Supplemental Figure Legends

Total Page:16

File Type:pdf, Size:1020Kb

Supplemental Figure Legends Supplemental Figure Legends Supplementary Figure S1 Generation and characterization of Frat-knockout mice (A) Frat/GBP orthologues are conserved amongst vertebrate species, including Xenopus (xl) zebrafish (dr), mice (mm), rats (rn) and humans (hs). Whereas lower organisms such as Xenopus and zebrafish contain one Frat/GBP gene, the human genome harbors two and the mouse genome three Frat homologues. mmFrat1 and mmFrat3 are 84% homologous on the protein level, whereas mmFrat1 and mmFrat2 share 68% amino-acid identity. A multiple- species alignment of Frat proteins reveals conserved domains in the N– and C-terminal regions. The IKEA-box, which is required for binding to GSK-3β, is conserved in all Frat family members. (B) Frat2– and Frat3-knockout mice were generated by replacing most of the coding sequence with a targeting cassette existing of a promoterless lacZ-reporter gene, followed by a PGK-promoter driven Hygromycin resistance marker to allow selection of targeted ES cells. Homologous recombination was analyzed by Southern Blot analysis using 3’ flanking probes. For Frat2 probe A allows detection of the shift of an endogenous 12 kb Xba fragment to a 9 kb fragment after homologous recombination; Probe B for Frat3 detects a shift in the endogenous 24 kb EcoRV fragment to an 18 kb knockout fragment after homologous recombination. (C) Since Frat1 and Frat2 are located on chromosome 19 only 15 kb apart in opposite orientations, a separate targeting vector was constructed to generate Frat1/Frat2 double- knockout mice. Frat2 was targeted with a PGK-Puromycin selection cassette in Frat1+/lacZ ES cells. Homologous recombination was verified by Southern Blot analysis. Clones in which the Frat1-knockout allele had undergone consecutive targeting of the Frat2 gene (Detectable with probe A as a shift from a 35 kb to a 17 kb EcoRV fragment) were used to generate double- knockout mice. (D) RT-PCR analysis on bone marrow (BM) or mouse embryo fibroblasts (MEF) with 3’ UTR primers reveals read-through from the targeting cassettes into the Frat1 and Frat2 3’UTR (upper two panels). RT-PCR analysis with gene-specific primers shows that Frat1, Frat2 and Frat3 are no longer expressed in Frat triple-knockout mice. GAPDH serves as a control. Lanes 1-4 RT-PCR; Lanes 5-8 no-RT control; Lanes 1,4,5,8 triple-heterozgygotes; Lanes 2,3,6,7 Frat triple-knockout mice. Supplementary Figure S2 Analysis of Frat3 imprinting status (A) Frat3 is located in the Prader-Willi syndrome region on mouse chromosome 7 (Chai et al. 2001; Kobayashi et al. 2002). It has acquired an imprinted status, causing Frat3 to be expressed solely from the paternal allele. Using Southern blot analysis similar to (Chai et al. 2001) we analyzed whether targeting of the Frat3 gene had left the imprinting status of the region intact. Indeed, the EagI site remains imprinted upon transmission of a maternal wildtype allele (and a paternal knockout allele, PKO) but not upon transmission of a paternal wildtype allele (and a maternal knockout allele, MKO). XbaI digest: wildtype allele 6.9 kb; knockout allele 4 kb XbaI/EagI digest: wildtype allele imprinted 6.9 kb; wildtype allele not imprinted 2.8 kb; knockout allele 4 kb. (B) Genomic DNA isolated from Frat3+/lacZ mice was digested with methylation-sensitive restriction enzymes (NruI or HpaII) or a methylation-insensitive enzyme as a control (MspI) and used as input for a Frat3-specific PCR that allows detection of a 380 bp wildtype and a 400 bp knockout band. In Frat3+/lacZ mice the wildtype allele was only detected upon maternal transmission of a wildtype allele (and a paternal knockout allele, PKO) and not upon transmission of a paternal wildtype allele (and a maternal knockout allele, MKO), indicating that only in the first case, methylation at the HpaII and NruI sites had protected the wildtype allele from restriction enzyme