Supplementary Information For

Total Page:16

File Type:pdf, Size:1020Kb

Supplementary Information For Supplementary Information for White shark genome: ancient elasmobranch adaptations associated with wound healing and the maintenance of genome stability. Nicholas J. Marraa,b,1,2, Michael J. Stanhopeb,1,3,4, Nathaniel Juec, Minghui Wangd, Qi Sund, Paulina Pavinski Bitarb, Vincent P. Richardse, Aleksey Komissarovf, Mike Raykog, Sergey Kliverf, Bryce J. Stanhopeb, Chuck Winklerh, Stephen J. O’Brienf,i, Agostinho Antunes,j,k, Salvador Jorgensenl, Mahmood S. Shivjia,3,4. authors to whom correspondence should be addressed: Michael J. Stanhope: [email protected] Mahmood S. Shivji: [email protected] Stephen J. O’Brien: [email protected] This PDF file includes: Figs. S1 to S3: pgs. 2-4 Table S1: pgs. 5-14 RESULTS; Shark Vision: pg. 15 Additional Methods; Transcriptome and Genome Annotation: pgs. 15-17 Captions for databases S1 to S8: pgs. 17-18. References for SI reference citations: pgs. 18-26 1 www.pnas.org/cgi/doi/10.1073/pnas.1819778116 Supplementary figures Fig. S1. K-mer plot of white shark genome sequence reads. Includes 23 bp lengths for all 2x150 bp reads (with the exclusion of the SWIFT kit sequences) included in the SOAPdenovo genome assembly, versus their frequency in the read pool. Several k-mer analyses were conducted to obtain an average estimate of genome size, of which this figure is representative. Genome size was calculated using the frequency of the k- mers present in the largest local maximum peak of the plot, using the following formula: G = Nk-mer/Ck-mer where G is genome size, N is the total number of error free k-mers expected from the sequence data, and C is the frequency of k-mers at the main peak. The plot also indicates a low level of heterozygosity (genomes with high heterozygosity have a secondary peak to the left of the local maximum), and a high repeat content, as shown by the large tail on the bottom right of the figure. There are also several areas of possible duplicated sequence, evidenced by the presence of several small secondary peaks to the right of the main genome size peak (magnified in the inset figure). The axes are plotted on a log-log scale to illustrate better the repeat tail. 2 Fig S2. Benchmarking Universal Single-Copy Orthologs (BUSCO). Assessment of the presence and completeness of Metazoan and Vertebrate BUSCO in the independently, de novo constructed genomes, of C. milli, R. typus, and C. carcharias as well as a multi- tissue transcriptome of C. carcharias. 3 Fig. S3. Break-down of the composition of the white shark genome assembly. The percent of the genome assembly belonging to introns and exons came from the MAKER output which provides an average intron and exon size along with the predicted number of genes. Segmental duplications were identified as sections of the genome that had >90% similarity to another portion of the genome over a continuous 1 kbp region as identified by BLASTN or that had 6 of 7 consecutive sliding windows of 1 kbp with a mean read depth of > mean + 3.5 SD, when the reads of the single end 150 bp library were mapped back to the genome. All repeat categories were determined by using the program RepeatMasker by using vertebrate + custom white shark repeats. The remaining percentage of the genome assembly not covered by these categories was defined as general 'Non-coding Regions'. 4 Table S1. Annotated list of positively selected genome stability related genes presented in Table 1 of the manuscript. Gene Protein name Role in genome Genome stability stability relevant GO terms CHEK2* Serine/threonine- DNA repair, apoptosis, DNA damage checkpoint; protein kinase tumor suppressor (1-8) double-strand break repair; Chk2 regulation of apoptotic process; regulation of signal transduction by p53 class mediator RFC5* Replication factor DNA repair; translesion DNA damage response, C subunit 5 synthesis; nucleotide detection of DNA damage; excision repair (9) DNA repair; translesion synthesis; nucleotide- excision repair FBXO45*! F-box/SPRY Regulates/degrades cellular response to DNA domain- tumor suppressor TP73 damage stimulus containing protein (10) 1 DICER1* Endoribonuclease siRNA and miRNA apoptotic DNA Dicer biogenesis; DNA repair fragmentation; miRNA (11-15) loading onto RISC involved in gene silencing by miRNA; conversion of ds siRNA