Mouse Rgs16 Conditional Knockout Project (CRISPR/Cas9)

Total Page:16

File Type:pdf, Size:1020Kb

Mouse Rgs16 Conditional Knockout Project (CRISPR/Cas9) https://www.alphaknockout.com Mouse Rgs16 Conditional Knockout Project (CRISPR/Cas9) Objective: To create a Rgs16 conditional knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Rgs16 gene (NCBI Reference Sequence: NM_011267 ; Ensembl: ENSMUSG00000026475 ) is located on Mouse chromosome 1. 5 exons are identified, with the ATG start codon in exon 1 and the TGA stop codon in exon 5 (Transcript: ENSMUST00000027748). Exon 3~4 will be selected as conditional knockout region (cKO region). Deletion of this region should result in the loss of function of the Mouse Rgs16 gene. To engineer the targeting vector, homologous arms and cKO region will be generated by PCR using BAC clone RP24-75F11 as template. Cas9, gRNA and targeting vector will be co-injected into fertilized eggs for cKO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Mice homozygous for a knock-out allele exhibit increased fatty acid oxidation and circulating ketone levels when fed a high-fat diet. Mice homozygous for a different knock-out allele exhibit impaired Th1 and Th2 chemotaxis and increased susceptibility toparasitic infection. Exon 3 starts from about 25.87% of the coding region. The knockout of Exon 3~4 will result in frameshift of the gene. The size of intron 2 for 5'-loxP site insertion: 646 bp, and the size of intron 4 for 3'-loxP site insertion: 1504 bp. The size of effective cKO region: ~1010 bp. The cKO region does not have any other known gene. Page 1 of 7 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele 5' gRNA region gRNA region 3' 1 2 3 4 5 Targeting vector Targeted allele Constitutive KO allele (After Cre recombination) Legends Homology arm Exon of mouse Rgs16 cKO region loxP site Page 2 of 7 https://www.alphaknockout.com Overview of the Dot Plot Window size: 10 bp Forward Reverse Complement Sequence 12 Note: The sequence of homologous arms and cKO region is aligned with itself to determine if there are tandem repeats. Tandem repeats are found in the dot plot matrix. It may be difficult to construct this targeting vector. Overview of the GC Content Distribution Window size: 300 bp Sequence 12 Summary: Full Length(7510bp) | A(23.28% 1748) | C(25.79% 1937) | T(25.85% 1941) | G(25.09% 1884) Note: The sequence of homologous arms and cKO region is analyzed to determine the GC content. Significant high GC-content regions are found. It may be difficult to construct this targeting vector. Page 3 of 7 https://www.alphaknockout.com BLAT Search Results (up) QUERY SCORE START END QSIZE IDENTITY CHROM STRAND START END SPAN ----------------------------------------------------------------------------------------------- browser details YourSeq 3000 1 3000 3000 100.0% chr1 + 153738371 153741370 3000 browser details YourSeq 169 70 286 3000 89.8% chr5 + 144084600 144084879 280 browser details YourSeq 160 61 290 3000 90.5% chr7 - 129130412 129130712 301 browser details YourSeq 154 18 218 3000 91.5% chr8 + 77125026 77272600 147575 browser details YourSeq 154 