1 6 5 0 180 ● 6 ● 1 2 4 7 6 13 8 1 63 4 4 ● 6 6 3 78 4 6 28 ● 6582 563 25 5728 42 6 03 0 52● 4 0 2 2● 2 8 1 0 2 7 4 53 3 2 7 5 2 58 7 36 5 4 ● 30 63 74 023 5 4 16 ● 8 705 3 70 4● 8 382 1 1 1 5● 02 42 0 0 18 328 2 74 748 4 35 84 6207 ● 6 87 5 2 1 0 80 1 6 6 37 0.75 3 5 7 1 34 67 2● 0.0 3 1 0 ● 4 2 1 4 70 ● 4 6 6 3 3 65 8 6302 13 13 ● 28 3 8 1 1 560 ● 15 6 8 7 3 6 1 6 ● 1 4 7 5 1 74 1 3 6 23 5 ● 47 ● 5 ● 2 7 6 ● 0 ● 8 ● 7 3 5 0 0 PC2 1 14 1 0 3 7 5 6● ● PC2 35 3 06 3 6 1 815 15 7 1 23 0571 42 1348 536 80 8 4 7 12 ● 1 ● 3 0 56 72 52 2 7 3● 5 5 ● 6 7 023 0 028 8 6 0 4870 3 4 6 658 685 41 48 3 574 0721 6● 1 6 247 1 ● 4 7 1 3 2 82 4 4 5 6 3 0 6 0 72 ● 4 8 8 6 4 ● 71 8 ● 7420 42 0257 41 28 85 1 30 7 2750 740 6 8 8 3● 1306 1 ● 7 0● 3 2 1 5 5 6 052 573 8 −0.506 3 8 41 45 0.50 ● 4 82 48 154 8 1 PC1 8 3 3 1 0.25 −1.0 frontalpole rostralmiddlefrontal parsorbitalis medialorbitofrontal rostralanteriorcingulate lateralorbitofrontal parstriangularis superiorfrontal caudalanteriorcingulate parsopercularis caudalmiddlefrontal temporalpole insula entorhinal precentral superiortemporal posteriorcingulate postcentral transversetemporal middletemporal paracentral parahippocampal inferiortemporal supramarginal fusiform bankssts isthmuscingulate precuneus superiorparietal lingual inferiorparietal cuneus pericalcarine lateraloccipital 0.00 frontalpole rostralmiddlefrontal parsorbitalis medialorbitofrontal rostralanteriorcingulate lateralorbitofrontal parstriangularis superiorfrontal caudalanteriorcingulate parsopercularis caudalmiddlefrontal temporalpole insula entorhinal precentral superiortemporal posteriorcingulate postcentral transversetemporal middletemporal paracentral parahippocampal inferiortemporal supramarginal fusiform bankssts isthmuscingulate precuneus superiorparietal lingual inferiorparietal cuneus pericalcarine lateraloccipital Figure E2. Loadings for the first two leading principal components (PC) of thickness measurements in 34 cortical regions: PC1 (panel A) and PC2 (panel B). Each cohort estimated the thickness of the 34 cortical regions (left and right hemispheres averaged), using FreeSurfer and carried out unrotated principal component analyses to obtain PC loading values for the first two leading PCs, which are indicated by colored lines: 0-indigo (ARIC), 1-light-blue (ASPSFam), 2-green (BIL&GIN), 3-yellow (CHS), 4-red (FHS), 5-purple (LIFE-Adult), 6-grey-blue (RS), 7-olive-green (SYS-Adult), and 8-mint-green (SYS- Adolescent). Black thicker lines indicate the median loading values across all the cohorts. The median loading values were then used to derive the ‘general’ PC score for each individual and later used as the response variable in the meta-GWAS analyses. CRHR1 A MAPT STH KANSL1 PC1 B DAAM1 PC2 Figure E3. Manhattan plots for the meta-analyses of GWAS for surface area PC1 (panel A) and PC2 (panel B) scores in CHARGE and UKBB cohorts. The GWAS meta- analyses were carried out as follows. Using the median PC loadings across the CHARGE consortium cohorts (Figure E1), each cohort derived the ‘general’ PC scores to be used as phenotypes in the genome-wide association tests in order to ensure ‘homogeneity’ in phenotype derivation. All the association tests were adjusted for age, sex and other cohort- specific confounding variables, such as study site and/or family structure. The cohort-specific GWAS results were then examined for quality-control with Easy QC software1, and meta- 2 analyzed with METAL using fixed effects models.
