Ribonuclease P: Anenzyme with an Essential Rnacomponent

Total Page:16

File Type:pdf, Size:1020Kb

Ribonuclease P: Anenzyme with an Essential Rnacomponent Proc. Natl. Acad. Sci. USA Vol. 75, No. 8, pp. 3717-3721, August 1978 Biochemistry Ribonuclease P: An enzyme with an essential RNA component (endoribonuclease/precursor tRNA substrates/RNA subunit) BENJAMIN C. STARK*, RYSZARD KOLEt, EMMA J. BOWMAN*, AND SIDNEY ALTMAN§ Department of Biology, Yale University, New Haven, Connecticut 06520 Communicated by Joseph G. Gall, June 8,1978 ABSTRACT The activity of ribonuclease P on precursor same buffer. The RNase P eluted after the rRNA peak and tRNA substrates from Escherichia coli can be abolished by before the 4S RNA peak. The active fractions were pooled and pretreatment of this enzyme with micrococcal nuclease or concentrated as described above and then applied to a Sephadex pancreatic ribonuclease A, as well as by proteases and by ther- mal denaturation. Highly purified RNase P exhibits one prom- G-200 column (1 X 90 cm) equilibrated and eluted in the same inent RNA and one prominent polypeptide com nent when manner as the Sepharose 4B column. The RNase P eluted just examined in polyacrylamide gels containing sodum dodecyl after the void volume. This material was again pooled, con- sulfate. The buoyant density in CsCl of RNase P, 1.71 g/ml, is centrated, redissolved in 2 M (NH4)2SO4 in buffer B, applied characteristic of a protein-RNA complex. The activity of RNase to an n-octyl-Sepharose (a gift of H. Taira, Yale University, P is inhibited by various RNA molecules. The presence of a New Haven, CT) column (0.5 X 2 cm) that had been equili- discrete RNA component in RNase P appears to be essential for brated with 2 M in buffer and then eluted with enzymatic function. A model is described for enzyme-substrate (NH4)2SO4 B, recognition in which this RNA component plays an important a linear gradient from 2 M (NH4)2SO4 in buffer B to buffer B. role. The active fractions eluted at about 0.5 M (NH4)2SO4. Scheme II. S30 made with 600 g of E. coli MRE 600 was Ribonuclease P (1-3) is necessary for the biosynthesis of the 5' further processed to yield a 0.2 M NH4Cl wash of ribosomes. termini of tRNA molecules in Escherichia coli. This enzyme The active RNase P in the salt wash was loaded onto a DEAE- recognizes some aspect of the structural conformation rather Sephadex column and purified as described (2). RNase P (1.8 than nucleotide sequence at is cleavage sites in tRNA precursor mg), purified through the DEAE-Sephadex step, was concen- molecules (4-6). Exactly how this recognition occurs is a matter trated by precipitation with (NH4)2SO4, resuspended in 0.5 ml for speculation (7). In this report we show that treatment of of buffer G [20 mM Tris-HCl, pH 7.6/0.50 M NH4CI/15 mM highly purified RNase P with either micrococcal nuclease (MN) Mg(OAc)2/6 mM 2-mercaptoethanol/5% sucrose], dialyzed or pancreatic ribonuclease A abolishes RNase P activity. Our against this buffer, and applied to a column of Sephadex G-200 most highly purified RNase P preparations contain one discrete (0.9 X 55 cm) equilibrated with buffer G. RNase P activity was RNA species and one discrete protein species, and several kinds eluted from this column with buffer G. One-third of the peak of RNA can inhibit the enzymatic activity. We conclude that fractions (0.4 mg of protein in 4 ml) was pooled and concen- the interaction of RNase P with its substrates depends on the trated by dialysis against buffer G containing 15% polyethylene presence