digestion. The knockout allele has become methylated, since it can be detected upon both paternal and maternal transmission following digestion with HpaII (but not following digestion with MspI). This has likely caused the lacZ reporter to be silenced, since we could not detect lacZ expression following X-gal staining of wholemount embryos or frozen sections of E11.5-E14.5, during which stages Frat3 expression is readily detected by RT-PCR. Thus, although imprinting at the wildtype allele remains intact, the knockout allele appears to have been silenced. However, since Frat3-deficient mice were also viable, healthy and fertile, we conclude that Frat3 is not involved in the Prader-Willi syndrome in mice. Supplementary Figure S3 Frat is expressed at sites that display active Wnt/β-catenin activity Since lacZ expression in Frat2+/lacZ mice is driven by the endogenous Frat2 promoter, analysis of the lacZ expression pattern provides a direct readout for Frat2 expression in vivo. Cryosections from developing embryos show that Frat2 is predominantly expressed in spinal ganglia (A) and brain ventricles (B) from early development onward. At later stages of development, lacZ activity was also detected in areas of the developing heart (C), the retina (D), the edge of the limbs (E) and in developing hair follicles and skin (F). Many of these sites overlap with areas where Wnt/β-catenin signaling has been reported to take place during mouse development (Maretto et al. 2003). Supplementary Figure S4 Frat is expressed in early lymphoid progenitors To characterize Frat expression during lymphoid differentiation, we used the fluorigenic substrate FDG to visualize β-galactosidase activity encoded by the lacZ knock-in allele present in both Frat1 and Frat2 heterozygous animals by flowcytometric detection. Staining patterns in immature B– en T-lymphocytes between Frat1+/lacZ and Frat2+/lacZ were identical (data not shown) and largely overlap with stages that display β-catenin/TCF activity. (A) A large proportion of differentiating T-cells in the thymus is FDG-positive (49%). During differentiation, immature T-cells rearrange the genomic loci encoding the β and α chains of the T-cell receptor (TCR). In both instances, successful rearrangement induces a burst of cellular proliferation. The earliest thymic population of CD4/CD8 double negative CD25-/CD44+ cells (DN1) does not express Frat. However, Frat expression is strongly induced in CD25+/CD44+ cells (DN2), which is the first subpopulation of cells committed to the T-cell lineage. Frat expression is maintained throughout DN3 and DN4. In DN3, T-cells rearrange the TCRβ locus and enter a proliferative phase after β selection. Subsequently, immature T-cells differentiate into CD4/CD8 double-positive (DP) T-cells and initiate TCRα rearrangements. DP T-cells express high levels of Frat. Upon positive selection, T-cells will downregulate CD4/CD8 expression, inititate CD69 expression and commit to either the CD4 or the CD8 lineage. Remarkably, the highest Frat expression (76% of the cells) is observed in the first subpopulation to be distinguished after the initiation of positive selection, the TCRβlo CD69+ DP T-cells, indicating that Frat might be further induced as an early immediate gene after the positive selection event. During lineage commitment to CD4 and CD8 SP T-cells, Frat expression is maintained in CD4 SP T-cells to a low extent. (B) During B-cell differentiation, Frat expression is mainly observed in immature B-cells (B220lo, CD43-) that do not yet express IgM on their cell surface. DN double-negative; DP double-positive; SP single-positive Supplementary Figure S5 T– and B-cell differentiation is not affected by the loss of Frat Flowcytometric analysis of single cell suspensions derived from bone marrow, thymus and spleen from young as well as old Frat-deficient mice did not reveal any subpopulations that were affected by the loss of Frat when compared to littermate