J. Biol. Chem.-2005-Léger-Silvestre-38177-85

10
Specific Role for Yeast Homologs of the Diamond Blackfan Anemia-associated Rps19 Protein in Ribosome Synthesis * Received for publication, June 24, 2005, and in revised form, August 8, 2005 Published, JBC Papers in Press, September 12, 2005, DOI 10.1074/jbc.M506916200 Isabelle Le ´ ger-Silvestre , Jacqueline Marie Caffrey §1 , Rosy Dawaliby ‡1 , Diana Alehandrovna Alvarez-Arias , Nicole Gas ‡2 , Salvatore J. Bertolone , Pierre-Emmanuel Gleizes ‡2,3 , and Steven Robert Ellis §¶4 From the Laboratoire de Biologie Mole ´culaire des Eucaryotes (UMR5099) and Institut d’Exploration Fonctionnelle des Ge ´nomes (IFR109), CNRS and Universite ´ Paul Sabatier, 118 route de Narbonne, 31062 Toulouse, France and the Departments of § Biochemistry and Molecular Biology, Chemistry, and Pediatrics, University of Louisville, Louisville, Kentucky 40292 Approximately 25% of cases of Diamond Blackfan anemia, a severe hypoplastic anemia, are linked to heterozygous mutations in the gene encoding ribosomal protein S19 that result in haploinsuf- ficiency for this protein. Here we show that deletion of either of the two genes encoding Rps19 in yeast severely affects the production of 40 S ribosomal subunits. Rps19 is an essential protein that is strictly required for maturation of the 3-end of 18 S rRNA. Depletion of Rps19 results in the accumulation of aberrant pre-40 S particles retained in the nucleus that fail to associate with pre-ribosomal fac- tors involved in late maturation steps, including Enp1, Tsr1, and Rio2. When introduced in yeast Rps19, amino acid substitutions found in Diamond Blackfan anemia patients induce defects in the processing of the pre-rRNA similar to those observed in cells under- expressing Rps19. These results uncover a pivotal role of Rps19 in the assembly and maturation of the pre-40 S particles and demon- strate for the first time the effect of Diamond Blackfan anemia- associated mutations on the function of Rps19, strongly connecting the pathology to ribosome biogenesis. Diamond Blackfan anemia (DBA) 5 is a severe hypoplastic anemia that generally presents early in infancy (1, 2). Other clinical features of DBA are heterogeneous with some patients presenting craniofacial abnor- malities, growth failure, predisposition to cancer, and other congenital abnormalities. Most cases of DBA arise spontaneously, with only a small proportion exhibiting familial transmission typically showing autoso- mal dominant inheritance. Approximately 25% of the DBA cases have been linked to mutations in the gene encoding ribosomal protein S19 (RPS19), and in these cases haploinsufficiency for this ribosomal protein gives rise to the disease (3, 4). The remaining cases are of unknown etiology. The Rps19 protein is a component of the small ribosomal subunit, and as such, defects in some aspect of ribosome structure, function, or synthesis may be an underlying cause of DBA. The human Rps19 pro- tein belongs to a family of ribosomal proteins restricted to eukaryotes and archea. Rps19 does not have a homolog in the eubacterial ribosome where the properties of individual ribosomal proteins have been most extensively studied. As such, little is known regarding the function of members of the eukaryotic Rps19 family. In prokaryotes, ribosomal pro- teins play critical roles in ribosomal assembly through their interactions with each other and rRNA (5). These interactions promote both steps in rRNA processing important for subunit maturation and the formation of active sites in mature subunits necessary for ribosome function. The precise functions of the ribosomal proteins in the production of the subunits in eukaryotes are poorly characterized, which calls for a more detailed investigation of the role played by Rps19 in ribosome synthesis and function. The yeast Saccharomyces cerevisiae has proven to be an outstanding system to investigate factors involved in eukaryotic ribosome synthesis, including ribosomal proteins (6 –10). To date, ribosome synthesis in yeast has been broken down to a number of well characterized interme- diates, and numerous factors required for discrete steps in the pathway have been identified (11–13). Ribosomal RNAs are initially synthesized as a polycistronic transcript containing 18 S, 5.8 S, and 25 S rRNAs (14). The mature rRNAs are derived from the primary transcript by a series of processing steps most of which occur within the nucleolus. Pre-rRNA processing occurs concomitantly with the assembly of ribosomal pro- teins onto the rRNA transcripts. The earliest pre-rRNAs are found in a large ribonucleoproteic precursor of 90 S, which splits into pre-40 S and pre-60 S particles after endonucleolytic cleavage in the internal tran- scribed spacer 1 (ITS1) at site A 2 (see Fig. 5). Through proteomic anal- ysis, non-ribosomal factors specifically associated with these pre-ribo- somal particles at various maturation stages have been identified (15). Pre-40 S particles have been found to contain a set of specific non- ribosomal proteins that associate in the nucleus and accompany the particles into the cytoplasm where they are required for the final matu- ration of the small subunit (16). In yeast, Rps19 is encoded by duplicated genes, RPS19A and RPS19B, which differ from one another by a single amino acid. The yeast Rps19 proteins have over 50% sequence identity with the human protein (Fig. 1). The sequence similarity spans the bulk of the yeast and human pro- teins and includes blocks of near complete identity. One of these blocks, residues 52– 63 in humans, is a hot spot for amino acid substitutions in patients with DBA (4). Other amino acid substitutions found in DBA patients that occur outside of this region also fall in residues that are either identical or have conservative substitutions between yeast and humans. Given this high degree of conservation, studies on the function of the yeast Rps19 protein should provide insight into the function of its human homolog and processes affected in Diamond Blackfan anemia. Here we show that Rps19 is an essential protein that is required for the synthesis of the small ribosomal subunit. This phenotype in Rps19- * This work was supported in part by grants from the Kentucky Lung Cancer Research Program and NHLBI, National Institutes of Health (to S. R. E.). The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1 Both authors contributed equally to this work. 2 Received funds from the CNRS, the French Ministry of Research (Actions Concerte ´es Incitatives Microbiologie) and the Association pour la Recherche contre le Cancer (Paris, France). 3 To whom correspondence may be addressed. Tel.: 33-561-335-926; Fax: 33-561-335- 886; E-mail: [email protected]. 4 To whom correspondence may be addressed. Tel.: 502-852-5222; Fax: 502-852-6222; E-mail: [email protected]. 5 The abbreviations used are: DBA, Diamond Blackfan anemia; FISH, fluorescence in situ hybridization; rRNA, ribosomal RNA; ITS1, internal transcribed spacer 1; TAP, tandem affinity purification; DAPI, 4,6-diamidino-2-phenylindole. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 280, NO. 46, pp. 38177–38185, November 18, 2005 © 2005 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A. NOVEMBER 18, 2005 • VOLUME 280 • NUMBER 46 JOURNAL OF BIOLOGICAL CHEMISTRY 38177 at ULB on October 19, 2015 http://www.jbc.org/ Downloaded from

Transcript of J. Biol. Chem.-2005-Léger-Silvestre-38177-85

Page 1: J. Biol. Chem.-2005-Léger-Silvestre-38177-85

Specific Role for Yeast Homologs of the Diamond BlackfanAnemia-associated Rps19 Protein in Ribosome Synthesis*

Received for publication, June 24, 2005, and in revised form, August 8, 2005 Published, JBC Papers in Press, September 12, 2005, DOI 10.1074/jbc.M506916200

Isabelle Leger-Silvestre‡, Jacqueline Marie Caffrey§1, Rosy Dawaliby‡1, Diana Alehandrovna Alvarez-Arias¶,Nicole Gas‡2, Salvatore J. Bertolone�, Pierre-Emmanuel Gleizes‡2,3, and Steven Robert Ellis§¶4