of RNA in the enzyme complex. glycol until the final volume was 0.3 ml. This sample was loaded in aliquots onto four 7% polyacrylamide gel columns (6 X 90 METHODS mm). The running buffer was 25 mM Tris-HCl/25 mM glycine Preparation of RNase P. What follows is an abbreviated at pH 9; the gel buffer was the same but at pH 7. The gels were description of RNase P purification schemes. Complete details run at 4 mA per column for 80 min at 40, at which time the will be published elsewhere. Crude extracts (S30) of E. coil bromphenol blue marker had run about 70 mm. Ten slices, MRE 600 were prepared by grinding of frozen cells with alu- spaced equidistant apart, were made from the top of the gel to mina as described (2). Two purification schemes were used, the dye front and eluted, after mincing, with buffer G at 00. starting with S30. Fractions were assayed for activity as described (2, 5). The final Scheme I. S30 made from 200 g of cells was diluted 1:5 with NH4Cl concentration in the reaction mixtures was 10 mM. buffer A [50 mM Tris-HCl, pH 7.5/60 mM NH4Cl/10 mM Assay of RNase P Activity. Radioactive substrates for RNase Mg(OAc)2/6 mM 2-mercaptoethanol] and loaded onto a P were prepared from E. coli A49 (RNase P ts) as described (2, DEAE-Sephadex A-50 column (12 X 25 cm) equilibrated with 5). These were used in assay mixtures under standard conditions buffer A. Bed volume of the column was about 2.5 liters. The (2, 5) unless otherwise noted. The extent of RNase P reaction column was washed successively with 3 liters of buffer A con- was quantitated by cutting appropriate slices out of polyac- taining 0.2 M NH4Cl, 0.3 M NH4Cl, and 0.4 M NH4Cl in suc- rylamide gels and assaying the amount of RNA in the slices by cessive washes. The final wash was with buffer A containing monitoring Cerenkov radiation. 0.5 M NH4Cl (now called buffer B) and the activity was eluted Abbreviations: NaDodSO4, sodium dodecyl sulfate; MN, micrococcal after about 1 liter of the last wash buffer had flowed through nuclease; buffer A, 50 mM Tris-HCl, pH 7.5/60 mM NH4Cl/10 mM the column. The pooled active fractions wer- precipitated with Mg(OAc)2/6 mM 2-mercaptoethanol; buffer B, 50 mM Tris-HCl, pH 0.6 g of (NH4)2SO4 per ml; the precipitate was resuspended and 7.5/0.5 M NH4Cl/10 mm Mg(OAc)2/6 mM 2-mercaptoethanol; dialyzed against buffer B to give a final volume of about 15 ml. buffer C, 20 mM Tris-HCl, pH 7.6/0.50 mM NH4Cl/15 mM This material was applied to a Sepharose 4B column (2.5 X 90 Mg(OAc)2/6 mM 2-mercaptoethanol/5% sucrose. * Present address: Department of Botany, Washington State University, cm) equilibrated with buffer B and eluted with 300 ml of the Pullman, WA 99163. t Permanent address: Institute of Biochemistry and Biophysics, The costs of publication of this article were defrayed in part by the Polish Academy of Sciences, Warsaw, Poland. payment of page charges. This article must therefore be hereby marked * Present address: Department of Human Genetics, Yale University, "advertisement" in accordance with 18 U.S.C. §1734 solely to indicate School of Medicine, New Haven, CT 06510. this fact. § To whom reprint requests should be addressed. 