Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Article in English | MEDLINE | ID: mdl-34725166

ABSTRACT

Small nucleolar (sno)RNAs guide posttranscriptional modifications essential for the biogenesis and function of their target. The majority of snoRNAs in higher eukaryotes are encoded within introns. They are first released from nascent transcripts in the form of a lariat and rapidly targeted by the debranching enzyme and nuclear exonucleases for linearization and further trimming. In this study, we report that some snoRNAs are encoded within unusually stable intronic RNAs. These intronic sequences can escape the debranching enzyme and accumulate as lariats. Stable lariats bearing a snoRNA, or slb-snoRNA, are associated with snoRNA binding proteins but do not guide posttranscriptional modification. While most slb-snoRNAs accumulate in the nucleus, some can be exported to the cytoplasm. We find that this export competes with snoRNA maturation. Slb-snoRNAs provide a previously unknown layer of regulation to snoRNA and snoRNA binding proteins.


Subject(s)
RNA, Circular/metabolism , RNA, Small Nucleolar/metabolism , 3T3 Cells , Animals , Female , Gene Expression Regulation , HeLa Cells , Humans , Introns , Mice , RNA, Guide, Kinetoplastida , Saccharomyces cerevisiae , Xenopus laevis
2.
RNA ; 27(11): 1363-1373, 2021 11.
Article in English | MEDLINE | ID: mdl-34385348

ABSTRACT

In eukaryotes, rRNAs and spliceosomal snRNAs are heavily modified post-transcriptionally. Pseudouridylation and 2'-O-methylation are the most abundant types of RNA modifications. They are mediated by modification guide RNAs, also known as small nucleolar (sno)RNAs and small Cajal body-specific (sca)RNAs. We used yeast and vertebrate cells to test guide activities predicted for a number of snoRNAs, based on their regions of complementarity with rRNAs. We showed that human SNORA24 is a genuine guide RNA for 18S-Ψ609, despite some noncanonical base-pairing with its target. At the same time, we found quite a few snoRNAs that have the ability to base-pair with rRNAs and can induce predicted modifications in artificial substrate RNAs, but do not modify the same target sequence within endogenous rRNA molecules. Furthermore, certain fragments of rRNAs can be modified by the endogenous yeast modification machinery when inserted into an artificial backbone RNA, even though the same sequences are not modified in endogenous yeast rRNAs. In Xenopus cells, a guide RNA generated from scaRNA, but not from snoRNA, could induce an additional pseudouridylation of U2 snRNA at position 60; both guide RNAs were equally active on a U2 snRNA-specific substrate in yeast cells. Thus, post-transcriptional modification of functionally important RNAs, such as rRNAs and snRNAs, is highly regulated and more complex than simply strong base-pairing between a guide RNA and substrate RNA. We discuss possible regulatory roles for these unexpected modifications.


Subject(s)
Coiled Bodies/metabolism , Pseudouridine/metabolism , RNA Processing, Post-Transcriptional , RNA, Guide, Kinetoplastida/metabolism , RNA, Small Nucleolar/metabolism , Spliceosomes/metabolism , Base Sequence , Coiled Bodies/genetics , Humans , Methylation , RNA, Guide, Kinetoplastida/genetics , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA, Small Nucleolar/chemistry , RNA, Small Nucleolar/genetics , Spliceosomes/genetics
3.
Genes Dev ; 35(15-16): 1123-1141, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34301768

ABSTRACT

Spliceosomal small nuclear RNAs (snRNAs) are modified by small Cajal body (CB)-specific ribonucleoproteins (scaRNPs) to ensure snRNP biogenesis and pre-mRNA splicing. However, the function and subcellular site of snRNA modification are largely unknown. We show that CB localization of the protein Nopp140 is essential for concentration of scaRNPs in that nuclear condensate; and that phosphorylation by casein kinase 2 (CK2) at ∼80 serines targets Nopp140 to CBs. Transiting through CBs, snRNAs are apparently modified by scaRNPs. Indeed, Nopp140 knockdown-mediated release of scaRNPs from CBs severely compromises 2'-O-methylation of spliceosomal snRNAs, identifying CBs as the site of scaRNP catalysis. Additionally, alternative splicing patterns change indicating that these modifications in U1, U2, U5, and U12 snRNAs safeguard splicing fidelity. Given the importance of CK2 in this pathway, compromised splicing could underlie the mode of action of small molecule CK2 inhibitors currently considered for therapy in cholangiocarcinoma, hematological malignancies, and COVID-19.