From the ‡Laboratoire de Biologie Moleculaire des Eucaryotes (UMR5099) and Institut d’Exploration Fonctionnelle des Genomes(IFR109), CNRS and Universite Paul Sabatier, 118 route de Narbonne, 31062 Toulouse, France and the Departments of§Biochemistry and Molecular Biology, ¶Chemistry, and �Pediatrics, University of Louisville, Louisville, Kentucky 40292

Approximately 25% of cases of Diamond Blackfan anemia, asevere hypoplastic anemia, are linked to heterozygous mutations inthe gene encoding ribosomal protein S19 that result in haploinsuf-ficiency for this protein. Here we show that deletion of either of thetwogenes encodingRps19 in yeast severely affects the productionof40 S ribosomal subunits. Rps19 is an essential protein that is strictlyrequired for maturation of the 3�-end of 18 S rRNA. Depletion ofRps19 results in the accumulation of aberrant pre-40 S particlesretained in the nucleus that fail to associate with pre-ribosomal fac-tors involved in late maturation steps, including Enp1, Tsr1, andRio2. When introduced in yeast Rps19, amino acid substitutionsfound in Diamond Blackfan anemia patients induce defects in theprocessing of the pre-rRNAsimilar to those observed in cells under-expressing Rps19. These results uncover a pivotal role of Rps19 inthe assembly and maturation of the pre-40 S particles and demon-strate for the first time the effect of Diamond Blackfan anemia-associatedmutations on the function of Rps19, strongly connectingthe pathology to ribosome biogenesis.

DiamondBlackfan anemia (DBA)5 is a severe hypoplastic anemia thatgenerally presents early in infancy (1, 2). Other clinical features of DBAare heterogeneous with some patients presenting craniofacial abnor-malities, growth failure, predisposition to cancer, and other congenitalabnormalities.Most cases of DBA arise spontaneously, with only a smallproportion exhibiting familial transmission typically showing autoso-mal dominant inheritance. Approximately 25% of the DBA cases havebeen linked to mutations in the gene encoding ribosomal protein S19(RPS19), and in these cases haploinsufficiency for this ribosomal proteingives rise to the disease (3, 4). The remaining cases are of unknownetiology.The Rps19 protein is a component of the small ribosomal subunit,

and as such, defects in some aspect of ribosome structure, function, orsynthesis may be an underlying cause of DBA. The human Rps19 pro-

tein belongs to a family of ribosomal proteins restricted to eukaryotesand archea. Rps19 does not have a homolog in the eubacterial ribosomewhere the properties of individual ribosomal proteins have been mostextensively studied. As such, little is known regarding the function ofmembers of the eukaryotic Rps19 family. In prokaryotes, ribosomal pro-teins play critical roles in ribosomal assembly through their interactionswith each other and rRNA (5). These interactions promote both steps inrRNA processing important for subunit maturation and the formationof active sites in mature subunits necessary for ribosome function. Theprecise functions of the ribosomal proteins in the production of thesubunits in eukaryotes are poorly characterized, which calls for a moredetailed investigation of the role played by Rps19 in ribosome synthesisand function.The yeast Saccharomyces cerevisiae has proven to be an outstanding

system to investigate factors involved in eukaryotic ribosome synthesis,including ribosomal proteins (6–10). To date, ribosome synthesis inyeast has been broken down to a number of well characterized interme-diates, and numerous factors required for discrete steps in the pathwayhave been identified (11–13). Ribosomal RNAs are initially synthesizedas a polycistronic transcript containing 18 S, 5.8 S, and 25 S rRNAs (14).Themature rRNAs are derived from the primary transcript by a series ofprocessing steps most of which occur within the nucleolus. Pre-rRNAprocessing occurs concomitantly with the assembly of ribosomal pro-teins onto the rRNA transcripts. The earliest pre-rRNAs are found in alarge ribonucleoproteic precursor of 90 S, which splits into pre-40 S andpre-60 S particles after endonucleolytic cleavage in the internal tran-scribed spacer 1 (ITS1) at site A2 (see Fig. 5). Through proteomic anal-ysis, non-ribosomal factors specifically associated with these pre-ribo-somal particles at various maturation stages have been identified (15).Pre-40 S particles have been found to contain a set of specific non-ribosomal proteins that associate in the nucleus and accompany theparticles into the cytoplasm where they are required for the final matu-ration of the small subunit (16).In yeast, Rps19 is encoded by duplicated genes, RPS19A and RPS19B,

which differ from one another by a single amino acid. The yeast Rps19proteins have over 50% sequence identity with the human protein (Fig.1). The sequence similarity spans the bulk of the yeast and human pro-teins and includes blocks of near complete identity. One of these blocks,residues 52–63 in humans, is a hot spot for amino acid substitutions inpatients with DBA (4). Other amino acid substitutions found in DBApatients that occur outside of this region also fall in residues that areeither identical or have conservative substitutions between yeast andhumans. Given this high degree of conservation, studies on the functionof the yeast Rps19 protein should provide insight into the function of itshuman homolog and processes affected in Diamond Blackfan anemia.Here we show that Rps19 is an essential protein that is required for

the synthesis of the small ribosomal subunit. This phenotype in Rps19-

* This work was supported in part by grants from the Kentucky Lung Cancer ResearchProgram and NHLBI, National Institutes of Health (to S. R. E.). The costs of publicationof this article were defrayed in part by the payment of page charges. This article musttherefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section1734 solely to indicate this fact.

1 Both authors contributed equally to this work.2 Received funds from the CNRS, the French Ministry of Research (Actions Concertees

Incitatives Microbiologie) and the Association pour la Recherche contre le Cancer(Paris, France).

3 To whom correspondence may be addressed. Tel.: 33-561-335-926; Fax: 33-561-335-886; E-mail: [email protected].

4 To whom correspondence may be addressed. Tel.: 502-852-5222; Fax: 502-852-6222;E-mail: [email protected].

5 The abbreviations used are: DBA, Diamond Blackfan anemia; FISH, fluorescence in situhybridization; rRNA, ribosomal RNA; ITS1, internal transcribed spacer 1; TAP, tandemaffinity purification; DAPI, 4�,6-diamidino-2-phenylindole.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 280, NO. 46, pp. 38177–38185, November 18, 2005© 2005 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

NOVEMBER 18, 2005 • VOLUME 280 • NUMBER 46 JOURNAL OF BIOLOGICAL CHEMISTRY 38177

at UL

B on O

ctober 19, 2015http://w

ww

.jbc.org/D

ownloaded from

Page 2: J. Biol. Chem.-2005-Léger-Silvestre-38177-85

depleted cells is linked to a clear pre-rRNA processing defect at site A2and to the nuclear retention of 40 S subunit precursors, which fail torecruit certain non-ribosomal factors involved in subunit maturation.In addition, a similar defect in pre-rRNA processing is induced by theexpression of yeast Rps19 mutants carrying missense mutations foundin DBA patients. These results indicate that Rps19 is a key protein inribosome biogenesis and establish a link between partial or total loss offunction of the DBA-linked protein Rps19 and a strong defect in pre-rRNA processing and subunit maturation.