Recommended publications
  • Open Full Page
    Research Article Distinct Genomic Profiles in Hereditary Breast Tumors Identified by Array-Based Comparative Genomic Hybridization Go¨ran Jo¨nsson,1 Tara L. Naylor,5 Johan Vallon-Christersson,1 Johan Staaf,1 Jia Huang,5 M. Renee Ward,5 Joel D. Greshock,5 Lena Luts,4 Ha˚kan Olsson,1 Nazneen Rahman,6 Michael Stratton,6 Markus Ringne´r,3 A˚ke Borg,1,2 and Barbara L. Weber5 1Department of Oncology, University Hospital; 2Lund Strategic Research Center for Stem Cell Biology and Cell Therapy and 3Department of Theoretical Physics, Lund University; and 4Department of Pathology, University Hospital, Lund, Sweden; 5Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania; and 6Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, United Kingdom Abstract of additional predisposing genes, although technical limitations Mutations in BRCA1 and BRCA2 account for a significant and the complexity of BRCA gene regulation and mutation proportion of hereditary breast cancers. Earlier studies have spectrum can probably explain why some disease-causing muta- shown that inherited and sporadic tumors progress along tions are missed (2). BRCA1 and BRCA2 function as classic tumor different somatic genetic pathways and that global gene suppressor genes with frequent loss of the wild-type allele in expression profiles distinguish between these groups. To tumors of mutation carriers. The BRCA1 protein has been determine whether genomic profiles similarly discriminate implicated in a broad range of cellular functions, including repair among BRCA1, BRCA2, and sporadic tumors, we established of double-strand breaks by homologous recombination, cell cycle DNA copy number profiles using comparative genomic checkpoint control, chromatin remodeling, and transcriptional hybridization to BAC-clone microarrays providing <1 Mb regulation.
    [Show full text]
  • Supplementary Data
    SUPPLEMENTARY DATA A cyclin D1-dependent transcriptional program predicts clinical outcome in mantle cell lymphoma Santiago Demajo et al. 1 SUPPLEMENTARY DATA INDEX Supplementary Methods p. 3 Supplementary References p. 8 Supplementary Tables (S1 to S5) p. 9 Supplementary Figures (S1 to S15) p. 17 2 SUPPLEMENTARY METHODS Western blot, immunoprecipitation, and qRT-PCR Western blot (WB) analysis was performed as previously described (1), using cyclin D1 (Santa Cruz Biotechnology, sc-753, RRID:AB_2070433) and tubulin (Sigma-Aldrich, T5168, RRID:AB_477579) antibodies. Co-immunoprecipitation assays were performed as described before (2), using cyclin D1 antibody (Santa Cruz Biotechnology, sc-8396, RRID:AB_627344) or control IgG (Santa Cruz Biotechnology, sc-2025, RRID:AB_737182) followed by protein G- magnetic beads (Invitrogen) incubation and elution with Glycine 100mM pH=2.5. Co-IP experiments were performed within five weeks after cell thawing. Cyclin D1 (Santa Cruz Biotechnology, sc-753), E2F4 (Bethyl, A302-134A, RRID:AB_1720353), FOXM1 (Santa Cruz Biotechnology, sc-502, RRID:AB_631523), and CBP (Santa Cruz Biotechnology, sc-7300, RRID:AB_626817) antibodies were used for WB detection. In figure 1A and supplementary figure S2A, the same blot was probed with cyclin D1 and tubulin antibodies by cutting the membrane. In figure 2H, cyclin D1 and CBP blots correspond to the same membrane while E2F4 and FOXM1 blots correspond to an independent membrane. Image acquisition was performed with ImageQuant LAS 4000 mini (GE Healthcare). Image processing and quantification were performed with Multi Gauge software (Fujifilm). For qRT-PCR analysis, cDNA was generated from 1 µg RNA with qScript cDNA Synthesis kit (Quantabio). qRT–PCR reaction was performed using SYBR green (Roche).