controls. Both overall cell numbers as well as the process of B– and T-cell differentiation as measured by the presence of specific subpopulations were not perturbed in the absence of Frat (also data not shown). Mice deficient for Tcf1 or for both Tcf1 and Lef1 show defects in specific stages of T-cell differentiation (Staal and Clevers 2000). Tcf1-/- animals show a progressive decline in T-cell maturation, due to specific blockades at the stage of actively proliferating CD4-CD8- (double- negative, DN) thymocytes (Verbeek et al. 1995; Schilham et al. 1998). Lef1-/- mice display defects in pro-B-cell proliferation (Reya et al. 2000) and in Tcf1-/-;Lef1-/- mice, T-cell differentiation is completely impaired from DN3 onward (Okamura et al. 1998). We therefore paid specific attention to these populations, but here, as in other subpopulations, we found no differences between Frat-TKO mice and control littermates. Additional References Chai, J.H., D.P. Locke, T. Ohta, J.M. Greally, and R.D. Nicholls. 2001. Retrotransposed genes such as Frat3 in the mouse Chromosome 7C Prader- Willi syndrome region acquire the imprinted status of their insertion site. Mamm Genome 12: 813-21. Kobayashi, S., T. Kohda, H. Ichikawa, A. Ogura, M. Ohki, T. Kaneko-Ishino, and F. Ishino. 2002. Paternal expression of a novel imprinted gene, Peg12/Frat3, in the mouse 7C region homologous to the Prader-Willi syndrome region. Biochem Biophys Res Commun 290: 403-8. Maretto, S., M. Cordenonsi, S. Dupont, P. Braghetta, V. Broccoli, A.B. Hassan, D. Volpin, G.M. Bressan, and S. Piccolo. 2003. Mapping Wnt/beta-catenin signaling during mouse development and in colorectal tumors. Proc Natl Acad Sci U S A 100: 3299- 304. Okamura, R.M., M. Sigvardsson, J. Galceran, S. Verbeek, H. Clevers, and R. Grosschedl. 1998. Redundant regulation of T cell differentiation and TCRalpha gene expression by the transcription factors LEF-1 and TCF-1. Immunity 8: 11-20. Reya, T., M. O'Riordan, R.
Recommended publications
  • Posters A.Pdf
    INVESTIGATING THE COUPLING MECHANISM IN THE E. COLI MULTIDRUG TRANSPORTER, MdfA, BY FLUORESCENCE SPECTROSCOPY N. Fluman, D. Cohen-Karni, E. Bibi Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel In bacteria, multidrug transporters couple the energetically favored import of protons to export of chemically-dissimilar drugs (substrates) from the cell. By this function, they render bacteria resistant against multiple drugs. In this work, fluorescence spectroscopy of purified protein is used to unravel the mechanism of coupling between protons and substrates in MdfA, an E. coli multidrug transporter. Intrinsic fluorescence of MdfA revealed that binding of an MdfA substrate, tetraphenylphosphonium (TPP), induced a conformational change in this transporter. The measured affinity of MdfA-TPP was increased in basic pH, raising a possibility that TPP might bind tighter to the deprotonated state of MdfA. Similar increases in affinity of TPP also occurred (1) in the presence of the substrate chloramphenicol, or (2) when MdfA is covalently labeled by the fluorophore monobromobimane at a putative chloramphenicol interacting site. We favor a mechanism by which basic pH, chloramphenicol binding, or labeling with monobromobimane, all induce a conformational change in MdfA, which results in deprotonation of the transporter and increase in the affinity of TPP. PHENOTYPE CHARACTERIZATION OF AZOSPIRILLUM BRASILENSE Sp7 ABC TRANSPORTER (wzm) MUTANT A. Lerner1,2, S. Burdman1, Y. Okon1,2 1Department of Plant Pathology and Microbiology, Faculty of Agricultural, Food and Environmental Quality Sciences, Hebrew University of Jerusalem, Rehovot, Israel, 2The Otto Warburg Center for Agricultural Biotechnology, Faculty of Agricultural, Food and Environmental Quality Sciences, Hebrew University of Jerusalem, Rehovot, Israel Azospirillum, a free-living nitrogen fixer, belongs to the plant growth promoting rhizobacteria (PGPR), living in close association with plant roots.