Subject(s)
Interstitial Cells of Cajal/metabolism , Methylation , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , RNA Splicing , RNA, Small Nuclear/metabolism , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/metabolism , Cholangiocarcinoma/drug therapy , Hematologic Neoplasms/drug therapy , Humans , Phosphorylation , RNA, Small Nuclear/chemistry , Ribonucleoproteins/metabolism , Spliceosomes/genetics , COVID-19 Drug Treatment
4.
bioRxiv ; 2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33948588

ABSTRACT

Spliceosomal small nuclear RNAs (snRNAs) are modified by small Cajal body (CB) specific ribonucleoproteins (scaRNPs) to ensure snRNP biogenesis and pre-mRNA splicing. However, the function and subcellular site of snRNA modification are largely unknown. We show that CB localization of the protein Nopp140 is essential for concentration of scaRNPs in that nuclear condensate; and that phosphorylation by casein kinase 2 (CK2) at some 80 serines targets Nopp140 to CBs. Transiting through CBs, snRNAs are apparently modified by scaRNPs. Indeed, Nopp140 knockdown-mediated release of scaRNPs from CBs severely compromises 2'-O-methylation of spliceosomal snRNAs, identifying CBs as the site of scaRNP catalysis. Additionally, alternative splicing patterns change indicating that these modifications in U1, U2, U5, and U12 snRNAs safeguard splicing fidelity. Given the importance of CK2 in this pathway, compromised splicing could underlie the mode of action of small molecule CK2 inhibitors currently considered for therapy in cholangiocarcinoma, hematological malignancies, and COVID-19.

5.
Exp Cell Res ; 401(2): 112523, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33675804

ABSTRACT

The lampbrush chromosomes (LBCs) in oocytes of the Mexican axolotl (Ambystoma mexicanum) were identified some time ago by their relative lengths and predicted centromeres, but they have never been associated completely with the mitotic karyotype, linkage maps or genome assembly. We identified 9 of the axolotl LBCs using RNAseq to identify actively transcribed genes and 13 BAC (bacterial artificial clone) probes containing pieces of active genes. Using read coverage analysis to find candidate centromere sequences, we developed a centromere probe that localizes to all 14 centromeres. Measurements of relative LBC arm lengths and polymerase III localization patterns enabled us to identify all LBCs. This study presents a relatively simple and reliable way to identify each axolotl LBC cytologically and to anchor chromosome-length sequences (from the axolotl genome assembly) to the physical LBCs by immunostaining and fluorescence in situ hybridization. Our data will facilitate a more detailed transcription analysis of individual LBC loops.


Subject(s)
Ambystoma mexicanum/genetics , Centromere/ultrastructure , Chromosomes/genetics , In Situ Hybridization, Fluorescence , Transcription, Genetic , Ambystoma mexicanum/immunology , Animals , Centromere/genetics , Chromosome Mapping , Chromosomes/immunology , Chromosomes, Artificial, Bacterial/genetics , Chromosomes, Artificial, Bacterial/immunology , Oocytes/growth & development , Oocytes/ultrastructure
6.
Exp Cell Res ; 398(1): 112386, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33220259

ABSTRACT

Here we describe novel spherical structures that are induced by cold shock on the lampbrush chromosomes (LBCs) of Xenopus laevis oocytes. We call these structures cold bodies or C-bodies. C-bodies are distributed symmetrically on homologous LBCs, with a pattern similar to that of 5S rDNA. Neither active transcription nor translation is necessary for their formation. Similar protrusions occur on the edges of some nucleoli. Endogenous LBCs as well as those derived from injected sperm form C-bodies under cold shock conditions. The function of C-bodies is unknown.


Subject(s)
Cell Nucleus Structures/ultrastructure , Cell Nucleus/ultrastructure , Chromosomes/ultrastructure , Cold-Shock Response , Oocytes/ultrastructure , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus Structures/metabolism , Chromosomes/genetics , Female , Oocytes/metabolism , Xenopus laevis
7.
J Cell Biol ; 219(5)2020 05 04.
Article in English | MEDLINE | ID: mdl-32211899

ABSTRACT

The synaptonemal complex (SC) is a tripartite protein scaffold that forms between homologous chromosomes during meiosis. Although the SC is essential for stable homologue pairing and crossover recombination in diverse eukaryotes, it is unknown how individual components assemble into the highly conserved SC structure. Here we report the biochemical identification of two new SC components, SYP-5 and SYP-6, in Caenorhabditis elegans. SYP-5 and SYP-6 are paralogous to each other and play redundant roles in synapsis, providing an explanation for why these genes have evaded previous genetic screens. Superresolution microscopy reveals that they localize between the chromosome axes and span the width of the SC in a head-to-head manner, similar to the orientation of other known transverse filament proteins. Using genetic redundancy and structure-function analyses to truncate C-terminal tails of SYP-5/6, we provide evidence supporting the role of SC in both limiting and promoting crossover formation.