32 290 3000 90.3% chr12 + 100680028 100680284 257 browser details YourSeq 154 32 247 3000 85.5% chr10 + 78083540 78083746 207 browser details YourSeq 151 32 310 3000 89.2% chr1 - 120288583 120288869 287 browser details YourSeq 150 61 279 3000 86.7% chrX + 95674842 95675043 202 browser details YourSeq 149 91 277 3000 93.3% chr10 - 91263654 91263851 198 browser details YourSeq 149 61 346 3000 90.0% chrX + 153218530 153219032 503 browser details YourSeq 148 61 244 3000 91.4% chr7 - 123441304 123646368 205065 browser details YourSeq 148 55 250 3000 90.3% chr10 - 6008641 6008903 263 browser details YourSeq 148 61 310 3000 83.1% chr1 + 161206800 161207017 218 browser details YourSeq 147 65 310 3000 87.8% chr11 + 106358349 106358610 262 browser details YourSeq 145 61 284 3000 83.5% chr9 + 94632660 94632863 204 browser details YourSeq 145 63 273 3000 88.7% chr9 + 75956686 75957095 410 browser details YourSeq 144 61 242 3000 92.5% chr12 - 91520068 91520249 182 browser details YourSeq 143 32 310 3000 83.1% chr10 + 23891263 23891516 254 browser details YourSeq 143 60 236 3000 91.0% chr10 + 18644464 18644641 178 browser details YourSeq 142 88 348 3000 84.1% chr13 - 93511673 93511922 250 Note: The 3000 bp section upstream of Exon 3 is BLAT searched against the genome. No significant similarity is found. BLAT Search Results (down) QUERY SCORE START END QSIZE IDENTITY CHROM STRAND START END SPAN ----------------------------------------------------------------------------------------------- browser details YourSeq 3000 1 3000 3000 100.0% chr1 + 153742381 153745380 3000 browser details YourSeq 73 855 1105 3000 77.3% chr16 - 13220994 13221165 172 browser details YourSeq 62 109 552 3000 77.2% chr13 + 53336467 53336821 355 browser details YourSeq 60 855 1080 3000 79.8% chr16 - 36003851 36004046 196 browser details YourSeq 60 856 981 3000 77.7% chr1 + 98072860 98072964 105 browser details YourSeq 58 868 993 3000 81.3% chr12 - 17519658 17519763 106 browser details YourSeq 51 855 1090 3000 72.6% chr12 - 16615019 16615197 179 browser details YourSeq 51 856 968 3000 79.0% chr12 - 3863649 3863741 93 browser details YourSeq 50 676 894 3000 91.6% chr17 - 7649479 7649887 409 browser details YourSeq 50 863 993 3000 93.2% chr13 - 98826687 98826833 147 browser details YourSeq 49 856 1087 3000 74.6% chr19 - 7390874 7391060 187 browser details YourSeq 48 863 981 3000 73.2% chr13 - 26433626 26433726 101 browser details YourSeq 47 855 1096 3000 92.8% chr13 - 53388866 53389401 536 browser details YourSeq 46 855 965 3000 76.0% chr1 + 175545436 175545526 91 browser details YourSeq 46 1095 1363 3000 67.5% chr1 + 169691840 169692288 449 browser details YourSeq 45 867 985 3000 74.6% chr10 - 41112590 41112687 98 browser details YourSeq 45 855 902 3000 98.0% chr14 + 61419795 61419848 54 browser details YourSeq 44 856 967 3000 72.0% chr16 + 96045432 96045508 77 browser details YourSeq 44 856 1059 3000 89.1% chr1 + 192514371 192514765 395 browser details YourSeq 43 862 967 3000 75.6% chr12 - 82540386 82540471 86 Note: The 3000 bp section downstream of Exon 4 is BLAT searched against the genome. No significant similarity is