to ss siRNA involved in RNA interference INO80B* INO80 complex Chromatin remodelling DNA repair; chromatin subunit B and DNA repair (16-18) remodelling; DNA recombination; cellular response to DNA damage stimulus DTL* Denticleless DNA damage response cellular response to DNA protein and translesion DNA damage stimulus; signal synthesis (19-25) transduction involved in G2 DNA damage checkpoint; POLD3* DNA polymerase DNA repair (26-30) nucleotide-excision repair, delta subunit 3 DNA incision, 5'-to lesion; telomere maintenance; translesion synthesis FEM1B* Protein fem-1 Apoptosis and DNA regulation of extrinsic homolog B repair (31,32) apoptotic signaling pathway via death domain receptors; regulation of 5 DNA damage checkpoint SIRT7* NAD-dependent DNA repair; chromatin chromatin organization; protein remodelling; apoptosis; NAD-dependent histone deacetylase regulates p53 (33-37) deacetylase activity (H3- sirtuin-7 K18 specific) PLK2* Serine/threonine- Cell cycle control; DNA damage response, protein kinase regulates tumor growth signal transduction by p53 PLK2 and apoptosis (38-42) class mediator resulting in cell cycle arrest; negative regulation of apoptotic process; CENPS* Centromere DNA repair (43-45) DNA repair; interstrand protein S cross-link repair; chromatin binding; cellular response to DNA damage stimulus; replication fork processing; CASS4* Cas scaffolding Cell adhesion and cell Cell adhesion protein family spreading; apoptosis member 4 (46-48) UFD1* Ubiquitin Protein negative regulation of recognition factor deubiquitination; core telomerase activity; in ER-associated component of p97- ubiquitin-dependent degradation UFD1-NPL4 complex ERAD pathway protein 1 involved in protein extraction from chromatin (49,50) AGT* Angiotensinogen Apotosis; cell regulation of apototic proliferation (51-54) process; regulation of cell proliferation; ERK1 and ERK2 cascade RPS6* 40S ribosomal Apoptosis (55,56) regulation of apoptotic protein S6 process; TOR signalling MYOG* Myogenin Regulation of cell positive regulation of cell proliferation and cell cycle arrest; regulation of cycle arrest (57,58) cell proliferation; chromatin DNA binding USP13* Ubiquitin Controls autophagy and regulation of autophagy; carboxyl-terminal p53 levels; cell cell proliferation; protein hydrolase 13 proliferation (59) deubiquitination PRIM1* DNA primase Okazaki fragment DNA replication, synthesis small subunit synthesis (60) of RNA primer; telomere maintenance via semi- conservative replication; G1/S transition of mitotic 6 cell cycle ALKBH7* Alpha- Necrosis (61) cellular response to DNA ketoglutarate damage stimulus; dependent programmed cell death; dioxygenase alkB regulation of homolog7 mitochondrial membrane permeability involved in programmed necrotic cell death BUD23* 18S rRNA Chromatin remodeling chromatin organization; (guanine-N(7))- (62) methyltransferase FIGL1# Fidgetin-like-1 Double strand break regulation of double-strand repair; regulation of break repair via meiotic recombination homologous (63,64) recombination; negative regulation of reciprocal meiotic recombination CTNNBL1# Beta-catenin-like Transcription coupled positive regulation of protein 1 repair; apoptosis (65-67) apoptotic process; mRNA splicing, via spliceosome CMTM7# CKLF-like Tumor suppressor (68) cytokine activity MARVEL transmembrane domain- containing protein 7 MDM4# Protein MDM4 p53 regulator (69-71) DNA damage response; signal transduction by p53 class mediator resulting in cell cycle arrest ARL6IP5# PRA1 family Apoptosis (72-79) intrinsic apoptotic protein 3 signaling pathway in response to oxidative stress; positive regulation of stress-activated MAPK cascade KIAA1324# UPF0577 protein Autophagy; tumor positive regulation of KIAA1324 suppressor (80-84) autophagosome assembly SALL4# Sal-like protein 4 DNA damage response; somatic stem cell stem cell maintenance population maintenance; (85-87) regulation of transcription, DNA-templated 7 PDCD2@! Programmed cell Apoptosis; regulation of positive regulation of death protein 2 stem cell proliferation apoptotic process; positive (88-90) regulation of hematopoietic stem cell proliferation PDCD4! Programmed cell Apoptosis; tumor positive regulation of death protein 4 suppressor (91-94) vascular associated smooth muscle cell apoptotic process; positive regulation of endothelial cell apoptotic