3717 Downloaded by guest on September 23, 2021 3718 Biochemistry: Stark et al. Proc. Natl. Acad. Sci. USA 75 (1978) Ribonuclease Inactivation of RNase P. One hundred mi- Ml of buffer G was then added and the sample was filtered croliters of RNase P (11 tig of protein purified through the through a Swinnex filter apparatus fitted with a Millipore DEAE-Sephadex step of scheme II was used in the experiments GAWP 01300 filter (0.45 Mum pore size) to remove the polyac- shown in Fig. 2A; 4 ug of protein purified through the nonde- rylamide beads and the pancreatic RNase A. This prevented naturing gel step of scheme II was used in the experiments interference of the pancreatic RNase A in subsequent assays shown in Fig. 2B) in buffer G was mixed with 50 Ml of 10 mM of the RNase P sample. Control reaction mixtures contained 50 CaCl2 and 6 ,ul of MN (1 mg/ml) in 1 mM NaCi. (MN from MAl of buffer G instead of the pancreatic RNase A suspension; Sigma Biochemicals, Worthington Biochemicals, Schwarz/ they were incubated and filtered in parallel with the actual Mann, and an authentic sample received as a kind gift from M. reaction mixtures in all experiments. Laskowski, Sr., were all equally effective. Whether or not the stock solutions were made up with bovine serum albumin had RESULTS no effect on the reaction.) The mixture was incubated for 30 min at 370 and then dialyzed extensively against buffer G at Structure of Highly Purified RNase P. RNase P. purified 40 to remove Ca2 . The RNase P was then assayed for activity by either scheme I or scheme II, exhibited a simple pattern using the precursor to E. coil tRNATYr as substrate. The final when analyzed in sodium dodecyl sulfate (NaDodSO4)/poly- volume (100,Ml) contained 30 mM Tris-HCI (pH 8), 5 mM acrylamide gels (Fig. 1). Scheme I enzyme, purified from a MgCI2, 0.1 mM EDTA, 0.1 mM 2-mercaptoethanol, and about crude extract of E. coli, showed one prominent band staining 104 cpm of substrate with appropriate amounts of treated en- with Coomassie brilliant blue (C5) and one staining with zyme. The final concentration of NH4Cl in the assay mixtures methylene blue (M2), protein and nucleic acid stains, respec- was about 50 mM. Incubation was for 10 min at 370; the mix- tively (Fig. 1A). Estimates of the purification factor (at least tures were then analyzed in acrylamide gels as described (2, 500-fold) of this enzyme were complicated by losses of activity 5).
Recommended publications
  • Supporting Information
    Supporting Information Figure S1. The functionality of the tagged Arp6 and Swr1 was confirmed by monitoring cell growth and sensitivity to hydeoxyurea (HU). Five-fold serial dilutions of each strain were plated on YPD with or without 50 mM HU and incubated at 30°C or 37°C for 3 days. Figure S2. Localization of Arp6 and Swr1 on chromosome 3. The binding of Arp6-FLAG (top), Swr1-FLAG (middle), and Arp6-FLAG in swr1 cells (bottom) are compared. The position of Tel 3L, Tel 3R, CEN3, and the RP gene are shown under the panels. Figure S3. Localization of Arp6 and Swr1 on chromosome 4. The binding of Arp6-FLAG (top), Swr1-FLAG (middle), and Arp6-FLAG in swr1 cells (bottom) in the whole chromosome region are compared. The position of Tel 4L, Tel 4R, CEN4, SWR1, and RP genes are shown under the panels. Figure S4. Localization of Arp6 and Swr1 on the region including the SWR1 gene of chromosome 4. The binding of Arp6- FLAG (top), Swr1-FLAG (middle), and Arp6-FLAG in swr1 cells (bottom) are compared. The position and orientation of the SWR1 gene is shown. Figure S5. Localization of Arp6 and Swr1 on chromosome 5. The binding of Arp6-FLAG (top), Swr1-FLAG (middle), and Arp6-FLAG in swr1 cells (bottom) are compared. The position of Tel 5L, Tel 5R, CEN5, and the RP genes are shown under the panels. Figure S6. Preferential localization of Arp6 and Swr1 in the 5′ end of genes. Vertical bars represent the binding ratio of proteins in each locus.