    [Show full text]
  • Monoclonal Antibodies
    NOTE Clinical Pathology Establishment of rat anti-canine DEP domain containing 1B (DEPDC1B) monoclonal antibodies Masaya IGASE1), Yuki MORINAGA1), Masahiro KATO2), Toshihiro TSUKUI2), Yusuke SAKAI3), Masaru OKUDA4) and Takuya MIZUNO1)* 1)Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi-shi, Yamaguchi 753-8511, Japan 2)Nippon Zenyaku Kogyo Co., Ltd., 1-1 Tairanoue, Sasagawa, Asaka-machi, Koriyama, Fukushima 963-0196, Japan 3)Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi-shi, Yamaguchi 753-8511, Japan 4)Laboratory of Veterinary Internal Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi-shi, Yamaguchi 753-8511, Japan ABSTRACT. DEP domain-containing 1B (DEPDC1B) is involved in the regulation of cell de- J. Vet. Med. Sci. adhesion and actin cytoskeleton activity during the G2/M transition of the cell cycle, and its 82(4): 483–487, 2020 overexpression has been proven to be associated with cancer progression in several human cancers. Canine DEPDC1B was identified as a gene that was overexpressed in canine lymphoma doi: 10.1292/jvms.19-0667 tissues in our previous study. However, in dogs, the protein expression of DEPDC1B remains to be determined due to the lack of a specific monoclonal antibody. Here, we developed rat monoclonal antibodies against canine DEPDC1B and characterized their applicability for immunodetection Received: 12 December 2019 assays. Our findings demonstrated that these antibodies are functional and can be important tools Accepted: 20 February 2020 to investigate the precise role of DEPDC1B in canine tumors. Advanced Epub: 6 March 2020 KEY WORDS: antibody, cancer, DEP domain-containing 1B, dog The DEP domain-containing 1B (DEPDC1B) gene was recently identified and localized at human chromosome 5 (5q12.1).
    [Show full text]
  • The Genetic Program of Pancreatic Beta-Cell Replication in Vivo
    Page 1 of 65 Diabetes The genetic program of pancreatic beta-cell replication in vivo Agnes Klochendler1, Inbal Caspi2, Noa Corem1, Maya Moran3, Oriel Friedlich1, Sharona Elgavish4, Yuval Nevo4, Aharon Helman1, Benjamin Glaser5, Amir Eden3, Shalev Itzkovitz2, Yuval Dor1,* 1Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel 2Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel. 3Department of Cell and Developmental Biology, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel 4Info-CORE, Bioinformatics Unit of the I-CORE Computation Center, The Hebrew University and Hadassah, The Institute for Medical Research Israel- Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel 5Endocrinology and Metabolism Service, Department of Internal Medicine, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel *Correspondence: [email protected] Running title: The genetic program of pancreatic β-cell replication 1 Diabetes Publish Ahead of Print, published online March 18, 2016 Diabetes Page 2 of 65 Abstract The molecular program underlying infrequent replication of pancreatic beta- cells remains largely inaccessible. Using transgenic mice expressing GFP in cycling cells we sorted live, replicating beta-cells and determined their transcriptome. Replicating beta-cells upregulate hundreds of proliferation- related genes, along with many novel putative cell cycle components. Strikingly, genes involved in beta-cell functions, namely glucose sensing and insulin secretion were repressed. Further studies using single molecule RNA in situ hybridization revealed that in fact, replicating beta-cells double the amount of RNA for most genes, but this upregulation excludes genes involved in beta-cell function.
    [Show full text]
  • 1 Title the Role of Mitotic Cell-Substrate Adhesion Re
    Manuscript Title The role of mitotic cell-substrate adhesion re-modeling in animal cell division. Authors Christina L. Dix1*, Helen K. Matthews1*, Marina Uroz3, Susannah McLaren1, Lucie Wolf2, Nicholas Heatley1, Zaw Win1, Pedro Almada1, Ricardo Henriques1, Michael Boutros2, Xavier Trepat3,4,5,6 and Buzz Baum1,7 *These authors contributed equally Affiliations 1 MRC - Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom. 2 Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), and Department for Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, 69120 Heidelberg, Germany 3 Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), 08028 Barcelona, Spain 4 Unitat de Biofisica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain 5 Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain 6 Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08028 Barcelona, Spain 7 Institute for the Physics of Living Systems, University College London, London WC1E 6BT, United Kingdom. 1 Contact information of lead contact and corresponding author: [email protected] Summary Animal cells undergo a dramatic series of shape changes as they divide, which depend on re-modeling of cell-substrate adhesions. Here, we show that while focal adhesion complexes are disassembled during mitotic rounding, integrins remain in place. These integrin-rich contacts connect mitotic cells to the underlying substrate throughout mitosis, guide polarized cell migration following mitotic exit, and are functionally important, since adherent cells undergo division failure when removed from the substrate. Further, the ability of cells to re-spread along pre-existing adhesive contacts is essential for division in cells compromised in their ability to construct a RhoGEF- dependent (Ect2) actomyosin ring.