    [Show full text]
  • Downloaded the “Top Edge” Version
    bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted December 6, 2019. 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 4.0 International license. 1 Drosophila models of pathogenic copy-number variant genes show global and 2 non-neuronal defects during development 3 Short title: Non-neuronal defects of fly homologs of CNV genes 4 Tanzeen Yusuff1,4, Matthew Jensen1,4, Sneha Yennawar1,4, Lucilla Pizzo1, Siddharth 5 Karthikeyan1, Dagny J. Gould1, Avik Sarker1, Yurika Matsui1,2, Janani Iyer1, Zhi-Chun Lai1,2, 6 and Santhosh Girirajan1,3* 7 8 1. Department of Biochemistry and Molecular Biology, Pennsylvania State University, 9 University Park, PA 16802 10 2. Department of Biology, Pennsylvania State University, University Park, PA 16802 11 3. Department of Anthropology, Pennsylvania State University, University Park, PA 16802 12 4 contributed equally to work 13 14 *Correspondence: 15 Santhosh Girirajan, MBBS, PhD 16 205A Life Sciences Building 17 Pennsylvania State University 18 University Park, PA 16802 19 E-mail: [email protected] 20 Phone: 814-865-0674 21 1 bioRxiv preprint doi: https://doi.org/10.1101/855338; this version posted December 6, 2019. 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 4.0 International license. 22 ABSTRACT 23 While rare pathogenic copy-number variants (CNVs) are associated with both neuronal and non- 24 neuronal phenotypes, functional studies evaluating these regions have focused on the molecular 25 basis of neuronal defects.
    [Show full text]
  • Analysis of the Indacaterol-Regulated Transcriptome in Human Airway
    Supplemental material to this article can be found at: http://jpet.aspetjournals.org/content/suppl/2018/04/13/jpet.118.249292.DC1 1521-0103/366/1/220–236$35.00 https://doi.org/10.1124/jpet.118.249292 THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS J Pharmacol Exp Ther 366:220–236, July 2018 Copyright ª 2018 by The American Society for Pharmacology and Experimental Therapeutics Analysis of the Indacaterol-Regulated Transcriptome in Human Airway Epithelial Cells Implicates Gene Expression Changes in the s Adverse and Therapeutic Effects of b2-Adrenoceptor Agonists Dong Yan, Omar Hamed, Taruna Joshi,1 Mahmoud M. Mostafa, Kyla C. Jamieson, Radhika Joshi, Robert Newton, and Mark A. Giembycz Departments of Physiology and Pharmacology (D.Y., O.H., T.J., K.C.J., R.J., M.A.G.) and Cell Biology and Anatomy (M.M.M., R.N.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada Received March 22, 2018; accepted April 11, 2018 Downloaded from ABSTRACT The contribution of gene expression changes to the adverse and activity, and positive regulation of neutrophil chemotaxis. The therapeutic effects of b2-adrenoceptor agonists in asthma was general enriched GO term extracellular space was also associ- investigated using human airway epithelial cells as a therapeu- ated with indacaterol-induced genes, and many of those, in- tically relevant target. Operational model-fitting established that cluding CRISPLD2, DMBT1, GAS1, and SOCS3, have putative jpet.aspetjournals.org the long-acting b2-adrenoceptor agonists (LABA) indacaterol, anti-inflammatory, antibacterial, and/or antiviral activity. Numer- salmeterol, formoterol, and picumeterol were full agonists on ous indacaterol-regulated genes were also induced or repressed BEAS-2B cells transfected with a cAMP-response element in BEAS-2B cells and human primary bronchial epithelial cells by reporter but differed in efficacy (indacaterol $ formoterol .