found. Page 4 of 7 https://www.alphaknockout.com Gene and protein information: Rgs16 regulator of G-protein signaling 16 [ Mus musculus (house mouse) ] Gene ID: 19734, updated on 12-Aug-2019 Gene summary Official Symbol Rgs16 provided by MGI Official Full Name regulator of G-protein signaling 16 provided by MGI Primary source MGI:MGI:108407 See related Ensembl:ENSMUSG00000026475 Gene type protein coding RefSeq status VALIDATED Organism Mus musculus Lineage Eukaryota; Metazoa; Chordata; Craniata; Vertebrata; Euteleostomi; Mammalia; Eutheria; Euarchontoglires; Glires; Rodentia; Myomorpha; Muroidea; Muridae; Murinae; Mus; Mus Also known as Rgsr; Rgs14 Expression Broad expression in liver E18 (RPKM 12.8), CNS E11.5 (RPKM 10.4) and 15 other tissues See more Orthologs human all Genomic context Location: 1 G3; 1 65.43 cM See Rgs16 in Genome Data Viewer Exon count: 5 Annotation release Status Assembly Chr Location 108 current GRCm38.p6 (GCF_000001635.26) 1 NC_000067.6 (153740353..153745468) Build 37.2 previous assembly MGSCv37 (GCF_000001635.18) 1 NC_000067.5 (155587483..155592598) Chromosome 1 - NC_000067.6 Page 5 of 7 https://www.alphaknockout.com Transcript information: This gene has 1 transcript Gene: Rgs16 ENSMUSG00000026475 Description regulator of G-protein signaling 16 [Source:MGI Symbol;Acc:MGI:108407] Gene Synonyms Rgsr Location Chromosome 1: 153,740,349-153,745,468 forward strand. GRCm38:CM000994.2 About this gene This gene has 1 transcript (splice variant), 161 orthologues, 23 paralogues, is a member of 1 Ensembl protein family and is associated with 11 phenotypes. Transcripts Name Transcript ID bp Protein Translation ID Biotype CCDS UniProt Flags Rgs16-201 ENSMUST00000027748.7 2339 201aa ENSMUSP00000027748.7 Protein coding CCDS15376 P97428 Q542U0 TSL:1 GENCODE basic APPRIS P1 25.12 kb Forward strand 153.735Mb 153.740Mb 153.745Mb 153.750Mb 153.755Mb Genes (Comprehensive set... Rgs16-201 >protein coding Rnasel-201 >protein coding Rnasel-205 >protein coding Gm28512-201 >lncRNA Rnasel-206 >protein coding Rnasel-203 >retained intron Rnasel-204 >protein coding Contigs AC154140.7 > Genes < C230024C17Rik-201lncRNA < Gm28513-201lncRNA (Comprehensive set... Regulatory Build 153.735Mb 153.740Mb 153.745Mb 153.750Mb 153.755Mb Reverse strand 25.12 kb Regulation Legend CTCF Enhancer Promoter Promoter Flank Gene Legend Protein Coding Ensembl protein coding merged Ensembl/Havana Non-Protein Coding processed transcript RNA gene Page 6 of 7 https://www.alphaknockout.com Transcript: ENSMUST00000027748 5.12 kb Forward strand Rgs16-201 >protein coding ENSMUSP00000027... PDB-ENSP mappings MobiDB lite Low complexity (Seg) Superfamily RGS domain superfamily SMART RGS domain Prints RGS domain Pfam RGS domain PROSITE profiles RGS domain PANTHER PTHR10845:SF187 PTHR10845 Gene3D RGS, subdomain 1/3 1.10.167.10 CDD cd08710 All sequence SNPs/i... Sequence variants (dbSNP and all other sources) Variant Legend missense variant synonymous variant Scale bar 0 20 40 60 80 100 120 140 160 180 201 We wish to acknowledge the following valuable scientific information resources: Ensembl, MGI, NCBI, UCSC. Page 7 of 7.