process; cell aging; negative regulation of apoptotic process NHP2! H/ACA Telomere maintenance telomere maintenance via ribonucleoprotein (95-97) telomerase complex subunit 2 RRS1! Ribosome p53 regulator (98-102) regulation of signal biogenesis transduction by p53 class regulatory protein mediator homolog *= white shark # = whale shark @ = elephant shark ! = elasmobranchs Annotation and references: Genome stability PS genes CHK2 Activated when DNA undergoes a double-strand break (1); more specifically, phosphatidylinositol kinase family protein (PIKK) ATM, phosphorylates site Thr68 and activates CHK2 (1,2). CHK2 then in turn, phosphorylates targets such as CDC25 (cell division control protein 25), responsible for removing phosphate from active site residues and activating the cyclin-dependent kinases (CDKs). Thus, CHK2’s inhibition of CDC25 phosphatases prevents entry of the cell into mitosis. The CHK2 protein also interacts with and stabilizes
Recommended publications
  • Deregulated Gene Expression Pathways in Myelodysplastic Syndrome Hematopoietic Stem Cells
    Leukemia (2010) 24, 756–764 & 2010 Macmillan Publishers Limited All rights reserved 0887-6924/10 $32.00 www.nature.com/leu ORIGINAL ARTICLE Deregulated gene expression pathways in myelodysplastic syndrome hematopoietic stem cells A Pellagatti1, M Cazzola2, A Giagounidis3, J Perry1, L Malcovati2, MG Della Porta2,MJa¨dersten4, S Killick5, A Verma6, CJ Norbury7, E Hellstro¨m-Lindberg4, JS Wainscoat1 and J Boultwood1 1LRF Molecular Haematology Unit, NDCLS, John Radcliffe Hospital, Oxford, UK; 2Department of Hematology Oncology, University of Pavia Medical School, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; 3Medizinische Klinik II, St Johannes Hospital, Duisburg, Germany; 4Division of Hematology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden; 5Department of Haematology, Royal Bournemouth Hospital, Bournemouth, UK; 6Albert Einstein College of Medicine, Bronx, NY, USA and 7Sir William Dunn School of Pathology, University of Oxford, Oxford, UK To gain insight into the molecular pathogenesis of the the World Health Organization.6,7 Patients with refractory myelodysplastic syndromes (MDS), we performed global gene anemia (RA) with or without ringed sideroblasts, according to expression profiling and pathway analysis on the hemato- poietic stem cells (HSC) of 183 MDS patients as compared with the the French–American–British classification, were subdivided HSC of 17 healthy controls. The most significantly deregulated based on the presence or absence of multilineage dysplasia. In pathways in MDS include interferon signaling, thrombopoietin addition, patients with RA with excess blasts (RAEB) were signaling and the Wnt pathways. Among the most signifi- subdivided into two categories, RAEB1 and RAEB2, based on the cantly deregulated gene pathways in early MDS are immuno- percentage of bone marrow blasts.
    [Show full text]
  • Coupling of Spliceosome Complexity to Intron Diversity
    bioRxiv preprint doi: https://doi.org/10.1101/2021.03.19.436190; this version posted March 20, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Coupling of spliceosome complexity to intron diversity Jade Sales-Lee1, Daniela S. Perry1, Bradley A. Bowser2, Jolene K. Diedrich3, Beiduo Rao1, Irene Beusch1, John R. Yates III3, Scott W. Roy4,6, and Hiten D. Madhani1,6,7 1Dept. of Biochemistry and Biophysics University of California – San Francisco San Francisco, CA 94158 2Dept. of Molecular and Cellular Biology University of California - Merced Merced, CA 95343 3Department of Molecular Medicine The Scripps Research Institute, La Jolla, CA 92037 4Dept. of Biology San Francisco State University San Francisco, CA 94132 5Chan-Zuckerberg Biohub San Francisco, CA 94158 6Corresponding authors: [email protected], [email protected] 7Lead Contact 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.19.436190; this version posted March 20, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. SUMMARY We determined that over 40 spliceosomal proteins are conserved between many fungal species and humans but were lost during the evolution of S. cerevisiae, an intron-poor yeast with unusually rigid splicing signals. We analyzed null mutations in a subset of these factors, most of which had not been investigated previously, in the intron-rich yeast Cryptococcus neoformans.