EXPERIMENTAL PROCEDURES

Yeast and Bacterial Strains—The yeast strains used in this studyweregenerated by the Saccharomyces genome deletion project consortiumand obtained from Research Genetics or Euroscarf. Media used in cul-tivating yeast were YPD (1% (w/v) yeast extract, 2% (w/v) peptone, and2% (w/v) glucose) and synthetic (0.67% (w/v) yeast nitrogen base with-out amino acids and 2% (w/v) glucose). Where appropriate, nutrientswere added to syntheticmedia in amounts specified by Sherman (17). Insome experiments glucose was replaced as a carbon source by 0.2%(w/v) sucrose and 2% (w/v) galactose. Diploids were sporulated on solidsporulationmedia (1% (w/v) potassium acetate, 0.1% (w/v) yeast extract,0.05% (w/v) glucose, and 2% (w/v) agar). Additional nutrients wereadded to sporulation media in amounts corresponding to 25% of thatused in synthetic media. A haploid yeast strain (GAL-RPS19), withchromosomal alleles of RPS19A and RPS19B disrupted and growth sup-ported by a plasmid-borne allele of RPS19A under control of the GAL1promoter, was created to analyze the effects of depletion of both Rps19proteins. The RPS19A gene was amplified from genomic DNA by PCRand subcloned into pFL38-GAL (10). This plasmid was transformedinto a diploid yeast strain heterozygous for RPS19A and RPS19B disrup-tions generated by crossing strains Y06271 (�rps19A/RPS19B) andY11142 (RPS19A/�rps19B) obtained from Euroscarf. Resulting trans-formants were sporulated, and tetrads were dissected on rich mediawith 2% (w/v) galactose as carbon sources. Haploid progeny were ini-tially tested for growth on 2% (w/v) glucose to identify strains whosegrowthwas galactose-dependent. The status of the chromosomal allelesof RPS19A and RPS19B in these strains was assessed using oligonucleo-tides flanking the kanMX disruption cassette in each gene. Two haploidstrainswere identified that had growth dependent on the plasmid-borneallele of RPS19A under control of the GAL1 promoter. The Escherichiacoli strain used in this study was XL1-Blue (Stratagene, La Jolla, CA).

RPS19 Mutations—The UAS and entire coding sequence of RPS19Awere amplified by PCRusing PfuUltraHotstart polymerase (Stratagene,La Jolla, CA). The PCR product was cloned into pRS315. Missensemutations in RPS19 reported for DBA patients were introduced at cor-

responding positions in the RPS19A gene using the Genetailor site-directed mutagenesis kit (Invitrogen). Mutations introduced intoRPS19A were confirmed by sequencing, which also ruled out the intro-duction of spurious mutations during the mutagenesis procedure. Plas-mids carrying either wild-type or mutant alleles of RPS19A were trans-formed into the GAL-RPS19 strain growth on 2% galactose. Thefunctions of RPS19A alleles on the pRS315 plasmid were analyzed aftershifting growth to media containing 2% glucose.

Polysome Analysis—Polysomes were prepared and fractionated on7–47% sucrose gradients as described byBaim et al. (18). Centrifugationwas generally carried out at 18,000 rpm for 16 h in an SW28.1 rotor(Beckman Instruments, Fullerton, CA). Sucrose gradients were frac-tionated, and absorbance at 254 nm was monitored using an ISCOmodel 185 gradient fractionator and a UA-6 absorbance detector. Datawere digitized using UN-SCAN-IT software (Silk Scientific, Orem, UT)or Adobe Photoshop.

Northern Analysis—Total RNA was prepared from yeast by hot phe-nol extraction (19), separated on a 1% agarose gel, and passively trans-ferred to a nylon membrane (Amersham Pharmacia Biotech). In Fig. 4,the membrane was hybridized overnight at 50 °C with the followingoligonucleotides: probe 5�-A0 (5�-GCAGATCTGACGATCACC-3�),probe A0–A1 (5�-GATCGTTCTCCCTTACCCAC-3�), probe 18 S (5�-CATGGCTTAATCTTTGAGAC-3�), probe D-A2 (5�-TTAAGCG-CAGGCCCGGCTGG-3�), probe A2–A3 (5�-GATTGCTCGAATGC-CCAAAG-3�), probe E-C2 (5�-GGCCAGCAATTTCAAGTTA-3�) andprobe 25 S (5�-CCATCTCCGGATAAACC-3�). In Fig. 8, membraneswere hybridized overnight at 37 °C with probe A2–A3 (5�-AT-GAAAACTCCACAGTG-3�) and probe 18S (5�-TGATCCTTCCG-CAGGTTCACCTACGGAAAC-3�). The probes (10 ng) were labeledwith [�-32P]ATP using T4 polynucleotide kinase (Promega). Afterhybridization, the blots were washed twice in 0.1% SSC, 0.1% SDS, or 3times in 6� SSC and analyzed by phosphorimaging.

Pulse-chase Analysis—Pulse-chase labeling of pre-rRNA was carriedout as described previously (7). The GAL-RPS19 strain was used in thepulse-chase experiments. Cells were initially grown in synthetic medialacking uracil containing 0.2% (w/v) sucrose and 2% (w/v) galactosefollowed by depletion of Rps19 by shifting to syntheticmedia containing2% (w/v) glucose. After 7 h in glucose-containing media, cells wererecovered from 40 ml of each culture (A600 0.2–0.3) by centrifugation.Cells from both cultures were each suspended in 1 ml of glucose-con-taining syntheticmedia lacking uracil andmethionine. Each culture waspulse-labeled for 2min at 30 °Cwith 250�Ci of [methyl-3H]methionine.Labeled cells in 250-�l aliquots were diluted into synthetic media con-taining 1mg/mlmethionine and either placed on ice (0 chase period) orincubated for 2, 5, or 10 min at 30 °C. After the chase period cells were

FIGURE 1. Alignment of yeast and human Rps19proteins. Species designations are indicated assuperscripts to the right of each sequence (Sc, S.cerevisiae; Hs, Homo sapiens). The numbers to theleft indicate the position of the first residue listedin the amino acid sequence of each protein. Theyeast Rps19B protein differs from the Rps19A pro-tein shown by an alanine for proline substitutionat residue 2. The line between the yeast andhuman sequences shows identical residues.Amino acid substitutions in DBA patients areunderlined and shown below the human sequence(4, 29). The sequences in bold represent thehotspot for DBA mutations. Some mutations in thehot spot region are found in more than onepatient.

Rps19 Is Required for 40 S Ribosomal Subunit Synthesis

38178 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 280 • NUMBER 46 • NOVEMBER 18, 2005

at UL

B on O

ctober 19, 2015http://w

ww

.jbc.org/D

ownloaded from

Page 3: J. Biol. Chem.-2005-Léger-Silvestre-38177-85

quickly chilled on ice prior to the isolation of total RNA. RNA wasfractionated on 1.5% agarose-formaldehyde gels and transferred toZeta-probe membrane. Membranes were baked for 2 h at 80 °C andexposed to BioMax MS film at �70 °C using a BioMax LE intensifyingscreen (Eastman Kodak Co., Rochester, NY). Film was exposed from 2weeks to 1 month. The long development time resulted from the factthat the strains in the deletion collection are methionine auxotrophs.Inclusion ofmethionine in the culturemediumduring and after the shiftto glucose reduces the efficiency of radiolabeling with [methyl-3H]me-thionine during the pulse period.

Co-immunoprecipitation and Immunolocalization of TAP Proteins—Affinity purification of TAP-taggedNoc4p, Enp1p, Tsr1p, Ltv1p, Rio2p,and analysis of co-precipitating RNAswere performed as described pre-viously (20). Immunolocalization of TAP proteins was performed asdescribed before (10). Briefly, spheroplasts were incubated for 2 h atroom temperature with rabbit anti-protein A antibodies (Sigma) inphosphate-buffered saline/bovine serum albumin (1:50,000 dilution).Fluorescence detection was achieved with Alexa Fluor 594-conjugatedgoat anti-rabbit IgG (H�L) antibodies (Molecular Probes), and DNAwas counterstained with DAPI. Images were captured with a CoolsnapES camera (Roper Scientifics) mounted on a DMRBmicroscope (Leica)using the Metavue software (Universal Imaging).

Fluorescence in Situ Hybridization and Electron Microscopy—Detec-tion of pre-rRNA by FISH was performed as described by Gleizes et al.(21). The sequence of the ITS1 oligonucleotidic probe is TT*GCACA-GAAATCTCT*CACCGTTTGGAAT*AGCAAGAAAGAAACT*TA-CAAGCT*T (ITS1 probe), whereT* represents amino-modified deoxy-thymidine conjugated to Cy3. Cells were prepared for electron micros-copy by high pressure freezing (EM Pact, Leica). The samples were thentransferred to �90 °C for cryosubstitution in acetone containing 0.2%uranyl acetate, 0.1% glutaraldehyde, 0.2% osmium tetroxide for 72 h.This fixative was then replaced by pure acetone, and temperature wasraised to �45 °C for embedding in Lowicryl HM20 resin. The resin waspolymerized under UV light for 2 days at �45 °C and 1 day at �20 °C.