    [Show full text]
  • Enhanced Phosphorylation of High-Mobility-Group Proteins In
    Proc. Nati. Acad. Sci. USA Vol. 78, No. 4, pp. 2189-2193, April 1981 Biochemistry Enhanced phosphorylation of high-mobility-group proteins in nuclease-sensitive mononucleosomes from butyrate-treated HeLa cells (hyperphosphorylation/transcriptionally active chromatin/sodium butyrate) BEATRIZ LEVY-WILSON Department of Molecular Biology and Biochemistry, University of California, Irvine, California 92717 Communicated by James F. Bonner, December 29, 1980 ABSTRACT The protein composition of nucleosome fractions MATERIALS AND METHODS differing in their sensitivity to micrococcal nuclease and derived from butyrate-treated or untreated HeLa cells has been com- Cell Cultures. HeLa cells, line S3, were grown in monolay- pared. Most of the high-mobility-group-14 (HMG-14) and HMG- ers, either in T-150 culture flasks or in roller bottles, at 370C 17 content of HeLa cell chromatin is associated with those nu- in minimal essential medium (Flow Laboratories, Rockville, cleosomes that are preferentially sensitive to micrococcal nu- MD) supplemented with 7% bovine calfserum (KC Biologicals) clease. Furthermore, electrophoresis of these two HMG proteins and penicillin (100 units/ml) and streptomycin (100 Ag/ml) from the transcriptionally active chromatin fraction MN, of bu- (Sigma). In experiments involving sodium butyrate, this com- tyrate-treated cells resolves them into a series ofbands. The mul- pound was added to the cultures at a final concentration of5 mM tiple band pattern of HMG-14 and -17 from butyrate-treated cells about 20 hr prior to harvesting of the cells. results from hyperphosphorylation rather than hyperacetylation. Preparation of Nuclei. HeLa cell nuclei were prepared as Phosphorylation of these two small nonhistone proteins may play described by Milcarek et al.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Ribonuclease P
    14 Ribonuclease P Sidney Altman Department of Molecular, Cellular and Developmental Biology Yale University New Haven, Connecticut 06520 Leif Kirsebom Department of Microbiology Biomedical Center Uppsala University Uppsala S-751 23, Sweden RNase P was first characterized in Escherichia coli as an enzyme that is required for the processing of the 5؅ termini of tRNA in the pathway of biosynthesis of tRNA from precursor tRNAs (ptRNAs; Altman and Smith 1971; for review, see Altman 1989). Subsequently, it was discov- ered that this enzyme consisted of one protein and one RNA subunit, the latter being the catalytic subunit (Guerrier-Takada et al. 1983). In retro- spect, it should not be surprising that an enzyme with the chemical compo- sition of RNase P exists. Ribosomes are also ribonucleoproteins (RNPs), but they are much more complicated than RNase P. It seems unreasonable that ribosomes, with three RNAs and about 50 proteins, would exist with- out less complex RNPs having come into existence first and having per- sisted throughout evolution. Indeed, relatively simple RNPs with and without catalytic activity have been discovered with regularity during the past 20 years. What important questions about RNase P have not yet been an- swered? Certainly, for the biochemist, problems not yet solved for E. coli RNase P include a full understanding of enzyme–substrate interac- tions, RNA–protein subunit interactions, and the chemical details of the hydrolysis reaction. In every case, much progress has been made, but much has yet to be learned. A second issue of interest to biochemists and “evolutionists” is the puzzle presented by the different compositions of RNase P from eubac- teria (one catalytic RNA and one protein subunit) and from eukaryotes (one RNA subunit and several protein subunits: no understanding yet of which subunit is responsible for catalysis).
    [Show full text]
  • Catalybc Rnas