    [Show full text]
  • Eine Dokumentvorlage Für Abschlussarbeiten Und Andere Wissenschaftliche Arbeiten, Insbesondere Bachelorarbeiten, Masterarbeiten
    1 Institute of Animal Science University of Hohenheim Prof. Dr. Ludwig Hölzle Transcriptomic analyses during infectious anemia in pigs DISSERTATION submitted in fulfillment of the requirements for the degree “Doktor der Agrarwissenschaften” (Dr. sc. agr. / PhD in Agricultural Science) to the Faculty of Agricultural Sciences presented by Sarah-Lena Mack born in Stuttgart, Germany 2018 2 Die vorliegende Arbeit wurde am 03.04.2019 von der Fakultät Agrarwissenschaften der Universität Hohenheim als "Dissertation zur Erlangung des Grades eines Doktors der Agrarwissenschaften" angenommen (Datum: vgl. Schreiben des Vorsitzenden des Pro- motionsausschusses an den/die Doktoranden/in). Tag der mündlichen Prüfung: 20.05.2019 Leiter/in der Prüfung: Prof. Dr. Jörn Bennewitz Berichterstatter/in 1. Prüfer/in: Prof. Dr. Ludwig Hölzle Mitberichterstatter/in, 2. Prüfer/in: Prof. Dr. Holger Stefanski ggf. weitere Berichter/in bzw. Prüfer/in: Prof. Dr. Mathias Ritzmann < Hohenheim, 18.12.2018, Sarah-Lena Mack > 3 Für meine Eltern 4 Table of contents Table of contents ......................................................................................................... 4 Table of figures ............................................................................................................ 7 List of tables ................................................................................................................ 8 List of abbreviations ................................................................................................... 9 Overview
    [Show full text]
  • Overexpressed DEPDC1B Contributes to the Progression of Hepatocellular Carcinoma by CDK1
    www.aging-us.com AGING 2021, Vol. 13, No. 16 Research Paper Overexpressed DEPDC1B contributes to the progression of hepatocellular carcinoma by CDK1 Xiao-Wei Dang1,*, Qi Pan2,*, Zhen-Hai Lin2,*, Hao-Hao Wang1, Lu-Hao Li1, Lin Li1, Dong-Qi Shen1, Pei-Ju Wang1 1Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China 2Department of Hepatic Surgery, The Cancer Hospital of Fudan University, Shanghai, China *Equal contribution Correspondence to: Xiao-Wei Dang; email: [email protected] Keywords: DEPDC1B, CDK1, proliferation, migration, hepatocellular carcinoma (HCC) Received: January 23, 2020 Accepted: February 16, 2021 Published: May 25, 2021 Copyright: © 2021 Dang et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Background: Hepatocellular carcinoma (HCC) is the main type of primary liver cancer and shows a heavy burden worldwide. Its recurrence and mortality rate are still uncontrolled by the usage of present treatments. More attention has been focused on exploring specific genes that play important roles in HCC procession, and the function of DEP domain containing 1B (DEPDC1B) in HCC has not been researched. Methods: Immunohistochemical staining was used to detect the expression level of DEPDC1B in tumor tissues and adjacent normal tissues. After DEPDC1B and CDK1 knockdown in cell lines HEP3B2.1-7 and SK-HEP-1, MTT assay and colony formation assay was used to detect cell growth, flow cytometry assay was used to investigate cell apoptosis and cell cycle, wound-healing assay and Transwell assay were used to examine the tumor cell migration.