    [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]
  • Functional Screening of Alzheimer Risk Loci Identifies PTK2B
    OPEN Molecular Psychiatry (2017) 22, 874–883 www.nature.com/mp ORIGINAL ARTICLE Functional screening of Alzheimer risk loci identifies PTK2B as an in vivo modulator and early marker of Tau pathology P Dourlen1,2,3, FJ Fernandez-Gomez4,5,6, C Dupont1,2,3, B Grenier-Boley1,2,3, C Bellenguez1,2,3, H Obriot4,5,6, R Caillierez4,5,6, Y Sottejeau1,2,3, J Chapuis1,2,3, A Bretteville1,2,3, F Abdelfettah1,2,3, C Delay1,2,3, N Malmanche1,2,3, H Soininen7, M Hiltunen7,8, M-C Galas4,5,6, P Amouyel1,2,3,9, N Sergeant4,5,6, L Buée4,5,6, J-C Lambert1,2,3,11 and B Dermaut1,2,3,10,11 A recent genome-wide association meta-analysis for Alzheimer’s disease (AD) identified 19 risk loci (in addition to APOE) in which the functional genes are unknown. Using Drosophila, we screened 296 constructs targeting orthologs of 54 candidate risk genes within these loci for their ability to modify Tau neurotoxicity by quantifying the size of 46000 eyes. Besides Drosophila Amph (ortholog of BIN1), which we previously implicated in Tau pathology, we identified p130CAS (CASS4), Eph (EPHA1), Fak (PTK2B) and Rab3-GEF (MADD) as Tau toxicity modulators. Of these, the focal adhesion kinase Fak behaved as a strong Tau toxicity suppressor in both the eye and an independent focal adhesion-related wing blister assay. Accordingly, the human Tau and PTK2B proteins biochemically interacted in vitro and PTK2B co-localized with hyperphosphorylated and oligomeric Tau in progressive pathological stages in the brains of AD patients and transgenic Tau mice.
    [Show full text]
  • Sirt7 Promotes Adipogenesis in the Mouse by Inhibiting Autocatalytic
    Sirt7 promotes adipogenesis in the mouse by inhibiting PNAS PLUS autocatalytic activation of Sirt1 Jian Fanga,1, Alessandro Iannia,1, Christian Smolkaa,b, Olesya Vakhrushevaa,c, Hendrik Noltea,d, Marcus Krügera,d, Astrid Wietelmanna, Nicolas G. Simonete, Juan M. Adrian-Segarraa, Alejandro Vaqueroe, Thomas Brauna,2, and Eva Bobera,2 aDepartment of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany; bMedizin III Kardiologie und Angiologie, Universitätsklinikum Freiburg, D-79106 Freiburg, Germany; cDepartment of Medicine, Hematology/Oncology, Goethe University, D-60595 Frankfurt am Main, Germany; dInstitute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), D-50931 Köln, Germany; and eCancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L’Hospitalet de Llobregat, Barcelona, Catalonia, Spain Edited by C. Ronald Kahn, Section of Integrative Physiology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, and approved August 23, 2017 (received for review April 26, 2017) Sirtuins (Sirt1–Sirt7) are NAD+-dependent protein deacetylases/ adipogenesis and accumulation of lipids in 3T3-L1 adipocytes by ADP ribosyltransferases, which play decisive roles in chromatin deacetylation of FOXO1, which represses PPARγ (10). The role of silencing, cell cycle regulation, cellular differentiation, and metab- Sirt6 in the regulation of adipogenic differentiation is less clear olism. Different sirtuins control similar cellular processes, suggest- although it is known that Sirt6 knockout (KO) mice suffer from ing a coordinated mode of action but information about potential reduced adipose tissue stores, while Sirt6 overexpressing mice cross-regulatory interactions within the sirtuin family is still lim- are protected against high-fat diet-induced obesity (11, 12).