    [Show full text]
  • Exploring the Relationship Between Gut Microbiota and Major Depressive Disorders
    E3S Web of Conferences 271, 03055 (2021) https://doi.org/10.1051/e3sconf/202127103055 ICEPE 2021 Exploring the Relationship between Gut Microbiota and Major Depressive Disorders Catherine Tian1 1Shanghai American School, Shanghai, China Abstract. Major Depressive Disorder (MDD) is a psychiatric disorder accompanied with a high rate of suicide, morbidity and mortality. With the symptom of an increasing or decreasing appetite, there is a possibility that MDD may have certain connections with gut microbiota, the colonies of microbes which reside in the human digestive system. In recent years, more and more studies started to demonstrate the links between MDD and gut microbiota from animal disease models and human metabolism studies. However, this relationship is still largely understudied, but it is very innovative since functional dissection of this relationship would furnish a new train of thought for more effective treatment of MDD. In this study, by using multiple genetic analytic tools including Allen Brain Atlas, genetic function analytical tools, and MicrobiomeAnalyst, I explored the genes that shows both expression in the brain and the digestive system to affirm that there is a connection between gut microbiota and the MDD. My approach finally identified 7 MDD genes likely to be associated with gut microbiota, implicating 3 molecular pathways: (1) Wnt Signaling, (2) citric acid cycle in the aerobic respiration, and (3) extracellular exosome signaling. These findings may shed light on new directions to understand the mechanism of MDD, potentially facilitating the development of probiotics for better psychiatric disorder treatment. 1 Introduction 1.1 Major Depressive Disorder Major Depressive Disorder (MDD) is a mood disorder that will affect the mood, behavior and other physical parts.
    [Show full text]
  • The Database of Chromosome Imbalance Regions and Genes
    Lo et al. BMC Cancer 2012, 12:235 http://www.biomedcentral.com/1471-2407/12/235 RESEARCH ARTICLE Open Access The database of chromosome imbalance regions and genes resided in lung cancer from Asian and Caucasian identified by array-comparative genomic hybridization Fang-Yi Lo1, Jer-Wei Chang1, I-Shou Chang2, Yann-Jang Chen3, Han-Shui Hsu4, Shiu-Feng Kathy Huang5, Fang-Yu Tsai2, Shih Sheng Jiang2, Rajani Kanteti6, Suvobroto Nandi6, Ravi Salgia6 and Yi-Ching Wang1* Abstract Background: Cancer-related genes show racial differences. Therefore, identification and characterization of DNA copy number alteration regions in different racial groups helps to dissect the mechanism of tumorigenesis. Methods: Array-comparative genomic hybridization (array-CGH) was analyzed for DNA copy number profile in 40 Asian and 20 Caucasian lung cancer patients. Three methods including MetaCore analysis for disease and pathway correlations, concordance analysis between array-CGH database and the expression array database, and literature search for copy number variation genes were performed to select novel lung cancer candidate genes. Four candidate oncogenes were validated for DNA copy number and mRNA and protein expression by quantitative polymerase chain reaction (qPCR), chromogenic in situ hybridization (CISH), reverse transcriptase-qPCR (RT-qPCR), and immunohistochemistry (IHC) in more patients. Results: We identified 20 chromosomal imbalance regions harboring 459 genes for Caucasian and 17 regions containing 476 genes for Asian lung cancer patients. Seven common chromosomal imbalance regions harboring 117 genes, included gain on 3p13-14, 6p22.1, 9q21.13, 13q14.1, and 17p13.3; and loss on 3p22.2-22.3 and 13q13.3 were found both in Asian and Caucasian patients.