RESULTS

Rps19 Is Essential for Cell Viability—To determine the extent towhich yeast cell growth and division is dependent on the RPS19 genes,strains deleted for one or the otherRPS19 gene (obtained from the YeastDeletion Collection) were crossed to create a diploid strain heterozy-gous for both RPS19 deletions. After sporulation, the resulting tetradswere dissected, and the haploidmeiotic progeny from individual tetradswere examined for growth on rich media at 30 °C. The results from thisanalysis indicate that disruption of either RPS19 gene alone reducesgrowth rate, whereas spores fail to germinate when both genes aredeleted (Fig. 2A). To examine whether Rps19 is required for vegetativegrowth, a strain (GAL-RPS19) was created where the chromosomalcopies of RPS19A and RPS19B are disrupted and a plasmid-borne copyof RPS19A is expressed under control of the GAL1 promoter. Thesedata reveal that growth ceases when cells are shifted to glucose andRPS19 expression is shut off, indicating that the yeast Rps19 proteins areessential for cell growth and division (Fig. 2B).

Shortage of Rps19 Preferentially Affects the Production of 40 SSubunits—To address whether deleting either RPS19 gene influencesthe translational machinery we prepared cell extracts from wild-typeand RPS19 deletion strains and passed these extracts through sucrosegradients to generate polysome profiles. Thismethod provides a steady-state measure of free ribosomal subunits and ribosomes engaged inprotein synthesis. Fig. 3 shows that extracts from the�rps19B strain hadreduced levels of free 40 S subunits and fewer polysomes relative to

wild-type. The overall rate of ribosome synthesis in the�rps19B strain isalso decreased, which is likely secondary to the 40 S subunit deficiencyand is correlated with reduced growth rate. Experiments with the GAL-RPS19 strain showed that the level of free 40 S subunits was reduced andfree 60 S subunits increased 3 h after a shift to glucose-containingmedia(Fig. 3, compareGAL-RSP19(Gal)withGAL-RPS19(Glu)). Under theseconditions the secondary effects on global ribosome synthesis are not asdramatic as those seen in the �rps19B strain. Data for the RPS19 dele-tion strains are similar to results reported for disruption of genes encod-ing other ribosomal proteins that are essential components of the 40 Sribosomal subunit (8, 22).

Production of 18 S rRNA Is Blocked in the Absence of Rps19—Thereduced amount of 40 S subunits in RPS19 deletion strains could be theconsequence of a defect in subunit synthesis or enhanced subunit deg-radation. We examined flux through the yeast rRNA-processing path-way in wild-type and GAL-RPS19 cells by pulse-chase analysis. Thestrains were grown in glucose containing medium for 7 h to depleteRps19 in the GAL-RP19 cells, and ribosomal RNA precursors were

FIGURE 2. Yeast Rps19 proteins are required for cell growth and division. A, out-growth of spores derived from a diploid heterozygous at the RPS19A/rps19A::kanMX andRPS19B/rps19B::kanMX loci. The heterozygous diploid was sporulated, and tetrads weredissected. Haploid progeny were grown on YPD plates at 30 °C. RPS19A/B genotypes arelisted below each tetrad. The genotype for double mutants was inferred. B, depletion ofRps19 by regulated expression from the GAL1 promoter inhibits growth. The strainlabeled GAL-RPS19 has the chromosomal copies of RPS19A and RPS19B deleted andplasmid-borne RPS19A under the control of the GAL1 promoter as the only potentialsource of functional Rps19 protein. The WT strain has wild-type chromosomal copies ofRPS19A and RPS19B in addition to the plasmid containing GAL1-RPS19A. Cells were main-tained on galactose-containing media and then shifted to glucose-containing media torepress expression from the GAL1 promoter. Cell growth was monitored by optical den-sity measurements at 600 nm.

Rps19 Is Required for 40 S Ribosomal Subunit Synthesis

NOVEMBER 18, 2005 • VOLUME 280 • NUMBER 46 JOURNAL OF BIOLOGICAL CHEMISTRY 38179

at UL

B on O

ctober 19, 2015http://w

ww

.jbc.org/D

ownloaded from

Page 4: J. Biol. Chem.-2005-Léger-Silvestre-38177-85

radiolabeled with a brief pulse of [methyl-3H]methionine. The radiola-beled methionine rapidly equilibrates with S-adenosylmethionine,which is used in the methylation of ribosomal RNA precursors. Meth-ylation occurs early in the synthesis of rRNA, efficiently labeling the 35Sprimary transcript. Processing was then followed after chase periods inthe presence of unlabeled methionine.Fig. 4A shows that during the labeling period wild-type cells rapidly

processed the 35 S/32 S precursors to 27 S and 20 S species, which werethen matured to 25 S and 18 S rRNAs in the first 2 min of the chaseperiod. The GAL-RPS19 strain, in contrast, showed delayed processingto the 35 S and 32 S precursors during the labeling period. Althoughthere was some delay in the production of 25 S rRNA upon depletion ofRps19, the amount of 25 S rRNA produced by 5 to 10 min of chaseapproached that observed in wild-type cells. The production of 18 SrRNA, on the other hand, was severely compromised in the absence ofRps19: maturation of the 18 S rRNA stopped after synthesis of a lateprecursor, which corresponds to 21 S pre-rRNA (see below). Thus,although flux through the entire rRNA processing pathway appearsreduced in Rps19-depleted cells mutants relative to wild-type, stepsleading to the production of 18 S rRNA are clearly impacted moreseverely than those leading to 25 S rRNA.

Rps19 Is Required for Pre-rRNA Processing at Site A2—To determinemore precisely the pre-rRNAprocessing defect(s) induced by the deple-tion of Rps19, we next performed Northern analysis on total RNAsisolated from GAL-RPS19 cells using oligonucleotidic probes (Fig. 4B).Cells were grown either on galactose or shifted for 4 and 8 h to glucose-containing medium to shut down Rps19 synthesis. An outline of theyeast rRNA processing pathway is shown in Fig. 5A.While the amount of 25 S rRNA stays roughly constant upon shutting

downRps19 expression the level of 18 S rRNAdecreases (Fig. 4B, probes18S�25S). The decrease in 18 S rRNA is accompanied by an increase in35 S and 23 S precursors indicating that cells depleted of Rps19 areinefficient at cleaving rRNA precursors at the A0 and A1 sites withinETS1 (Fig. 4B, probes 5�-A0 and A0–A1). Blots with an oligonucleotidethat hybridizes to sequences between the D and A2 cleavage sitesrevealed a severe reduction of the amount of 20 S pre-rRNA, a normalintermediate in the pathway leading to 40 S subunits, and an increase inwhat appears to be 21 S pre-rRNA, which extends from the 5�-end ofmature 18 S rRNA to the A3 cleavage site (Fig. 4B, probe D-A2). Anoligonucleotide complementary to the A2–A3 segment confirmed thatthis band was 21 S pre-rRNA (Fig. 4B, probe A2–A3). This probe alsoshowed that the 23 S rRNA intermediate extends past the A2 cleavagesite. Thus, cells depleted of Rps19 are defective in cleavage at the A2 sitewithin ITS1, as illustrated in Fig. 5B. The relative amounts of 23 S and 21S rRNA precursors accumulating in the Rps19-depleted cells indicate

that the primary defect is in ITS1 with secondary effects on cleavagewithin ETS1, consistent with previous reports on the coupling betweencleavage at sites A0, A1, and A2 sites (14). In parallel, the 27 S-A2 pre-cursor was decreased in cells depleted of Rps19 providing further sup-port for the influence of Rps19 on cleavage at the A2 site (Fig. 4B, probeA2–A3). In contrast, the levels of 27 S-A3 and 27 S-B showed only amodest reduction in Rps19-depleted cells (Fig. 4B, probe E-C2) indicat-ing that relatively efficient cleavage of 35 S and 32 S precursors at site A3can still generate normal intermediates in the pathway leading tomature 25 S rRNA. Very similar pre-rRNA profiles were obtained whenonly one RPS19 gene was deleted: the major processing defect observedin these strains was accumulation of 21 S pre-rRNA (data not shown).These data indicate that Rps19 is necessary for efficient maturation ofthe pre-rRNA ETS1 and pinpoint the strict requirement of this riboso-mal protein for processing at the A2 site in the ITS1.