    Felix Loosli ITG/KIT (Karlsruhe) Developmental neurobiology of vertebrates Introduc*on to Gene*cs Cataly@c RNAs Lewin‘s GeneX Chapter 23 Albert‘s Molecular Biology of the Cell 5th ed. Chapter 6 Ribozyme : a RNA that has catalytic activity Group I and II self splicing introns Ribonuclease P (RNase P: t-RNA maturaon) Viroids Ribosome Evolu@onary consideraons Tetrahymena thermophilus Thomas R. Cech rRNA self-splicing Splicing occurred without added cell extract Sidney Altman Cataly@c RNA of RNase P which contains a RNA molecule essen@al for catalysis rRNA genes Mitochondrial/Chloroplast genes Bound G nucleo@de (cofactor) aacks phosphodiester bond Two consecuve transesterificaons; no ATP required 2nd structure of RNA is essen@al promoter β-gal β-gal tetrahymena intron transfect E. coli transcrip@on and splicing blue colonies on agar plate GTP conc. >rRNA conc. change of 2nd structure of products (products are ‘removed’ from the reac@on) -> reac@on proceeds to comple@on In vitro self splicing is very slow, in vivo proteins have accessory funcons Group I introns share a common 2nd structure: core structure is the ac@ve site Group I introns can also catalyse: Sequence specific RNA cleavage (ribo endonuclease) RNA ligase Phospatase (not related to self-splicing ac@vity) Group II Introns May Code for Multifunction Proteins • Group II introns can autosplice in vitro, but are usually assisted by protein activities encoded by the intron • A single open reading frame codes for a protein with reverse transcriptase activity, maturase activity, a DNA-binding
    [Show full text]
  • Termin Translat Trna Utr Mutat Protein Signal
    Drugs & Chemicals 1: Tumor Suppressor Protein p53 2: Heterogeneous-Nuclear Ribonucleo- (1029) proteins (14) activ apoptosi arf cell express function inactiv induc altern assai associ bind mdm2 mutat p53 p73 pathwai protein regul complex detect exon famili genom respons suppress suppressor tumor wild-typ interact intron isoform nuclear protein sensit site specif splice suggest variant 3: RNA, Transfer (110) 4: DNA Primers (1987) codon contain differ eukaryot gene initi amplifi analysi chain clone detect dna express mrna protein region ribosom rna fragment gene genotyp mutat pcr sequenc site speci suggest synthesi polymorph popul primer reaction region restrict sequenc speci termin translat trna utr 5: Saccharomyces cerevisiae Proteins 6: Apoptosis Regulatory Proteins (291) (733) activ apoptosi apoptosis-induc albican bud candida cerevisia complex encod apoptot bcl-2 caspas caspase-8 cell eukaryot fission function growth interact involv death fasl induc induct ligand methyl necrosi pathwai program sensit surviv trail mutant pomb protein requir saccharomyc strain suggest yeast 7: Plant Proteins (414) 8: Membrane Proteins (1608) access arabidopsi cultivar flower hybrid leaf leav apoptosi cell conserv domain express function gene human identifi inhibitor line maiz plant pollen rice root seed mammalian membran mice mous mutant seedl speci thaliana tomato transgen wheat mutat protein signal suggest transport 1 9: Tumor Suppressor Proteins (815) 10: 1-Phosphatidylinositol 3-Kinase activ arrest cell cycl cyclin damag delet dna (441) 3-kinas activ
    [Show full text]
  • Detection of a Novel Avian Influenza a (H7N9)
    Fan et al. BMC Infectious Diseases 2014, 14:541 http://www.biomedcentral.com/1471-2334/14/541 RESEARCH ARTICLE Open Access Detection of a novel avian influenza A (H7N9) virus in humans by multiplex one-step real-time RT-PCR assay Jian Fan1†, David Cui1†, Siuying Lau3, Guoliang Xie1, Xichao Guo1, Shufa Zheng1, Xiaofeng Huang3, Shigui Yang2, Xianzhi Yang1, Zhaoxia Huo1, Fei Yu1, Jianzhou Lou1, Li Tian1, Xuefen Li1, Yuejiao Dong1, Qiaoyun Zhu1 and Yu Chen1,2* Abstract Background: A novel avian influenza A (H7N9) virus emerged in eastern China in February 2013. 413 confirmed human cases, including 157 deaths, have been recorded as of July 31, 2014. Methods: Clinical specimens, including throat swabs, sputum or tracheal aspirates, etc., were obtained from patients exhibiting influenza-like illness (ILIs), especially from those having pneumonia and a history of occupational exposure to poultry and wild birds. RNA was extracted from these samples and a multiplex one-step real-time RT-PCR assay was developed to specifically detect the influenza A virus (FluA). PCR primers targeted the conserved M and Rnase P (RP) genes, as well as the hemagglutinin and neuraminidase genes of the H7N9 virus. Results: The multiplex assay specifically detected the avian H7N9 virus, and no cross-reaction with other common respiratory pathogens was observed. The detection limit of the assay was approximately 0.05 50% tissue culture infective doses (TCID50), or 100 copies per reaction. Positive detection of the H7N9 virus in sputum/tracheal aspirates was higher than in throat swabs during the surveillance of patients with ILIs. Additionally, detection of the matrix (M) and Rnase P genes aided in the determination of the novel avian H7N9 virus and ensured the quality of the clinical samples.