    [Show full text]
  • The Expression Profile of FRAT1 in Human Gliomas
    BRAIN RESEARCH 1320 (2010) 152– 158 available at www.sciencedirect.com www.elsevier.com/locate/brainres Research Report The expression profile of FRAT1 in human gliomas Geng Guoa,1, Xinggang Maoa,1, Peng Wanga,1, Bolin Liua, Xiang Zhanga,⁎, Xiaofan Jianga, Chengliang Zhongb, Junli Huoa, Ji Jinc, Yuzhen Zhuod aDepartment of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 15# West Chang Le Road, Xi'an, Shaanxi Province 710032, People's Republic of China bDepartment of Health Statistics, Faculty of Preventative Medicine, Fourth Military Medical University, 17# West Chang Le Road, Xi'an, Shaanxi Province 710032, People's Republic of China cShanxi Medical University, 56# South Xin Jian Road, Taiyuan, Shanxi Province 030001, People's Republic of China dTianjin Institute of Acute Abdomen, Tianjin Nankai Hospital, 122# San Wei Road, Tianjin 300100, People's Republic of China ARTICLE INFO ABSTRACT Article history: FRAT1 was originally characterized as a protein frequently rearranged in advanced T cell Accepted 13 January 2010 lymphoma, which inhibits GSK-3-mediated phosphorylation of β-catenin and positively Available online 21 January 2010 regulates the Wnt signaling pathway. FRAT1 has been identified as a proto-oncogene involved in tumorigenesis. Previous studies have shown that FRAT1 is strikingly Keywords: overexpressed in some human cancers. However, the relationship between FRAT1 and FRAT1 human gliomas is unclear. In this study, we detected the expression of FRAT1 in human β-catenin gliomas by immunohistochemistry, Western blot and RT-PCR. FRAT1 was found to be Glioma specifically expressed in the majority of glioma samples, and their expression levels Immunohistochemistry increased markedly with the increase of WHO grades.
    [Show full text]
  • GSK-3) in Dictyostelium Discoideum and the Role of GSK-3 Binding Partners in Dictyostelium Discoideum and Mammalian Cells
    Investigation into the role of Glycogen Synthase Kinase 3 (GSK-3) in Dictyostelium discoideum and the role of GSK-3 binding partners in Dictyostelium discoideum and mammalian cells Josephine E. Forde School of Biosciences Cardiff University PhD Thesis Submitted: November 2009 UMI Number: U585B60 All rights reserved INFORMATION TO ALL USERS The quality of this reproduction is dependent upon the quality of the copy submitted. In the unlikely event that the author did not send a complete manuscript and there are missing pages, these will be noted. Also, if material had to be removed, a note will indicate the deletion. Dissertation Publishing UMI U585B60 Published by ProQuest LLC 2013. Copyright in the Dissertation held by the Author. Microform Edition © ProQuest LLC. All rights reserved. This work is protected against unauthorized copying under Title 17, United States Code. ProQuest LLC 789 East Eisenhower Parkway P.O. Box 1346 Ann Arbor, Ml 48106-1346 Ca r d if f UNIVERSITY PRIFYSGOL DECLARATION C a e RD yi §> This work has not previously been accepted in substance for any degree and is not concurrently submitted in candidature for any degree. Signed ........... d & c k . - . ...................... (candidate) Date ... ! H | Q s | a o \ p .......... STATEMENT 1 This thesis is being submitted in partial fulfillment of the requirements for the degree of .................... (insert MCh, MD, MPhil, PhD etc, as appropriate) Signed .......d & 0 ( L : ...................... (candidate) Date ........... STATEMENT 2 This thesis is the result of my own independent work/investigation, except where otherwise stated. Other sources are acknowledged by explicit references. Signed d 6 < L - . ............................ (candidate) Date .