Recommended publications
  • 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]
  • Related Malignant Phenotypes in the Nf1-Deficient MPNST
    Published OnlineFirst February 19, 2013; DOI: 10.1158/1541-7786.MCR-12-0593 Molecular Cancer Genomics Research RAS/MEK–Independent Gene Expression Reveals BMP2- Related Malignant Phenotypes in the Nf1-Deficient MPNST Daochun Sun1, Ramsi Haddad2,3, Janice M. Kraniak2, Steven D. Horne1, and Michael A. Tainsky1,2 Abstract Malignant peripheral nerve sheath tumor (MPNST) is a type of soft tissue sarcoma that occurs in carriers of germline mutations in Nf1 gene as well as sporadically. Neurofibromin, encoded by the Nf1 gene, functions as a GTPase-activating protein (GAP) whose mutation leads to activation of wt-RAS and mitogen-activated protein kinase (MAPK) signaling in neurofibromatosis type I (NF1) patients' tumors. However, therapeutic targeting of RAS and MAPK have had limited success in this disease. In this study, we modulated NRAS, mitogen-activated protein/extracellular signal–regulated kinase (MEK)1/2, and neurofibromin levels in MPNST cells and determined gene expression changes to evaluate the regulation of signaling pathways in MPNST cells. Gene expression changes due to neurofibromin modulation but independent of NRAS and MEK1/2 regulation in MPNST cells indicated bone morphogenetic protein 2 (Bmp2) signaling as a key pathway. The BMP2-SMAD1/5/8 pathway was activated in NF1-associated MPNST cells and inhibition of BMP2 signaling by LDN-193189 or short hairpin RNA (shRNA) to BMP2 decreased the motility and invasion of NF1-associated MPNST cells. The pathway-specific gene changes provide a greater understanding of the complex role of neurofibromin in MPNST pathology and novel targets for drug discovery. Mol Cancer Res; 11(6); 616–27.
    [Show full text]
  • Molecular Architecture of G O and the Structural Basis for RGS16
    Molecular architecture of G␣o and the structural basis for RGS16-mediated deactivation Kevin C. Slep*†, Michele A. Kercher‡§, Thomas Wieland¶ʈ, Ching-Kang Chen¶, Melvin I. Simon¶**, and Paul B. Sigler‡§†† *Department of Biology, University of North Carolina, Chapel Hill, NC 27599; ‡Department of Molecular Biophysics and Biochemistry, and §Howard Hughes Medical Institute, Yale University, New Haven, CT 06511; and ¶Division of Biology, California Institute of Technology, Mail Code 147-75, Pasadena, CA 91125 Contributed by Melvin I. Simon, February 16, 2008 (sent for review December 18, 2007) Heterotrimeric G proteins relay extracellular cues from heptahelical (11). With the number of RGS proteins greatly exceeding the transmembrane receptors to downstream effector molecules. number of G␣ subunits, RGS proteins are likely to be finely tuned, Composed of an ␣ subunit with intrinsic GTPase activity and a ␤␥ titrated, and localized to regulate specific signaling pathways within heterodimer, the trimeric complex dissociates upon receptor- aG␣ subunit’s repertoire of effector targets. To mediate the mediated nucleotide exchange on the ␣ subunit, enabling each specificity and fidelity requisite for accurate signal transmission, the component to engage downstream effector targets for either G␣ subunit and its binding partners (GPCR, G␤␥, RGS, and activation or inhibition as dictated in a particular pathway. To effector) must have specific reciprocating molecular determinants mitigate excessive effector engagement and concomitant signal to minimize the convergence of independent signaling pathways. transmission, the G␣ subunit’s intrinsic activation timer (the rate of Thus, a comprehensive understanding of the molecular basis for G␣ GTP hydrolysis) is regulated spatially and temporally by a class of engagement with its activators, regulators, and effectors is critical GTPase accelerating proteins (GAPs) known as the regulator of G for elucidating specificity determinants.