    [Show full text]
  • Supplementary Text and Figures.Pdf
    Supplemental Figure S2. PRIM1 immunoblots (20-75 kDa). (A) Corresponds to Figure 2D, with additional bands seen in P2 at 75 kDa and 25 kDa. (B) Independent experiment with cell lysates from C and P2 in which additional bands at 25 kDa not evident. (C) Corresponds to Figure 2D. No additional band visible at 25 kDa. (D) Validation of monoclonal antibody (8G10) with siRNA to PRIM1 in HeLa cells. siLUC, luciferase negative control. siPRIM1, siRNA targeting PRIM1 transcript. Vinculin used as a loading control. Supplemental Figure S3: Predicted destabilisation of PRIM1 by the C301R substitution. (A) Cysteine 301 lies in a buried hydrophobic region in PRIM1. DNA primase dimer crystal structure (PDB: 4BPU). PRIM1 residues shaded according to solvent accessibility. Cysteine 301, substituted to arginine in P5, lies in a buried hydrophobic region. (B) Cys301 is evolutionarily conserved in vertebrates. Neither arginine or other large or charged amino acids are observed at this position in other species. (C) The C301R substitution observed in P5 is predicted to lead to destabilisation of all available PRIM1 crystal structures. In contrast, substitution with leucine or threonine, as observed in some orthologous proteins, is not predicted to lead to destabilisation. Data points, predicted changes in free-energy (ΔΔG) from FoldX plotted for each of 14 available crystal structures. (D) Immunoblotting of RPE1 cells transfected with dual reporter constructs (as described in Figure 3B and Materials and Methods) confirms production of PRIM1-GFP and FLAG-SR at expected molecular weights and shows reduced protein levels for the C301R variant. n=2 experiments shown. C301R protein levels relative to WT after normalization to actin loading control indicated underneath the blot.
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [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]
  • Essential Genes and Their Role in Autism Spectrum Disorder
    University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2017 Essential Genes And Their Role In Autism Spectrum Disorder Xiao Ji University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Bioinformatics Commons, and the Genetics Commons Recommended Citation Ji, Xiao, "Essential Genes And Their Role In Autism Spectrum Disorder" (2017). Publicly Accessible Penn Dissertations. 2369. https://repository.upenn.edu/edissertations/2369 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/2369 For more information, please contact [email protected]. Essential Genes And Their Role In Autism Spectrum Disorder Abstract Essential genes (EGs) play central roles in fundamental cellular processes and are required for the survival of an organism. EGs are enriched for human disease genes and are under strong purifying selection. This intolerance to deleterious mutations, commonly observed haploinsufficiency and the importance of EGs in pre- and postnatal development suggests a possible cumulative effect of deleterious variants in EGs on complex neurodevelopmental disorders. Autism spectrum disorder (ASD) is a heterogeneous, highly heritable neurodevelopmental syndrome characterized by impaired social interaction, communication and repetitive behavior. More and more genetic evidence points to a polygenic model of ASD and it is estimated that hundreds of genes contribute to ASD. The central question addressed in this dissertation is whether genes with a strong effect on survival and fitness (i.e. EGs) play a specific oler in ASD risk. I compiled a comprehensive catalog of 3,915 mammalian EGs by combining human orthologs of lethal genes in knockout mice and genes responsible for cell-based essentiality.
    [Show full text]
  • 1) (As of December 2018) and the Latest GWAS of AD (2