    [Show full text]
  • 1471-2105-8-217.Pdf
    BMC Bioinformatics BioMed Central Software Open Access GenMAPP 2: new features and resources for pathway analysis Nathan Salomonis1,2, Kristina Hanspers1, Alexander C Zambon1, Karen Vranizan1,3, Steven C Lawlor1, Kam D Dahlquist4, Scott W Doniger5, Josh Stuart6, Bruce R Conklin1,2,7,8 and Alexander R Pico*1 Address: 1Gladstone Institute of Cardiovascular Disease, 1650 Owens Street, San Francisco, CA 94158 USA, 2Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA, 3Functional Genomics Laboratory, University of California, Berkeley, CA 94720 USA, 4Department of Biology, Loyola Marymount University, 1 LMU Drive, MS 8220, Los Angeles, CA 90045 USA, 5Computational Biology Graduate Program, Washington University School of Medicine, St. Louis, MO 63108 USA, 6Department of Biomolecular Engineering, University of California, Santa Cruz, CA 95064 USA, 7Department of Medicine, University of California, San Francisco, CA 94143 USA and 8Department of Molecular and Cellular Pharmacology, University of California, San Francisco, CA 94143 USA Email: Nathan Salomonis - [email protected]; Kristina Hanspers - [email protected]; Alexander C Zambon - [email protected]; Karen Vranizan - [email protected]; Steven C Lawlor - [email protected]; Kam D Dahlquist - [email protected]; Scott W Doniger - [email protected]; Josh Stuart - [email protected]; Bruce R Conklin - [email protected]; Alexander R Pico* - [email protected] * Corresponding author Published: 24 June 2007 Received: 16 November 2006 Accepted: 24 June 2007 BMC Bioinformatics 2007, 8:217 doi:10.1186/1471-2105-8-217 This article is available from: http://www.biomedcentral.com/1471-2105/8/217 © 2007 Salomonis et al; licensee BioMed Central Ltd.
    [Show full text]
  • Rewirable Gene Regulatory Networks in the Preimplantation Embryonic Development of Three Mammalian Species
    Downloaded from genome.cshlp.org on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press Research Rewirable gene regulatory networks in the preimplantation embryonic development of three mammalian species Dan Xie,1,9 Chieh-Chun Chen,1,9 Leon M. Ptaszek,2,3,4,9 Shu Xiao,1 Xiaoyi Cao,5 Fang Fang,6 Huck H. Ng,6 Harris A. Lewin,7 Chad Cowan,3,4 and Sheng Zhong1,7,8,10 1Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA; 2Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA; 3Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA; 4Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA; 5Center for Biophysics and Computational Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA; 6Gene Regulation Laboratory, Genome Institute of Singapore, 138672 Singapore, Singapore; 7Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA; 8Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA Mammalian preimplantation embryonic development (PED) is thought to be governed by highly conserved processes. While it had been suggested that some plasticity of conserved signaling networks exists among different mammalian species, it was not known to what extent modulation of the genomes and the regulatory proteins could ‘‘rewire’’ the gene regulatory networks (GRN) that control PED. We therefore generated global transcriptional profiles from three mam- malian species (human, mouse, and bovine) at representative stages of PED, including: zygote, two-cell, four-cell, eight-cell, 16-cell, morula and blastocyst.
    [Show full text]
  • Peptidomic Discovery of Short Open Reading Frame-Encoded Peptides in Human Cells
    Peptidomic discovery of short open reading frame-encoded peptides in human cells The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Slavoff, Sarah A., Andrew J. Mitchell, Adam G. Schwaid, Moran N. Cabili, Jiao Ma, Joshua Z. Levin, Amir D. Karger, Bogdan A. Budnik, John L. Rinn, and Alan Saghatelian. 2013. “Peptidomic discovery of short open reading frame-encoded peptides in human cells.” Nature chemical biology 9 (1): 59-64. doi:10.1038/nchembio.1120. http:// dx.doi.org/10.1038/nchembio.1120. Published Version doi:10.1038/nchembio.1120 Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:11717493 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA NIH Public Access Author Manuscript Nat Chem Biol. Author manuscript; available in PMC 2013 July 01. NIH-PA Author ManuscriptPublished NIH-PA Author Manuscript in final edited NIH-PA Author Manuscript form as: Nat Chem Biol. 2013 January ; 9(1): 59–64. doi:10.1038/nchembio.1120. Peptidomic discovery of short open reading frame-encoded peptides in human cells Sarah A. Slavoff1, Andrew J. Mitchell2,*, Adam G. Schwaid1,*, Moran N. Cabili3,4,5, Jiao Ma1, Joshua Z. Levin6, Amir D. Karger7, Bogdan A. Budnik8, John L. Rinn3,5, and Alan Saghatelian1,† 1Department of Chemistry and Chemical Biology, Harvard University,