Nuclear Retention of Pre-40 S Particles—Wenext examined the intra-cellular fate of the 21 S pre-rRNA-containing particles accumulatingupon depletion of Rps19 and determined whether they can be exportedto the cytoplasm or are retained in the nucleus and degraded. Fig. 6Ashows the localization by fluorescence in situ hybridization of thepre-40 S particles with a probe complementary to the D-A2 domain ofITS1, in wild-type and mutant cells. As previously observed (10), wild-type cells display labeling in the nucleolus, where most ribosome bio-genesis is performed, and in the cytoplasmwhere conversion of the 20 Spre-rRNA to 18 S mature RNA occurs. In GAL-RPS19 cells depleted ofRps19, one observes a striking decrease of the cytoplasmic signal, whichindicates that the pre-40 S particles are no longer efficiently exportedfrom the nucleus. Precursors to the small subunit accumulate in thenucleus with most of the signal being restricted to the nucleolus, whichcan be distinguished from the nucleoplasmdue to very low stainingwithDAPI. Labelingwith a probe complementary to theA2–A3 segment alsoyields a nucleoplasmic signal in Rps19-depleted cells and not in wildtype, consistent with the nuclear accumulation of pre-40 S particlescontaining 21 S pre-rRNA (data not shown). Single deletion of RPS19Aor RPS19B is sufficient to induce a similar phenotype (Fig. 6B). Thesedata are very reminiscent of our previous observations of cells depletedof Rps18, which is also required for efficient cleavage at the A2 cleavagesite within ITS1 (8, 10). The strong nucleolar signal is compatible with adefect in early pre-rRNA cleavage steps taking place in the nucleolus.Together with the accumulation of pre-rRNA precursors in the

nucleus, another conspicuous feature in a majority of Rps19-depletedcells is the fact that the nucleolus (as seenwith theD-A2 probe) becomesround and appears to detach from the DNA.We visualized this pheno-type at a higher resolution by electron microscopy (Fig. 6C). In severalsections, we observed what appeared to be the vacuole engulfing the

FIGURE 3. Strains containing disrupted alleles of RPS19 are deficient in 40 S ribosomal subunits. Cell extracts were prepared as described by Baim et al. (18) and loaded onto7– 47% sucrose gradients. Gradients were centrifuged at 18,000 rpm for 16 h. Gradients were broken down using an ISCO 185 gradient fractionator and absorbance at 254nm wasmeasured using a UA-6 absorbance detector. Strain designations are listed above each panel. The wild-type and �rps19B strains were grown in YPD media and harvested inmid-exponential phase. The GAL-RPS19 strain was grown in synthetic complete media containing 2% galactose and shifted to media containing 2% glucose. Cells were harvestedboth prior to the nutritional shift, GAL-RPS19(Gal), and after 3 h in glucose, GAL-RPS19(Glc).

Rps19 Is Required for 40 S Ribosomal Subunit Synthesis

38180 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 280 • NUMBER 46 • NOVEMBER 18, 2005

at UL

B on O

ctober 19, 2015http://w

ww

.jbc.org/D

ownloaded from

Page 5: J. Biol. Chem.-2005-Léger-Silvestre-38177-85

nucleolus. These images strongly suggest that microautophagy of thenucleus is induced by reduced levels of Rps19, a mechanism previouslyproposed to be involved in down-regulation of ribosome biogenesis(23).

Rps19 Is Necessary for Recruitment of Pre-ribosomal Factors—To getinsight into the stage at which the assembly of the 40 S particle is

blocked upon depletion of Rps19, we examined association of severalnon-ribosomal factors with pre-rRNAs in the GAL-RPS19 strain. Weused TAP-tagged genes encoding Noc4, Enp1, Tsr1, and Rio2 as ameans of studying the association of these proteins with pre-ribosomalparticles. In wild-type cells, Noc4 is found associated with 90 S particles,whereas Tsr1 and Rio2 are present in pre-40 S particles. Enp1 is foundassociated with both types of pre-ribosomes. The tagged proteins wereprecipitated with IgG-Sepharose beads, and the co-purifying pre-rR-NAs were analyzed by Northern blots with the D-A2 probe (Fig. 7A).Detection of the 25 S rRNA, which does not associate with these taggedproteins, showed that less than 0.1% of this RNA (relative to input)co-precipitated non-specifically (not shown).In wild-type cells, Noc4 co-precipitated with RNA species found in

the 90 S particles (35 S and 32 S), but not significantly with 20 S pre-rRNA. As shown previously, the precursor containing 20 S rRNA wasinstead isolated in significant amounts in co-precipitates with Enp1,Tsr1, and Rio2. Upon depletion of Rps19, this pattern was dramaticallyaltered. The amount of pre-rRNA co-precipitated with Enp1, Tsr1, and

FIGURE 4. Analysis of ribosomal RNA processing in Rps19-depleted cells. A, pulse-chase analysis of rRNA synthesis. Wild-type and GAL1-RPS19 cells were grown on galac-tose and then shifted to glucose-containing media. Seven hours after the nutritionalshift, cells were harvested, concentrated, and pulse-labeled with [3H]methionine asdescribed under “Experimental Procedures.” After pulse labeling, cells were chased for 0,2, 5, and 10 min in media containing unlabeled methionine. Total RNA was isolated fromcells at each time point, fractionated on formaldehyde agarose gels, and transferred toZetaprobe membrane. The membrane was exposed to Kodak BioMax MS film at �70 °Cwith a Kodax LE-intensifying screen. B, Northern analysis was performed on total RNAsisolated from GAL-RPS19 cells grown on galactose (Gal) or shifted for 4 and 8 h to glu-cose-containing media (Glc). RNAs were fractionated on formaldehyde-agarose gels andtransferred to nylon membranes. Membranes were blotted with oligonucleotidic probescomplementary to mature rRNAs or to transcribed spacers listed under “ExperimentalProcedures.”

FIGURE 5. Pre-rRNA processing in S. cerevisiae. A, major pre-rRNA processing pathway.Letters designate the endonucleolytic and exonucleolytic cleavage points. Circles indi-cate the cleavages required to proceed to the next step. B, alternative pathway in Rps19-depleted cells. Cleavage at point A3 substitutes deficient processing at point A2 to yieldthe 21 S and 27S-A3 pre-rRNAs. The latter is then normally converted to the mature 5.8 Sand 25 S species, whereas processing of the former is blocked.

Rps19 Is Required for 40 S Ribosomal Subunit Synthesis

NOVEMBER 18, 2005 • VOLUME 280 • NUMBER 46 JOURNAL OF BIOLOGICAL CHEMISTRY 38181

at UL

B on O

ctober 19, 2015http://w

ww

.jbc.org/D

ownloaded from

Page 6: J. Biol. Chem.-2005-Léger-Silvestre-38177-85

Rio2 was drastically reduced, indicating that these proteins no longerassociate with pre-ribosomes in the absence of Rps19. Interestingly, theamount of 21 S pre-rRNA co-precipitating with Noc4 was significantlyhigher under these conditions, which suggests that some of the proteinsassociated with the 90 S precursors remain in the abnormal pre-40 Sparticles, which accumulate in the absence of Rps19.We next localized the TAP-tagged proteins by immunofluorescence.