    [Show full text]
  • AB079. Upregulation of DEPDC1B Correlates with Tumor
    Translational Andrology and Urology, Vol 6, Suppl 3 August 2017 S243 Keywords: Prostate cancer (PCa); SPC25; prognosis; proliferation; cells via activating Wnt/β-catenin signaling. However, the cell cycle clinical relevance and functional roles of DEPDC1B in PCa remain unclear. In this study, we found that the expression doi: 10.21037/tau.2017.s078 levels of DEPDC1B in PCa tissues were significantly higher than that in non-tumor tissues. Furthermore, our results Cite this abstract as: Cui F, Hu J, Tan J, Tang H. Knockdown showed DEPDC1B was upregulated in high pathology of SPC25 inhibits cell proliferation and cycle progression in stage PCa. Kaplan-Meier analysis showed that Lower prostate cancer. Transl Androl Urol 2017;6(Suppl 3):AB078. DEPDC1B expression level was associated with better doi: 10.21037/tau.2017.s078 survival of PCa patients. GO and KEGG pathway analysis of DEPDC1B co-expressed genes showed DEPDC1B played an important role in regulating PCa proliferation and cell cycle progression. We believed that this study will provide a potential new therapeutic and prognostic target AB079. Upregulation of for prostate cancer. Keywords: Prostate cancer (PCa); DEPDC1B; prognosis; tumor DEPDC1B correlates with progression; cell tumor progression and predicts doi: 10.21037/tau.2017.s079 a poor prognosis in prostate Cite this abstract as: Cui F, Hu J, Tan J, Tang H. Upregulation cancer of DEPDC1B correlates with tumor progression and predicts a poor prognosis in prostate cancer. Transl Androl Urol 2017;6(Suppl 3):AB079. doi: 10.21037/tau.2017.s079 Feilun Cui, Jiangpeng Hu, Jian Tan, Huaming Tang Department of Urology, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang 212000, China AB080.
    [Show full text]
  • Cell Cycle Arrest Through Indirect Transcriptional Repression by P53: I Have a DREAM
    Cell Death and Differentiation (2018) 25, 114–132 Official journal of the Cell Death Differentiation Association OPEN www.nature.com/cdd Review Cell cycle arrest through indirect transcriptional repression by p53: I have a DREAM Kurt Engeland1 Activation of the p53 tumor suppressor can lead to cell cycle arrest. The key mechanism of p53-mediated arrest is transcriptional downregulation of many cell cycle genes. In recent years it has become evident that p53-dependent repression is controlled by the p53–p21–DREAM–E2F/CHR pathway (p53–DREAM pathway). DREAM is a transcriptional repressor that binds to E2F or CHR promoter sites. Gene regulation and deregulation by DREAM shares many mechanistic characteristics with the retinoblastoma pRB tumor suppressor that acts through E2F elements. However, because of its binding to E2F and CHR elements, DREAM regulates a larger set of target genes leading to regulatory functions distinct from pRB/E2F. The p53–DREAM pathway controls more than 250 mostly cell cycle-associated genes. The functional spectrum of these pathway targets spans from the G1 phase to the end of mitosis. Consequently, through downregulating the expression of gene products which are essential for progression through the cell cycle, the p53–DREAM pathway participates in the control of all checkpoints from DNA synthesis to cytokinesis including G1/S, G2/M and spindle assembly checkpoints. Therefore, defects in the p53–DREAM pathway contribute to a general loss of checkpoint control. Furthermore, deregulation of DREAM target genes promotes chromosomal instability and aneuploidy of cancer cells. Also, DREAM regulation is abrogated by the human papilloma virus HPV E7 protein linking the p53–DREAM pathway to carcinogenesis by HPV.Another feature of the pathway is that it downregulates many genes involved in DNA repair and telomere maintenance as well as Fanconi anemia.
    [Show full text]
  • Unravelling the Role of the Microenvironment in Chronic Lymphocytic Leukaemia: T Helper Cell Support of Cancer Cells
    Unravelling the role of the microenvironment in chronic lymphocytic leukaemia: T helper cell support of cancer cells Doctoral thesis by Anna Maria Parente Ribes Centre for Immune Regulation, Department of Immunology, Institute of Clinical Medicine, University of Oslo, Norway 2018 © Anna Maria Parente Ribes, 2018 Series of dissertations submitted to the Faculty of Medicine, University of Oslo ISBN 978-82-8377-230-2 All rights reserved. No part of this publication may be reproduced or transmitted, in any form or by any means, without permission. Cover: Hanne Baadsgaard Utigard. Print production: Reprosentralen, University of Oslo. TABLE OF CONTENTS ACKNOWLEDGEMENTS ....................................................................................................... 5 ABBREVIATIONS .................................................................................................................... 7 LIST OF PUBLICATIONS ....................................................................................................... 8 INTRODUCTION .................................................................................................................... 10 B cells .................................................................................................................................... 10 B cell development ............................................................................................................ 10 Central and peripheral tolerance ..................................................................................