    [Show full text]
  • Identification of Endogenous Adenomatous Polyposis Coli Interaction Partners and B-Catenin– Independent Targets by Proteomics
    Published OnlineFirst June 3, 2019; DOI: 10.1158/1541-7786.MCR-18-1154 Cancer "-omics" Molecular Cancer Research Identification of Endogenous Adenomatous Polyposis Coli Interaction Partners and b-Catenin– Independent Targets by Proteomics Olesja Popow1,2,3,Joao~ A. Paulo2, Michael H. Tatham4, Melanie S. Volk3, Alejandro Rojas-Fernandez5, Nicolas Loyer3, Ian P. Newton3, Jens Januschke3, Kevin M. Haigis1,6, and Inke Nathke€ 3 Abstract Adenomatous Polyposis Coli (APC) is the most frequently tion between endogenous MINK1 and APC and further mutated gene in colorectal cancer. APC negatively regulates confirmed the negative, and b-catenin–independent, regu- the Wnt signaling pathway by promoting the degradation of lation of MINK1 by APC. Increased Mink1/Msn levels were b-catenin, but the extent to which APC exerts Wnt/b-cate- also observed in mouse intestinal tissue and Drosophila nin–independent tumor-suppressive activity is unclear. To follicular cells expressing mutant Apc/APC when compared identify interaction partners and b-catenin–independent with wild-type tissue/cells. Collectively, our results highlight targets of endogenous, full-length APC, we applied label- the extent and importance of Wnt-independent APC func- free and multiplexed tandem mass tag-based mass spec- tions in epithelial biology and disease. trometry. Affinity enrichment-mass spectrometry identified more than 150 previously unidentified APC interaction Implications: The tumor-suppressive function of APC, the partners. Moreover, our global proteomic analysis revealed most frequently mutated gene in colorectal cancer, is mainly that roughly half of the protein expression changes that attributed to its role in b-catenin/Wnt signaling. Our study occur in response to APC loss are independent of b-catenin.
    [Show full text]
  • Insulin-Like Growth Factor 1 Is a Direct HOXA9 Target Important for Hematopoietic Transformation
    Leukemia (2015) 29, 901–908 © 2015 Macmillan Publishers Limited All rights reserved 0887-6924/15 www.nature.com/leu ORIGINAL ARTICLE Insulin-like growth factor 1 is a direct HOXA9 target important for hematopoietic transformation J Steger, E Füller, M-P Garcia-Cuellar, K Hetzner and RK Slany HOX homeobox proteins are key oncogenic drivers in hematopoietic malignancies. Here we demonstrate that HOXA1, HOXA6 and predominantly HOXA9 are able to induce the production of insulin-like growth factor 1 (Igf1). In chromatin immunoprecipitations, HOXA9 bound directly to the putative promoter and a DNase-hypersensitive region in the first intron of the Igf1 gene. Transcription rates of the Igf1 gene paralleled HOXA9 activity. Primary cells transformed by HOXA9 expressed functional Igf1 receptors and activated the protein kinase Akt in response to Igf1 stimulation, suggesting the existence of an autocrine signaling loop. Genomic deletion of the Igf1 gene by Cre-mediated recombination increased sensitivity toward apoptosis after serum starvation. In addition, the leukemogenic potential of Igf1-negative, HOXA9-transformed cells was impaired, leading to a significant delay in disease development on transplantation into recipient animals. Leukemia (2015) 29, 901–908; doi:10.1038/leu.2014.287 INTRODUCTION factor FGF2 (fibroblast growth factor) are regulated by HOX 14,15 HOX homeobox genes are increasingly acknowledged as impor- proteins. Apart from these examples and despite the tant drivers of malignant transformation in the hematopoietic discovery of numerous HOXA9-binding sites across the genome 16 system. In line with their major regulatory role in hematopoietic by chromatin immunoprecipitation-sequencing ChIP-Seq, func- stem and precursor cell populations, HOX transcription needs to tionally characterized HOX targets are scarce.
    [Show full text]