As seen in Fig. 7B, Noc4-TAP, which is a nucleolar protein in wild-typecells, was also localized in part to the nucleoplasm in GAL-RPS19-de-pleted cells. This patternwas very similar to that of the pre-40 S particles(Fig. 6A), consistent with the association of Noc4 with the 21 S pre-rRNA detected by TAP purification. In contrast, the late associatingRio2 remained mostly cytoplasmic as in wild-type cells. Enp1 and Tsr1localization switched from partially cytoplasmic in wild-type cells tofully nuclear in Rps19-depleted cells. The distribution of these two pro-teins within the nucleus was homogeneous, and thus differed from theITS1 FISH signal, which remains predominantly nucleolar under theseconditions. We therefore infer that, although not binding to pre-40 Sparticles, these proteins are nevertheless imported in the nucleus.These data show that Rps19 is necessary for recruitment of certain

non-ribosomal factors normally found associated with pre-40 S parti-cles. Absence of these factors may preclude processing at site A2 andfurther maturation of the precursors to the 18 S rRNA.

MissenseMutations Identified in DBA Patients Influence Growth andrRNA Maturation When Incorporated into the Yeast RPS19A Gene—The results described above indicate that null alleles of RPS19A andRPS19B influence rRNA processing by interfering with cleavage at theA2 site within ITS1. Most of the mutations identified to date in theRPS19 gene of DBA patients would be similar to these null alleles eitherthrough large deletions, splicing defects, or frameshift mutations thatwould interfere with the production of functional Rps19 protein. Inaddition, several patients have missense mutations in the RPS19 gene,which are presumably responsible for the disease (Fig. 1). We reporthere a novel missense mutation identified in a DBA patient, whichresults in a proline for leucine substitution at position 64 in the humanprotein. This change occurs in the Rps19 consensus sequence adjacentto the hot spot for missense mutations in DBA patients. This polymor-phism has not been reported in the RPS19 sequence of over 50 normalcontrol individuals and as such, may represent a pathogenic lesion.6

However, whether this mutation or other reported missense mutationsassociated with DBA affect Rps19 function has not been determined.Many of these missense mutations fall in regions of Rps19 highly con-served between yeast and humans. Thus, we analyzed the effects ofseveral of these missense mutations on growth and pre-rRNA process-ing in yeast.

6 H. Gazda, personal communication, Dana Farber Cancer Institute.

FIGURE 6. Depletion of Rps19 results in nuclearretention of pre-40 S particles and inducesmicroautophagy of the nucleus. A, detection ofpre-18 S rRNA by FISH with a probe hybridizingwithin the D-A2 domain of ITS1. Wild-type andGAL-RPS19 cells cultured in galactose (Gal) con-taining medium were put in the presence of glu-cose (Glc) for 4 h before fixation. Arrowheads pointto the nucleoplasm revealed by DAPI staining. Thecytoplasm is indicated by an asterisk. Images ofwild-type and Gal-RPS19 cells were created withthe same exposure time. B, examples of cellsdeleted of either RPS19A or RPS19B treated as in A.C, electron micrographs of wild-type and GAL-RPS19 cells after 4 h of culture in glucose-contain-ing medium. Insets illustrate parallel situations asseen by FISH with the D-A2 probe. Cy, cytoplasm;CW, cell wall; Np, nucleoplasm; Nu, nucleolus; Va,vacuole.

Rps19 Is Required for 40 S Ribosomal Subunit Synthesis

38182 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 280 • NUMBER 46 • NOVEMBER 18, 2005

at UL

B on O

ctober 19, 2015http://w

ww

.jbc.org/D

ownloaded from

Page 7: J. Biol. Chem.-2005-Léger-Silvestre-38177-85

The GAL-RPS19 strain was transformed with a second plasmid car-rying mutant alleles of RPS19A under control of its own promoter.When this strain is grown on glucose-containing media, the mutantalleles of RPS19A are the primary source of functional Rps19 proteinavailable to support growth. As shown in Fig. 8A, the I65P substitution,similar to the L64P mutation found in the DBA patient reported here,completely abolishes the ability of the mutant Rps19A protein to sup-port growth on glucose indicating that this missense mutation has asevere effect on Rps19 function. Two other mutations studied, R63Q(position 62 in the human sequence) and I15F, also have demonstrableeffects on the ability of the yeast Rps19A protein to support growth. Incontrast, two other missense mutations, A62S (codon 61 in humans)and G121S (codon 120 in humans), do not influence the ability of theyeast protein to support growth. Interestingly, A62S is no longer con-sidered a pathogenic alteration in DBA patients, and as such, would notbe considered likely to have an affect on Rps19 function.7

We next determined the influence of these mutations on ribosomesynthesis by isolating RNA from each of these strains 22 h after shiftingfrom galactose- to glucose-containing media. Northern blot analysis(Fig. 8B) revealed that strains where growth is compromised by thepresence of a DBA-related missense mutation in RPS19A also showincreased 21 S rRNA relative to strains where growth is not affected.Moreover, the pre-40 S particles produced in these mutants wereretained in the nucleus and accumulated in the nucleolus, as seen by insitu hybridization (Fig. 8C). Thus, there is a close correspondencebetween the ability of DBA-relatedmissensemutations to affect growthin yeast and their ability to abrogate the function of Rps19 in ribosomebiogenesis.

DISCUSSION

The gene encoding ribosomal protein S19 is the only gene known tobe involved in the pathogenesis of Diamond Blackfan anemia. Approx-imately 25% of DBA patients have mutations in RPS19. The remainingcases of DBA are of unknown etiology. Studies in the yeast S. cerevisiaehave shown that some ribosomal proteins are required for the efficient7 S. Ball, personal communication, St. George’s Hospital Medical School.

FIGURE 7. Behavior of pre-ribosomal factors inRps19-depleted cells. A, detection of pre-18 SrRNAs co-purifying with TAP-tagged Noc4, Enp1,Tsr1, and Rio2 in Rps19-depleted cells. GAL-RPS19cells expressing one of the TAP-tagged proteinswere grown for 4 h in YP-glucose medium, andtotal extracts were submitted to immunoprecipi-tation with IgG-Sepharose. Co-precipitating RNAswere revealed after Northern blot with a probehybridizing between point D and A2 in ITS1. Allsamples, were treated in the same conditions; onerepresentative input lane (0.1% of the input) is dis-played. The levels of the TAP-tagged proteinswere checked in inputs by Western blotting withan anti-protein A antibody. Semi-quantification ofthe fraction of precipitated RNA was performed byPhosphorImager analysis. B, immunolocalizationof TAP-tagged Noc4, Enp1, Tsr1, and Rio2 in WTand GAL-RPS19 strains cultured 4 h in the pres-ence of glucose. Immunofluorescence was carriedout with anti-protein A antibodies. Arrowheadsindicate the nucleoplasm as visualized by DNAstaining with DAPI. The cytoplasm is designatedby asterisks.