    [Show full text]
  • Identification of Transcriptional Mechanisms Downstream of Nf1 Gene Defeciency in Malignant Peripheral Nerve Sheath Tumors Daochun Sun Wayne State University
    Wayne State University DigitalCommons@WayneState Wayne State University Dissertations 1-1-2012 Identification of transcriptional mechanisms downstream of nf1 gene defeciency in malignant peripheral nerve sheath tumors Daochun Sun Wayne State University, Follow this and additional works at: http://digitalcommons.wayne.edu/oa_dissertations Recommended Citation Sun, Daochun, "Identification of transcriptional mechanisms downstream of nf1 gene defeciency in malignant peripheral nerve sheath tumors" (2012). Wayne State University Dissertations. Paper 558. This Open Access Dissertation is brought to you for free and open access by DigitalCommons@WayneState. It has been accepted for inclusion in Wayne State University Dissertations by an authorized administrator of DigitalCommons@WayneState. IDENTIFICATION OF TRANSCRIPTIONAL MECHANISMS DOWNSTREAM OF NF1 GENE DEFECIENCY IN MALIGNANT PERIPHERAL NERVE SHEATH TUMORS by DAOCHUN SUN DISSERTATION Submitted to the Graduate School of Wayne State University, Detroit, Michigan in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY 2012 MAJOR: MOLECULAR BIOLOGY AND GENETICS Approved by: _______________________________________ Advisor Date _______________________________________ _______________________________________ _______________________________________ © COPYRIGHT BY DAOCHUN SUN 2012 All Rights Reserved DEDICATION This work is dedicated to my parents and my wife Ze Zheng for their continuous support and understanding during the years of my education. I could not achieve my goal without them. ii ACKNOWLEDGMENTS I would like to express tremendous appreciation to my mentor, Dr. Michael Tainsky. His guidance and encouragement throughout this project made this dissertation come true. I would also like to thank my committee members, Dr. Raymond Mattingly and Dr. John Reiners Jr. for their sustained attention to this project during the monthly NF1 group meetings and committee meetings, Dr.
    [Show full text]
  • Supplementary Table 1
    Supplementary Table 1. 492 genes are unique to 0 h post-heat timepoint. The name, p-value, fold change, location and family of each gene are indicated. Genes were filtered for an absolute value log2 ration 1.5 and a significance value of p ≤ 0.05. Symbol p-value Log Gene Name Location Family Ratio ABCA13 1.87E-02 3.292 ATP-binding cassette, sub-family unknown transporter A (ABC1), member 13 ABCB1 1.93E-02 −1.819 ATP-binding cassette, sub-family Plasma transporter B (MDR/TAP), member 1 Membrane ABCC3 2.83E-02 2.016 ATP-binding cassette, sub-family Plasma transporter C (CFTR/MRP), member 3 Membrane ABHD6 7.79E-03 −2.717 abhydrolase domain containing 6 Cytoplasm enzyme ACAT1 4.10E-02 3.009 acetyl-CoA acetyltransferase 1 Cytoplasm enzyme ACBD4 2.66E-03 1.722 acyl-CoA binding domain unknown other containing 4 ACSL5 1.86E-02 −2.876 acyl-CoA synthetase long-chain Cytoplasm enzyme family member 5 ADAM23 3.33E-02 −3.008 ADAM metallopeptidase domain Plasma peptidase 23 Membrane ADAM29 5.58E-03 3.463 ADAM metallopeptidase domain Plasma peptidase 29 Membrane ADAMTS17 2.67E-04 3.051 ADAM metallopeptidase with Extracellular other thrombospondin type 1 motif, 17 Space ADCYAP1R1 1.20E-02 1.848 adenylate cyclase activating Plasma G-protein polypeptide 1 (pituitary) receptor Membrane coupled type I receptor ADH6 (includes 4.02E-02 −1.845 alcohol dehydrogenase 6 (class Cytoplasm enzyme EG:130) V) AHSA2 1.54E-04 −1.6 AHA1, activator of heat shock unknown other 90kDa protein ATPase homolog 2 (yeast) AK5 3.32E-02 1.658 adenylate kinase 5 Cytoplasm kinase AK7