    SUPPLEMENTARY FIGURES downstream intergenic ncRNA_exonic upstream ●936 ●918 group downstream intergenic ncRNA_exonic upstream group exonic exonicintronicintronic ncRNA_intronic ncRNA_intronicUTR3 UTR3 3.8% 1.2%1.5%1.9% 3.8% 5.4%5.4% 750 0.3% 3.8%1.2%1.5%1.9% ●700 5.4% ●670 0.3% 500 45.8% 40.240.2% % 45.8% ●329 ●274 250 ●223 Number (GWAS SNPs/studies) Number (GWAS ●128 ●105 45.8% ●54 ●57 ●58 ●48 ●42 ●46 ●50 ●30 ●3740.2% ● ●17 ●25 ●4 ●6 ●12 0 ● 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016 2017 2018 2019 Year Supplementary Figure S1. GWAS of AD since 2007. The figure is based on data from the GWAS Catalog (1) (as of December 2018) and the latest GWAS of AD (2). The green area shows the total number of AD-associated SNPs, and the purple area shows the total number of GWAS of AD. The insert chart shows the proportions of different types of all 936 AD-associated SNPs. 1 100 200 RPS27A TGFB2 BIN1 C4BPB MSH2 PROC UGT1A1 RAB1A TTN DISC1 50 PDCL3 COL4A3 CD55 ERCC3 100 USP21 C4BPA ITSN2 PTPRF MPZ FMN2 INPP5D CEP85 FNBP1L CSF1 CD46 ADAMTS4 PRKRA SPRED2 0 CTNNA2 DGKD ADCY10 ZAP70 LIMS2 PDE1A PROX1 0 CHRNB2 CR1 HSPG2 SH3BGRL3 DAB1 CTBS FCER1G MAP3K2 AD risk score or log10(P value) IL6R CDC73 CD34 AD risk score or log10(P value) −50 B4GALT3 IL19 0 50 100 150 200 250 0 50 100 150 200 Chromosome 1 (Mb) Chromosome 2 (Mb) ATP2B2 LTF ARF4 MECOM PAK2 EPHB1 40 VHL PRSS42 ARL6IP5 150 COL25A1 TDGF1 RPSA CCR2 CCR1 IL1RAP IRAK2 20 PTPRG 100 FLNB TF CX3CR1 IL17RD SH3RF1 FGG FANCD2 LIMD1 CCR5 50 0 WDR1 PDGFRA EIF4E FGB AD risk score or log10(P value) AD risk
    [Show full text]
  • Molecular Anatomy and Regulation of a Stable Replisome at a Paused Eukaryotic DNA Replication Fork
    Downloaded from genesdev.cshlp.org on September 29, 2021 - Published by Cold Spring Harbor Laboratory Press Molecular anatomy and regulation of a stable replisome at a paused eukaryotic DNA replication fork Arturo Calzada,1,2,3 Ben Hodgson,1,3 Masato Kanemaki,1 Avelino Bueno,2 and Karim Labib1,4 1Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Manchester M20 4BX, United Kingdom; 2Cancer Research Institute, University of Salamanca/CSIC, 37007 Salamanca, Spain Eukaryotic cells regulate the progression and integrity of DNA replication forks to maintain genomic stability and couple DNA synthesis to other processes. The budding yeast proteins Mrc1 and Tof1 associate with the putative MCM–Cdc45 helicase and limit progression of the replisome when nucleotides are depleted, and the checkpoint kinases Mec1 and Rad53 stabilize such stalled forks and prevent disassembly of the replisome. Forks also pause transiently during unperturbed chromosome replication, at sites where nonnucleosomal proteins bind DNA tightly. We describe a method for inducing prolonged pausing of forks at protein barriers assembled at unique sites on a yeast chromosome, allowing us to examine for the first time the effects of pausing upon replisome integrity. We show that paused forks maintain an intact replisome that contains Mrc1, Tof1, MCM–Cdc45, GINS, and DNA polymerases ␣ and ␧ and that recruits the Rrm3 helicase. Surprisingly, pausing does not require Mrc1, although Tof1 and Csm3 are both important. In addition, the integrity of the paused forks does not require Mec1, Rad53, or recombination. We also show that paused forks at analogous barriers in the rDNA are regulated similarly. These data indicate that paused and stalled eukaryotic replisomes resemble each other but are regulated differently.
    [Show full text]
  • Supplementary Table S1. Correlation Between the Mutant P53-Interacting Partners and PTTG3P, PTTG1 and PTTG2, Based on Data from Starbase V3.0 Database
    Supplementary Table S1. Correlation between the mutant p53-interacting partners and PTTG3P, PTTG1 and PTTG2, based on data from StarBase v3.0 database. PTTG3P PTTG1 PTTG2 Gene ID Coefficient-R p-value Coefficient-R p-value Coefficient-R p-value NF-YA ENSG00000001167 −0.077 8.59e-2 −0.210 2.09e-6 −0.122 6.23e-3 NF-YB ENSG00000120837 0.176 7.12e-5 0.227 2.82e-7 0.094 3.59e-2 NF-YC ENSG00000066136 0.124 5.45e-3 0.124 5.40e-3 0.051 2.51e-1 Sp1 ENSG00000185591 −0.014 7.50e-1 −0.201 5.82e-6 −0.072 1.07e-1 Ets-1 ENSG00000134954 −0.096 3.14e-2 −0.257 4.83e-9 0.034 4.46e-1 VDR ENSG00000111424 −0.091 4.10e-2 −0.216 1.03e-6 0.014 7.48e-1 SREBP-2 ENSG00000198911 −0.064 1.53e-1 −0.147 9.27e-4 −0.073 1.01e-1 TopBP1 ENSG00000163781 0.067 1.36e-1 0.051 2.57e-1 −0.020 6.57e-1 Pin1 ENSG00000127445 0.250 1.40e-8 0.571 9.56e-45 0.187 2.52e-5 MRE11 ENSG00000020922 0.063 1.56e-1 −0.007 8.81e-1 −0.024 5.93e-1 PML ENSG00000140464 0.072 1.05e-1 0.217 9.36e-7 0.166 1.85e-4 p63 ENSG00000073282 −0.120 7.04e-3 −0.283 1.08e-10 −0.198 7.71e-6 p73 ENSG00000078900 0.104 2.03e-2 0.258 4.67e-9 0.097 3.02e-2 Supplementary Table S2.
    [Show full text]