Rps19 Is Required for 40 S Ribosomal Subunit Synthesis

NOVEMBER 18, 2005 • VOLUME 280 • NUMBER 46 JOURNAL OF BIOLOGICAL CHEMISTRY 38183

at UL

B on O

ctober 19, 2015http://w

ww

.jbc.org/D

ownloaded from

Page 8: J. Biol. Chem.-2005-Léger-Silvestre-38177-85

maturation of ribosomal subunits (6–8, 10). These studies have linkedindividual ribosomal proteins to specific steps in rRNA processing dur-ing subunit maturation. We therefore turned to yeast to gain a betterunderstanding of the function of the Rps19 family of proteins. Disrup-tion of either of the yeast RPS19 genes caused a reduction in growth rateand a deficiency of 40 S ribosomal subunits. Disruption of both RPS19genes is lethal. This observation is consistent with recent data showingthat a homozygous RPS19 deletion in mice is lethal prior to the blasto-cyst stage indicating that the mammalian Rps19 protein also plays acritical role in cell growth and division (24).Our data reveal a critical role of Rps19 in the production of the small

ribosomal subunit. The most striking defect induced by depletion ofRps19 is a strong inhibition of cleavage at site A2 within ITS1 and of thesubsequent maturation of the 3�-end of the 18 S rRNA. The endonu-cleolytic cleavage at A2 takes place within the nucleolus and results inrelease of pre-40 S and pre-60 S particles from the 90 S ribosomal pre-cursor. Absence of Rps19 preferentially affects the production of 40 Sribosomal subunits, as cleavage at site A3 still occurs, giving rise to anormal intermediate in the pathway leading to 60 S subunits. The 21 Spre-rRNA is not a dead-end product of the 40 S production pathway,and it may be converted into mature 18 S rRNA, as seen with somemutations in the RRP5 gene (25, 26). However, the 21 S rRNA-contain-ing pre-40 S particles, which accumulate in Rps19-depleted cells areretained in the nucleus and are inefficiently if at all, processed tomature40 S subunits.In addition to the structural components of the ribosome, numerous

other non-ribosomal factors must assemble with the pre-ribosomes forvarious steps in pre-rRNAprocessing and transport. Our data show thatthe aberrant pre-40 S particles that accumulate in Rps19-depleted cellslack the non-ribosomal factors Enp1, Tsr1, and Rio2, which indicate amajor failure in the assembly of the pre-40 S particles. In wild-type cells,

Tsr1 and Rio2 are found associated with pre-40 S subunits after the 90 Spre-ribosome is split by cleavage at A2. They are not required for A2cleavage, but are instead needed for cleavage at site D, which yieldsmature 18 S rRNA in the cytoplasm (10, 27). The failure of aberrantpre-40 S subunits found in cells depleted of Rps19 to recruit Rio2 couldbe a consequence of the retention of these particles within the nucleus/nucleolus, as Rio2 is predominantly cytoplasmic. Tsr1, on the otherhand, is present in the nucleus and the nucleolus suggesting that theaberrant pre-40 S subunits may be unable to recruit Tsr1 even withinthe nucleolus. In wild-type cells, Enp1 is a component of early 90 S andlate 40 S pre-ribosomes (16). Thermosensitive mutants of ENP1 show astrong accumulation of 21 S rRNAat non-permissive temperatures sim-ilar to the phenotype observed here forRPS19mutants (28). Thus Rps19and Enp1 could cooperate within the 90 S pre-ribosomes to facilitatecleavage at A2. In addition, the presence of Noc4, a component of thesmall subunit processome, in the aberrant pre-40 S particles that accu-mulate in Rps19-depleted cells indicates that dissociation of factors spe-cific to the 90 S particles may also be defective. Together, these dataindicate that Rps19 acts as an essential protein for some of the earlyremodeling steps in the maturation of pre-ribosomes to 40 S ribosomalsubunits.Rps19 haploinsufficiency is thought to give rise to DBA. In many

cases, DBA patients harbor mutations in an RPS19 gene that deletes thegene entirely, interferes with pre-mRNA splicing, or affects translationby introducing frameshifts or premature termination codons (4, 29).Less frequently, DBA patients have been identified that have missensemutations in RPS19. These missense mutations, particularly those thatarise spontaneously, are considered pathogenic if they are not found inthe general population and/or in unaffected family members. As such,they are classified as pathogenic solely on the basis of sequence analysis.The observation, that many of these missense mutations fall in regions

FIGURE 8. Missense mutations found in DBApatients influence rRNA processing and affectthe ability of RPS19A to support growth inyeast. The GAL-RPS19 strain was transformedwith a second plasmid (pRS315) harboring allelesof RPS19A under the control of its own promoter.A, growth assay. Cultures were grown overnight insynthetic media lacking uracil and leucine withgalactose as carbon source, diluted to 0.1 A600,subjected to 10-fold serial dilutions, and spottedon rich media containing galactose (Gal) or glu-cose (Glc). RPS19A alleles carried on the pRS315plasmid are listed above each dilution series. B,Northern analysis. Strains were grown overnight insynthetic media lacking uracil and leucine withgalactose as carbon source, transferred to glu-cose-containing media, and grown for 22 h beforecells were harvested and total RNA prepared. Fil-ters were hybridized with probe A2–A3 and aprobe to mature 18 S rRNA. Values below each lanerepresent the ratio of 21 S pre-rRNA to 18 S rRNAderived from PhosphorImager analysis. The ratioin cells with RPS19A was arbitrarily set at 1. Othervalues were normalized against the RPS19A value.A.U., arbitrary units. C, pre-18 S rRNA FISH with aprobe complementary to the D-A2 segment ofITS1. Strains were grown for 4 h in glucose-con-taining media. Arrows point to the nucleoplasm asvisualized by DNA staining with DAPI. Asterisksindicate the cytoplasm.

Rps19 Is Required for 40 S Ribosomal Subunit Synthesis

38184 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 280 • NUMBER 46 • NOVEMBER 18, 2005

at UL

B on O

ctober 19, 2015http://w

ww

.jbc.org/D

ownloaded from

Page 9: J. Biol. Chem.-2005-Léger-Silvestre-38177-85

ofRPS19 highly conserved between the human and yeast genes, led us toassess the effect of these amino acid substitutions on the function of theyeast protein. Three of the mutations studied, including a novel muta-tion reported here, have measurable effects on Rps19 function in yeast.These missense mutations yield alterations of pre-rRNA processingequivalent to those observed in cells lacking Rps19, indicating that theyresult in the loss of function of the protein. Of the two mutations thatdid not affect the function of the yeast protein, the A62S mutation is nolonger considered pathogenic and, as such, would not be expected toaffect function. The reason why the G121S does not affect the functionof the yeast protein is less clear. It is possible that the glycine to serinecodon change at position 120 in the human sequence may be an innoc-uous polymorphism rather than a pathogenic mutation. Alternatively,the affect of this change on functionmay bemore specific to the humanprotein and not be manifested by the yeast protein. Because the identi-fication of pathogenic mutations in RPS19 is important for assessinggenotype/phenotype correlations in DBA, these data reveal a criticalneed for the ability to monitor Rps19 function in mammalian systems.Other diseases have recently been identified that result from defects

in genes known to affect ribosome synthesis. The X-linked form ofdyskeratosis congenita is caused by mutations in the dyskerin gene,which encodes a pseudouracil synthase involved in ribosomal RNAprocessing and telomere maintenance (30, 31), whereas the TreacherCollins syndrome geneTCOF1 is involved in ribosomal DNA transcrip-tion (32). Each of these diseases, including DBA, has defining character-istics that differ from one another, but all exhibit extreme clinical het-erogeneity with some overlap in secondary characteristics betweendiseases. These data indicate that additional genetic or environmentalfactors significantly influence the nature and course of each disease.Other diseases that may result from defects in ribosome synthesis thatalso show considerable clinical heterogeneity include cartilage hair hyp-oplasia (33) and Shwachman-Diamond syndrome (34, 35). Thus, thefailure to synthesize normal amounts of ribosomal subunits is a contrib-uting factor to the clinical profile of a growing number of humandiseases.

Acknowledgments—We thank Valerie Choesmel, Sebastien Ferreira-Cerca,Cecile Guinefoleau, and Marlene Faubladier for their participation to thisstudy.