    [Show full text]
  • Egfr Activates a Taz-Driven Oncogenic Program in Glioblastoma
    EGFR ACTIVATES A TAZ-DRIVEN ONCOGENIC PROGRAM IN GLIOBLASTOMA by Minling Gao A thesis submitted to Johns Hopkins University in conformity with the requirements for the degree of Doctor of Philosophy Baltimore, Maryland March 2020 ©2020 Minling Gao All rights reserved Abstract Hyperactivated EGFR signaling is associated with about 45% of Glioblastoma (GBM), the most aggressive and lethal primary brain tumor in humans. However, the oncogenic transcriptional events driven by EGFR are still incompletely understood. We studied the role of the transcription factor TAZ to better understand master transcriptional regulators in mediating the EGFR signaling pathway in GBM. The transcriptional coactivator with PDZ- binding motif (TAZ) and its paralog gene, the Yes-associated protein (YAP) are two transcriptional co-activators that play important roles in multiple cancer types and are regulated in a context-dependent manner by various upstream signaling pathways, e.g. the Hippo, WNT and GPCR signaling. In GBM cells, TAZ functions as an oncogene that drives mesenchymal transition and radioresistance. This thesis intends to broaden our understanding of EGFR signaling and TAZ regulation in GBM. In patient-derived GBM cell models, EGF induced TAZ and its known gene targets through EGFR and downstream tyrosine kinases (ERK1/2 and STAT3). In GBM cells with EGFRvIII, an EGF-independent and constitutively active mutation, TAZ showed EGF- independent hyperactivation when compared to EGFRvIII-negative cells. These results revealed a novel EGFR-TAZ signaling axis in GBM cells. The second contribution of this thesis is that we performed next-generation sequencing to establish the first genome-wide map of EGF-induced TAZ target genes.
    [Show full text]
  • The Human Gene Connectome As a Map of Short Cuts for Morbid Allele Discovery
    The human gene connectome as a map of short cuts for morbid allele discovery Yuval Itana,1, Shen-Ying Zhanga,b, Guillaume Vogta,b, Avinash Abhyankara, Melina Hermana, Patrick Nitschkec, Dror Friedd, Lluis Quintana-Murcie, Laurent Abela,b, and Jean-Laurent Casanovaa,b,f aSt. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065; bLaboratory of Human Genetics of Infectious Diseases, Necker Branch, Paris Descartes University, Institut National de la Santé et de la Recherche Médicale U980, Necker Medical School, 75015 Paris, France; cPlateforme Bioinformatique, Université Paris Descartes, 75116 Paris, France; dDepartment of Computer Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; eUnit of Human Evolutionary Genetics, Centre National de la Recherche Scientifique, Unité de Recherche Associée 3012, Institut Pasteur, F-75015 Paris, France; and fPediatric Immunology-Hematology Unit, Necker Hospital for Sick Children, 75015 Paris, France Edited* by Bruce Beutler, University of Texas Southwestern Medical Center, Dallas, TX, and approved February 15, 2013 (received for review October 19, 2012) High-throughput genomic data reveal thousands of gene variants to detect a single mutated gene, with the other polymorphic genes per patient, and it is often difficult to determine which of these being of less interest. This goes some way to explaining why, variants underlies disease in a given individual. However, at the despite the abundance of NGS data, the discovery of disease- population level, there may be some degree of phenotypic homo- causing alleles from such data remains somewhat limited. geneity, with alterations of specific physiological pathways under- We developed the human gene connectome (HGC) to over- come this problem.