    [Show full text]
  • Mitotic Checkpoints and Chromosome Instability Are Strong Predictors of Clinical Outcome in Gastrointestinal Stromal Tumors
    MITOTIC CHECKPOINTS AND CHROMOSOME INSTABILITY ARE STRONG PREDICTORS OF CLINICAL OUTCOME IN GASTROINTESTINAL STROMAL TUMORS. Pauline Lagarde1,2, Gaëlle Pérot1, Audrey Kauffmann3, Céline Brulard1, Valérie Dapremont2, Isabelle Hostein2, Agnès Neuville1,2, Agnieszka Wozniak4, Raf Sciot5, Patrick Schöffski4, Alain Aurias1,6, Jean-Michel Coindre1,2,7 Maria Debiec-Rychter8, Frédéric Chibon1,2. Supplemental data NM cases deletion frequency. frequency. deletion NM cases Mand between difference the highest setswith of theprobe a view isdetailed panel Bottom frequently. sorted totheless deleted theprobe are frequently from more and thefrequency deletion represent Yaxes inblue. are cases (NM) metastatic for non- frequencies Corresponding inmetastatic (red). probe (M)cases sets figureSupplementary 1: 100 100 20 40 60 80 20 40 60 80 0 0 chr14 1 chr14 88 chr14 175 chr14 262 chr9 -MTAP 349 chr9 -MTAP 436 523 chr9-CDKN2A 610 Histogram presenting the 2000 more frequently deleted deleted frequently the 2000 more presenting Histogram chr9-CDKN2A 697 chr9-CDKN2A 784 chr9-CDKN2B 871 chr9-CDKN2B 958 chr9-CDKN2B 1045 chr22 1132 chr22 1219 chr22 1306 chr22 1393 1480 1567 M NM 1654 1741 1828 1915 M NM GIST14 GIST2 GIST16 GIST3 GIST19 GIST63 GIST9 GIST38 GIST61 GIST39 GIST56 GIST37 GIST47 GIST58 GIST28 GIST5 GIST17 GIST57 GIST47 GIST58 GIST28 GIST5 GIST17 GIST57 CDKN2A Supplementary figure 2: Chromosome 9 genomic profiles of the 18 metastatic GISTs (upper panel). Deletions and gains are indicated in green and red, respectively; and color intensity is proportional to copy number changes. A detailed view is given (bottom panel) for the 6 cases presenting a homozygous 9p21 deletion targeting CDKN2A locus (dark green).
    [Show full text]
  • DEPDC1B Regulates the Progression of Human Chordoma Through
    www.nature.com/cddis ARTICLE OPEN DEPDC1B regulates the progression of human chordoma through UBE2T-mediated ubiquitination of BIRC5 ✉ ✉ Liang Wang1,4, Liang Tang2,4, Ruijun Xu2, Junpeng Ma1, Kaibing Tian1, Yanbin Liu2, Yanghu Lu2, Zhen Wu1 and Xiaodong Zhu3 © The Author(s) 2021 Chordoma is a rare bone malignancy with a high rate of local recurrence and distant metastasis. Although DEP domain-containing protein 1B (DEPDC1B) is implicated in a variety of malignancies, its relationship with chordoma is unclear. In this study, the biological role and molecular mechanism of DEPDC1B in chordoma were explored. The function of DEPDC1B in chordoma cells was clarified through loss-of-function assays in vitro and in vivo. Furthermore, molecular mechanism of DEPDC1B in chordoma cells was recognized by RNA sequencing and Co-Immunoprecipitation (Co-IP) assay. The malignant behaviors of DEPDC1B knockdown chordoma cells was significantly inhibited, which was characterized by reduced proliferation, enhanced apoptosis, and hindered migration. Consistently, decreased expression of DEPDC1B suppressed tumor growth in xenograft mice. Mechanically, DEPDC1B affected the ubiquitination of baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5) through ubiquitin-conjugating enzyme E2T (UBE2T). Simultaneous downregulation of BIRC5 and DEPDC1B may exacerbate the inhibitory effects of chordoma. Moreover, BIRC5 overexpression reduced the inhibitory effects of DEPDC1B knockdown in chordoma cells. In conclusion, DEPDC1B regulates the progression of human chordoma through UBE2T-mediated ubiquitination of BIRC5, suggesting that it may be a promising candidate target with potential therapeutic value. Cell Death and Disease (2021) 12:753 ; https://doi.org/10.1038/s41419-021-04026-7 INTRODUCTION of 529 amino acids, which includes an N-terminal DEP domain and a Chordoma is a rare, slow-growing primary bone malignancy that C-terminal Rho-GAP (GTPase activating protein)-like domain [10].
    [Show full text]