REFERENCES1. Freedman, M. H. (2000) Baillieres Clin. Hematol. 13, 391–4062. Willig, T.-N., Gazda, H., and Sieff, C. A. (2000) Hematology 7, 85–943. Draptchinskaia, N., Gustavsson, P., Andersson, B., Pettersson, M., Willig, T. N., Di-

anzani, I., Ball, S., Tchernia, G., Klar, J., Matsson, H., Tentler, D., Mohandas, N.,Carlsson, B., and Dahl, N. (1999) Nat. Genet. 21, 169–175

4. Willig, T. N., Draptchinskaia, N., Dianzani, I., Ball, S., Niemeyer, C., Ramenghi, U.,Orfali, K., Gustavsson, P., Garelli, E., Brusco, A., Tiemann, C., Perignon, J. L.,Bouchier, C., Cicchiello, L., Mohandas, N., and Tchernia, G. (1999) Blood 94,4294–4306

5. Nomura, M. (1990) in The Ribosome: Structure, Function, and Evolution (Hill, W. E.,

Dahlberg, A., Garrett, R. A., Moore, P. B., Schlessinger, D., andWarner, J. R., eds) pp.3–55, American Society for Microbiology, Washington, D. C.

6. Baudin-Baillieu, A., Tollervey, D., Cullin, C., and Lacroute, F. (1997) Mol. Cell. Biol.17, 5023–5032

7. Ford, C. L., Randall-Whitis, L., and Ellis, S. R. (1999) Cancer Res. 59, 704–7108. Tabb-Massey, A., Caffrey, J. M., Logsden, P., Taylor, S., Trent, J. O., and Ellis, S.R.

(2003) Nucleic Acids Res. 31, 6798–68059. Jakovljevic, J., deMayolo, P. A.,Miles, T.D.,Nguyen, T.M., Leger-Silvestre, I., Gas,N.,

and Woolford, J. L. (2004)Mol. Cell 14, 331–34210. Leger-Silvestre, I., Milkereit, P., Ferreira-Cerca, S., Saveanu, C., Rousselle, J.-C.,

Choesmel, V., Guinefoleau, C., Gas, N., and Gleizes, P. -E. (2004) EMBO J. 23,2336–2347

11. Fatica, A., and Tollervey, D. (2002) Curr. Opin. Cell Biol. 14, 313–31812. Fromont-Racine M., Senger B., Saveanu C., and Fasiolo F. (2003) Gene 313, 17–4213. Tschochner H., and Hurt E. (2003) Trends Cell Biol. 13, 255–26314. Venema, J., and Tollervey, D. (1999) Annu. Rev. Genet. 33, 261–31115. Milkereit, P., Kuhn, T., Gas, N., and Tschochner, H. (2003) Nucleic Acids Res. 31,

799–80416. Schafer, T., Strauss, D., Petfalski, E., Tollervey, D., and Hurt, E. (2003) EMBO J. 22,

1370–138017. Sherman, F. (1991)Methods Enzymol. 194, 1–2118. Baim, S. B., Pietras, D. F., Eustice, D. C., and Sherman, F. (1985) Mol. Cell. Biol. 5,

1839–184619. Schmitt, M. E., Brown, T. A., and Trumpower, B. L. (1990) Nucleic Acids Res. 18,

3091–309220. Dez, C., Noaillac-Depeyre, J., Caizergues-Ferrer, M., and Henry, Y. (2002)Mol. Cell.

Biol. 22, 7053–706521. Gleizes, P. E., Noaillac-Depeyre, J., Leger-Silvestre, I., Teulieres, F., Dauxois, J. Y.,

Pommet, D., Azum-Gelade, M. C., and Gas, N. (2001) J. Cell Biol. 155, 923–93622. Demianova, M., Formosa, T. G., and Ellis, S. R. (1996) J. Biol. Chem. 271,

11383–1139123. Roberts, P., Moshitch-Moshkovitz, S., Kvam, E., O’Toole, E., Winey, M., and Gold-

farb, D. S. (2003)Mol. Biol. Cell 14, 129–14124. Matsson, H., Davey, E. J., Draptchinskaia, N., Hamaguchi, I., Ooka, A., Leveen, P.,

Forsberg, E., Karlsson, S., and Dahl, N. (2004)Mol. Cell. Biol. 24, 4032–403725. Torchet, C., and Hermann-Le Denmat, S. (2000) RNA (N. Y.) 6, 1498–150826. Vos,H. R., Bax, R, Faber, A.W., Vos, J. C., andRaue,H.A. (2004)NucleicAcids Res.32,

5827–583327. Vanrobays, E., Gelugne, J. P., Gleizes, P. E., and Caizergues-Ferrer, M. (2003) Mol.

Cell. Biol. 23, 2083–209528. Chen,W., Bucaria, J., Band, D. A., Sutton, A., and Sternglanz, R. (2003)Nucleic Acids

Res. 31, 690–69929. Gazda, H. T., Zhong, R., Long, L., Niewiadomska, E., Lipton, J. L., Ploszynska, A.,

Zaucha, J.M., Vlachos, A., Atsidaflos, E., Viskochil, D. H., Niemeyer, C.M.,Meerpohl,J. J., Rokicka-Milewska, R., Pospisilova, D., Wiktor-Jedrzejczak, W., Nathan, D. G.,Beggs, A. H., and Sieff, C. A. (2004) Br. J. Hemaetol. 127, 105–113

30. Ruggero, D., Grisendi, S., Piazza, F., Rego, E., Mari, F., Rao, P. H., Cordon-Cardo, C.,and Pandolfi, P. P. (2003) Science 299, 259–262

31. Mochizuki, Y., He, J., Kulkarni, S., Bessler, M., and Mason, P. J. (2004) Proc. Natl.Acad. Sci. U. S. A. 101, 10756–10761

32. Valdez, B. C.,Henning,D., So, R. B., Dixon, J., andDixon,M. J. (2004)Proc.Natl. Acad.Sci. U. S. A. 101, 10709–10714

33. Ridanpaa, M., van Eenennaam, H., Pelin, K., Chadwick, R., Johnson, C., Yuan, B., vanVenrooij, W., Pruijn, G., Salmela, R., Rockas, S., Makitie, O., Kaitila, I., and de laChapelle, A. (2001) Cell 104, 195–203

34. Rothbaum,R., Perrault, J., Vlachos, A., Cipolli,M., Alter, B. P., Burroughs, S., Durie, P.,Elghtany, M. T., Grand, R., Hubbard, V., Rommens, J., and Rossi, T. (2002) J. Pediatr.141, 266–270

35. Boocock, G. R., Morrison, J. A., Popovic, M., Richards, N., Ellis, L., Durie, P. R., andRommens, J. M. (2003) Nat. Genet. 33, 97–101

Rps19 Is Required for 40 S Ribosomal Subunit Synthesis

NOVEMBER 18, 2005 • VOLUME 280 • NUMBER 46 JOURNAL OF BIOLOGICAL CHEMISTRY 38185

at UL

B on O

ctober 19, 2015http://w

ww

.jbc.org/D

ownloaded from

Page 10: J. Biol. Chem.-2005-Léger-Silvestre-38177-85

Steven Robert EllisBertolone, Pierre-Emmanuel Gleizes andAlvarez-Arias, Nicole Gas, Salvatore J.

AlehandrovnaCaffrey, Rosy Dawaliby, Diana Isabelle Léger-Silvestre, Jacqueline Marie  Rps19 Protein in Ribosome SynthesisDiamond Blackfan Anemia-associated Specific Role for Yeast Homologs of theModification, and Degradation:Protein Synthesis, Post-Translation

doi: 10.1074/jbc.M506916200 originally published online September 12, 20052005, 280:38177-38185.J. Biol. Chem. 

  10.1074/jbc.M506916200Access the most updated version of this article at doi:

  .JBC Affinity SitesFind articles, minireviews, Reflections and Classics on similar topics on the

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

  http://www.jbc.org/content/280/46/38177.full.html#ref-list-1

This article cites 34 references, 20 of which can be accessed free at

at UL

B on O

ctober 19, 2015http://w

ww

.jbc.org/D

ownloaded from