    [Show full text]
  • The Human Regulator of G-Protein Signaling Protein 6 Gene (RGS6) Maps Between Markers WI-5202 and D14S277 on Chromosome 14Q24.3
    138J Hum Genet (1999) 44:138–140 © Jpn Soc Hum Genet and Springer-Verlag 1999 BRIEF REPORT—GENE MAPPING Naohiko Seki · Atsushi Hattori · Akiko Hayashi Sumie Kozuma · Tada-aki Hori · Toshiyuki Saito The human regulator of G-protein signaling protein 6 gene (RGS6) maps between markers WI-5202 and D14S277 on chromosome 14q24.3 Received: September 14, 1998 / Accepted: November 1, 1998 Abstract The recently discovered regulators of G-protein proteins by activating the intrinsic GTPase activity of the α signaling proteins, termed the RGS family, have been subunits (for reviews, see Dohlman and Thorner 1997; shown to modulate the functioning of G-proteins by activat- Koelle 1997; Neer 1997). ing the intrinsic guanosine triphosphatase (GTPase) activity At present, full or partial sequences of the mRNAs for of the α subunits. Here, we report the chromosomal loca- numerous putative RGS proteins have been identified in tion and tissue expression of the human regulator of RGS6 mammalian species (De Vries et al. 1995; Chen et al. 1996; gene. The messenger RNA was ubiquitously expressed in Druey et al. 1996; Koelle and Horvitz, 1996; Siderovski et al. various tissues. Polymerase chain reaction (PCR)-based 1996; Gold et al. 1997; Snow et al. 1997; 1998; Seki et al. analysis with a human/rodent monochromosomal hybrid 1998a). The cDNA sequence of human RGS6 has recently panel and a radiation hybrid panel indicated that the gene been registered with the public database (accession number was mapped between genetic markers WI-5202 and AF073920). We are attempting to carry out systematic chro- D14S277 on chromosome 14q24.3 region.
    [Show full text]
  • Hedgehog Inhibitor Sonidegib Potentiates 177Lu-Octreotate
    Spetz et al. BMC Cancer (2017) 17:528 DOI 10.1186/s12885-017-3524-x RESEARCH ARTICLE Open Access Hedgehog inhibitor sonidegib potentiates 177Lu-octreotate therapy of GOT1 human small intestine neuroendocrine tumors in nude mice Johan Spetz1* , Britta Langen1, Nils Rudqvist1, Toshima Z. Parris2, Khalil Helou2, Ola Nilsson3 and Eva Forssell-Aronsson1 Abstract Background: 177Lu-octreotate can be used to treat somatostatin receptor expressing neuroendocrine tumors. It is highly effective in animal models, but clinical studies have so far only demonstrated low cure rates. Hedgehog inhibitors have shown therapeutic effect as monotherapy in neuroendocrine tumor model systems and might be one option to enhance the efficacy of 177Lu-octreotate therapy. The aim of this study was to determine the therapeutic effect of combination therapy using 177Lu-octreotate and the Hedgehog signaling pathway inhibitor sonidegib. Methods: GOT1-bearing BALB/c nude mice were treated with either sonidegib (80 mg/kg twice a week via oral gavage), a single injection of 30 MBq 177Lu-octreotate i.v., or a combination of both. Untreated animals served as controls. Tumor size was measured twice-weekly using calipers. The animals were killed 41 d after injection followed by excision of the tumors. Total RNA was extracted from each tumor sample and then subjected to gene expression analysis. Gene expression patterns were compared with those of untreated controls using Nexus Expression 3.0, IPA and Gene Ontology terms. Western blot was carried out on total protein extracted from the tumor samples to analyze activation-states of the Hh and PI3K/AKT/mTOR pathways. Results: Sonidegib monotherapy resulted in inhibition of tumor growth, while a significant reduction in mean tumor volume was observed after 177Lu-octreotate monotherapy and combination therapy.