    [Show full text]
  • Nucleosome Phasing and Micrococcal Nuclease Cleavage of African Green Monkey Component a DNA (Nucleosomes and Chromatdn/Primate Dnas/DNA Sequence) PHILLIP R
    Proc. Natl Acad. Sci. USA Vol. 79, pp. 118-122, January 1982 Cell Biology Nucleosome phasing and micrococcal nuclease cleavage of African green monkey component a DNA (nucleosomes and chromatdn/primate DNAs/DNA sequence) PHILLIP R. MUSICH*, FRED L. BROWN, AND JOSEPH J. MAIO Department of Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461 Communicated by Harry Eagle, September 14, 1981 ABSTRACT The micrococcal nuclease cleavage of intact nu- gestion was made that these results simply reflect a DNA se- clear chromatin from African green monkey cells and ofthe com- quence specificity of the micrococcal nuclease. Nucleosome pletely deproteinized sequences was studied by using high-reso- phasing would not have to be invoked. Although phase rela- lution analytical and DNA sequencing gels and secondary restric- tionships have been observed for other repetitive and 5S DNA tion enzyme analysis. When deproteinized component a DNAwas (5, 10), some tRNA coding regions (11), gene domains (12), and used as substrate, not all phosphodiester bonds in the 172-base- regions of the simian virus 40 genome (13, 14), these results pair repeat units were cleaved with equal frequency by the nu- would also now be in doubt if micrococcal nuclease shows site clease. A distinct preference for the cleavage of A-T rather than specificity with deproteinized DNA that could give rise to spu- G-C bonds was observed: however, A+T-richness in itselfdid not rious phase relationships in chromatin. confer susceptibility to cleavage by micrococcal nuclease. The re- We performed detailed sequence analysis ofthe micrococcal sults suggested that, in deproteinized DNA, nuclease cleavage at a particular dinucleotides may be influenced more by the effect of nuclease products of completely deproteinized component adjacent sequences than by the composition of the dinucleotide.
    [Show full text]
  • Human Induced Pluripotent Stem Cell–Derived Podocytes Mature Into Vascularized Glomeruli Upon Experimental Transplantation
    BASIC RESEARCH www.jasn.org Human Induced Pluripotent Stem Cell–Derived Podocytes Mature into Vascularized Glomeruli upon Experimental Transplantation † Sazia Sharmin,* Atsuhiro Taguchi,* Yusuke Kaku,* Yasuhiro Yoshimura,* Tomoko Ohmori,* ‡ † ‡ Tetsushi Sakuma, Masashi Mukoyama, Takashi Yamamoto, Hidetake Kurihara,§ and | Ryuichi Nishinakamura* *Department of Kidney Development, Institute of Molecular Embryology and Genetics, and †Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; ‡Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan; §Division of Anatomy, Juntendo University School of Medicine, Tokyo, Japan; and |Japan Science and Technology Agency, CREST, Kumamoto, Japan ABSTRACT Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator–like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in
    [Show full text]
  • A Role of Human Rnase P Subunits, Rpp29 and Rpp21, in Homology
    www.nature.com/scientificreports OPEN A role of human RNase P subunits, Rpp29 and Rpp21, in homology directed-repair of double-strand Received: 22 December 2016 Accepted: 22 March 2017 breaks Published: xx xx xxxx Enas R. Abu-Zhayia1, Hanan Khoury-Haddad1, Noga Guttmann-Raviv1, Raphael Serruya2, Nayef Jarrous2 & Nabieh Ayoub1 DNA damage response (DDR) is needed to repair damaged DNA for genomic integrity preservation. Defective DDR causes accumulation of deleterious mutations and DNA lesions that can lead to genomic instabilities and carcinogenesis. Identifying new players in the DDR, therefore, is essential to advance the understanding of the molecular mechanisms by which cells keep their genetic material intact. Here, we show that the core protein subunits Rpp29 and Rpp21 of human RNase P complex are implicated in DDR. We demonstrate that Rpp29 and Rpp21 depletion impairs double-strand break (DSB) repair by homology-directed repair (HDR), but has no deleterious effect on the integrity of non-homologous end joining. We also demonstrate that Rpp29 and Rpp21, but not Rpp14, Rpp25 and Rpp38, are rapidly and transiently recruited to laser-microirradiated sites. Rpp29 and Rpp21 bind poly ADP-ribose moieties and are recruited to DNA damage sites in a PARP1-dependent manner. Remarkably, depletion of the catalytic H1 RNA subunit diminishes their recruitment to laser-microirradiated regions. Moreover, RNase P activity is augmented after DNA damage in a PARP1-dependent manner. Altogether, our results describe a previously unrecognized function of the RNase P subunits, Rpp29 and Rpp21, in fine- tuning HDR of DSBs. The human genome is susceptible to endogenous and exogenous DNA damaging agents1, 2.