    [Show full text]
  • De-Repression of the RAC Activator ELMO1 in Cancer Stem Cells
    RESEARCH ARTICLE De-repression of the RAC activator ELMO1 in cancer stem cells drives progression of TGFb-deficient squamous cell carcinoma from transition zones Heather A McCauley1*, Ve´ ronique Chevrier2, Daniel Birnbaum2, Ge´ raldine Guasch1,2* 1Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, United States; 2Centre de Recherche en Cance´rologie de Marseille (CRCM), Inserm, U1068, F-13009, CNRS, UMR7258, F-13009, Institut Paoli- Calmettes, F-13009, Aix-Marseille University, UM 105, F-13284, Marseille, France Abstract Squamous cell carcinomas occurring at transition zones are highly malignant tumors with poor prognosis. The identity of the cell population and the signaling pathways involved in the progression of transition zone squamous cell carcinoma are poorly understood, hence representing limited options for targeted therapies. Here, we identify a highly tumorigenic cancer stem cell population in a mouse model of transitional epithelial carcinoma and uncover a novel mechanism by which loss of TGFb receptor II (Tgfbr2) mediates invasion and metastasis through de-repression of ELMO1, a RAC-activating guanine exchange factor, specifically in cancer stem cells of transition zone tumors. We identify ELMO1 as a novel target of TGFb signaling and show that restoration of Tgfbr2 results in a complete block of ELMO1 in vivo. Knocking down Elmo1 impairs metastasis of carcinoma cells to the lung, thereby providing insights into the mechanisms of progression of *For correspondence: Heather. Tgfbr2-deficient invasive transition zone squamous cell carcinoma. [email protected] (HAM); DOI: 10.7554/eLife.22914.001 geraldine.guasch-grangeon@ inserm.fr (GG) Competing interests: The authors declare that no Introduction competing interests exist.
    [Show full text]
  • A Major Qualifying Project Report Submitted to The
    A MAJOR QUALIFYING PROJECT REPORT SUBMITTED TO THE FACULTY OF THE WORCESTER POLYTECHNIC INSTITUTE IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF BACHELOR OF SCIENCE BY DONGNI ZHANG, MA DATE: NOVEMBER 10TH, 2014 SPONSORED BY: WORCESTER POLYTECHNIC INSTITUTE PROJECT ADVISOR: PROFESSOR ZHEYANG WU Table of Contents ChaptEr 1: Introduction ........................................................................................................................................... 3 ChaptEr 2: MEthodology .......................................................................................................................................... 4 ResEarch design ................................................................................................................................................... 4 Seed gene detection ............................................................................................................................................ 5 CandidatE gEne detEction .................................................................................................................................... 5 AnnotatE nsSNVs according to protEin binding sitEs ........................................................................................... 6 Likelihood ratio tEsts (LRT) .................................................................................................................................. 6 LRT for burdEn scorE wEightEd by binding sitE information (LRT-BS) ................................................................
    [Show full text]
  • The Genetic Basis of Intradural Spinal Tumors and Its Impact on Clinical Treatment
    NEUROSURGICAL FOCUS Neurosurg Focus 39 (2):E3, 2015 The genetic basis of intradural spinal tumors and its impact on clinical treatment Michael Karsy, MD, PhD, Jian Guan, MD, Walavan Sivakumar, MD, Jayson A. Neil, MD, Meic H. Schmidt, MD, MBA, and Mark A. Mahan, MD Department of Neurosurgery, Clinical Neurosciences Center, The University of Utah, Salt Lake City, Utah Genetic alterations in the cells of intradural spinal tumors can have a significant impact on the treatment options, coun- seling, and prognosis for patients. Although surgery is the primary therapy for most intradural tumors, radiochemothera- peutic modalities and targeted interventions play an ever-evolving role in treating aggressive cancers and in addressing cancer recurrence in long-term survivors. Recent studies have helped delineate specific genetic and molecular differ- ences between intradural spinal tumors and their intracranial counterparts and have also identified significant variation in therapeutic effects on these tumors. This review discusses the genetic and molecular alterations in the most common intradural spinal tumors in both adult and pediatric patients, including nerve sheath tumors (that is, neurofibroma and schwannoma), meningioma, ependymoma, astrocytoma (that is, low-grade glioma, anaplastic astrocytoma, and glioblas- toma), hemangioblastoma, and medulloblastoma. It also examines the genetics of metastatic tumors to the spinal cord, arising either from the CNS or from systemic sources. Importantly, the impact of this knowledge on therapeutic options
    [Show full text]