    [Show full text]
  • Mechanisms of Interaction Between Haemophilus Parainfluenzae and Streptococcus Mitis
    University of Rhode Island DigitalCommons@URI Open Access Dissertations 2021 MECHANISMS OF INTERACTION BETWEEN HAEMOPHILUS PARAINFLUENZAE AND STREPTOCOCCUS MITIS Dasith Perera Follow this and additional works at: https://digitalcommons.uri.edu/oa_diss MECHANISMS OF INTERACTION BETWEEN HAEMOPHILUS PARAINFLUENZAE AND STREPTOCOCCUS MITIS BY DASITH PERERA A DISSERTATION SUBMITTED IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY IN CELL & MOLECULAR BIOLOGY UNIVERSITY OF RHODE ISLAND 2021 DOCTOR OF PHILOSOPHY DISSERTATION OF DASITH PERERA APPROVED: Thesis Committee: Matthew Ramsey, Major professor David Nelson David Rowley Brenton DeBoef DEAN OF THE GRADUATE SCHOOL UNIVERSITY OF RHODE ISLAND 2021 ABSTRACT The human oral cavity is a complex polymicrobial environment, home to an array of microbes that play roles in health and disease. Oral bacteria have been shown to cause an array of systemic diseases and are particularly concerning to type II diabetics (T2D) with numerous predispositions that exacerbate bacterial infection. In this dissertation, we investigated the serum of healthy subjects and T2D subjects to determine whether we see greater translocation of oral bacteria into the bloodstream of T2D indiviDuals. We didn’t observe any significant enrichment of oral taxa, however we detected the presence of an emerging pathogen, Acinetobacter baumannii that is also associated with impaired inflammation in T2D. While some are associated with disease, many oral taxa are important in the pre- vention of disease. In this dissertation we investigated the interactions between two abundant health-associated commensal microbes, Haemophilus parainfluenzae and Streptococcus mitis. We demonstrated that H. parainfluenzae typically exists adjacent to Mitis group streptococci in vivo in healthy subjects.
    [Show full text]
  • 4. Chemical Stability of Nucleic Acids
    4. Chemical stability of nucleic acids NH2 N 8 N H O 2 Heterocycle: stable - O N P O N H except for reactions with alkylating O HO agents, nitrite and radicals Cleavage of the N-glycosidic bond Purinyl-N-glycosides are cleaved by acid HO OH or hydrazine treatment Cleavage of the phosphodiester linkage: RNA: hydrolyzed by 0.3 M KOH DNA: stable Chemical hydrolysis of phosphoesters Comparison: Ester hydrolysis of carboxylic acid O - O HO- O - R O-R´ + O-R´ R O-R´ R OH OH O tetrahedral transition state Acyl cleavage R O- + HO-R´ Proof: Reaction in 18O labeled water O + H2O, H O + R O R OH HO Alkyl cleavage in specific cases only Chemical hydrolysis of phosphoesters 1.Phosphotriester Apical ligand 1 M NaOH OMe O O 24 h, 20 °C Equatorial ligand - P O P MeO OMe MeO P MeO OMe OMe OMe O- + OH MeOH Trimethyl phosphate Dimethyl phosphate Transition state: dsp3-hybrid! Phosphoryl cleavage Chemical hydrolysis of phosphoesters 2.Phosphodiester 1 M NaOH O 100 °C O P P MeO O-Me MeO O- + MeOH O- O- HO- Dimethyl phosphate Methyl phosphate Alkyl cleavage Half life time: 16 days! Chemical hydrolysis of phosphoesters 2.Phosphodiester HO- H Cyclic phosphodiester (ring strain!) H O - O HO O O O P P O- O O-Me O P O - O- O O O- 2-Hydroxyethyl-methyl-phosphate 2-Hydroxyethyl phosphate Δ H = -26.8 kJ/mol Cf: Dimethyl phosphate: -7.5 kJ/ mol Half life time at 25 °C 25 min! Neighboring group participation Anchimeric assistance OH O H+ O OH OH O-Me + P P P O O H2O O O-Me O O O 2-Hydroxyethyl-methyl Ethylene-phosphate phosphate O 70% 30% P O O „Pseudorotation“: Interchange
    [Show full text]