Subject(s)
Caenorhabditis elegans/genetics , Chromosomal Proteins, Non-Histone/genetics , Recombination, Genetic/genetics , Synaptonemal Complex/genetics , Animals , Chromosome Pairing/genetics , Chromosomes/genetics , Crossing Over, Genetic/genetics , Meiosis/genetics , Mutation/genetics
8.
Nat Protoc ; 15(3): 1188-1208, 2020 03.
Article in English | MEDLINE | ID: mdl-32051613

ABSTRACT

During DNA replication, the genetic information of a cell is copied. Subsequently, identical genetic information is segregated reliably to the two daughter cells through cell division. Meanwhile, DNA replication is intrinsically linked to the process of chromatin duplication, which is required for regulating gene expression and establishing cell identities. Understanding how chromatin is established, maintained or changed during DNA replication represents a fundamental question in biology. Recently, we developed a method to directly visualize chromatin components at individual replication forks undergoing DNA replication. This method builds upon the existing chromatin fiber technique and combines it with cell type-specific chromatin labeling and superresolution microscopy. In this method, a short pulse of nucleoside analog labels replicative regions in the cells of interest. Chromatin fibers are subsequently isolated and attached to a glass slide, after which a laminar flow of lysis buffer extends the lysed chromatin fibers parallel with the direction of the flow. Fibers are then immunostained for different chromatin-associated proteins and mounted for visualization using superresolution microscopy. Replication foci, or 'bubbles,' are identified by the presence of the incorporated nucleoside analog. For researchers experienced in molecular biology and superresolution microscopy, this protocol typically takes 2-3 d from sample preparation to data acquisition, with an additional day for data processing and quantification.


Subject(s)
Chromatin , DNA Replication/physiology , DNA/genetics , Epigenesis, Genetic/physiology , Animals , Cell Line , Drosophila melanogaster , Microscopy, Fluorescence/methods , Optical Imaging/methods , Single Molecule Imaging/methods
9.
Mol Biol Evol ; 37(1): 149-166, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31553476

ABSTRACT

Small nucleolar RNAs (snoRNAs) function primarily as guide RNAs for posttranscriptional modification of rRNAs and spliceosomal snRNAs, both of which are functionally important and evolutionarily conserved molecules. It is commonly believed that snoRNAs and the modifications they mediate are highly conserved across species. However, most relevant data on snoRNA annotation and RNA modification are limited to studies on human and yeast. Here, we used RNA-sequencing data from the giant oocyte nucleus of the frog Xenopus tropicalis to annotate a nearly complete set of snoRNAs. We compared the frog data with snoRNA sets from human and other vertebrate genomes, including mammals, birds, reptiles, and fish. We identified many Xenopus-specific (or nonhuman) snoRNAs and Xenopus-specific domains in snoRNAs from conserved RNA families. We predicted that some of these nonhuman snoRNAs and domains mediate modifications at unexpected positions in rRNAs and snRNAs. These modifications were mapped as predicted when RNA modification assays were applied to RNA from nine vertebrate species: frogs X. tropicalis and X. laevis, newt Notophthalmus viridescens, axolotl Ambystoma mexicanum, whiptail lizard Aspidoscelis neomexicana, zebrafish Danio rerio, chicken, mouse, and human. This analysis revealed that only a subset of RNA modifications is evolutionarily conserved and that modification patterns may vary even between closely related species. We speculate that each functional domain in snoRNAs (half of an snoRNA) may evolve independently and shuffle between different snoRNAs.


Subject(s)
RNA, Small Nucleolar/genetics , Xenopus/genetics , Animals , Genome , Humans , Molecular Sequence Annotation , Point Mutation
10.
Genes Dev ; 33(23-24): 1617-1618, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31792016

ABSTRACT

Although the nucleolus was first described in the early 19th century from both animal and plant cells, human nucleoli and particularly the five human nucleolus organizers have not been well characterized. In this issue of Genes & Development, van Sluis and colleagues (pp. 1688-1701) present a detailed molecular analysis of these organizers, which occur on the short arms of five human chromosomes. The near identity of these arms suggests extensive interchromosomal exchange during evolutionary history.


Subject(s)
Cell Nucleolus , Nucleolus Organizer Region , Animals , DNA, Ribosomal , Humans
11.
Biomolecules ; 9(9)2019 09 07.
Article in English | MEDLINE | ID: mdl-31500270

ABSTRACT

Site-specific 2'-O-ribose methylation is an abundant post-transcriptional modification mediated by small non-coding nuclear RNAs known as box C/D modification guide RNAs. The minimal structural requirements for these guide RNAs to function in higher eukaryotes are still unclear. To address this question, we generated a series of mutant variants of Drosophila box C/D scaRNA:MeU2-C28 and tested their modification guide activities in the Xenopus oocyte system. Our data suggest that box C/D guide RNA function requires either a terminal or an internal consensus kink-turn structure. We identified the minimal functional box C/D guide RNA. It consists of a single-domain molecule with (i) a terminal stem with a consensus kink-turn domain, (ii) one box C and box D connected by a 14-nucleotide antisense element and (iii) a one-nucleotide spacer between the box C and the antisense element. In this single domain RNA, the sequence of the spacer is more important than its length. We suggest that the secondary structure of box C/D RNAs, essential for guide RNA function, is more complex than generally supposed. At the same time, the expression of functional extremely short single-domain box C/D RNAs is possible in higher eukaryotes.


Subject(s)
RNA, Guide, Kinetoplastida/metabolism , RNA, Long Noncoding/metabolism , Animals , Female , Nucleic Acid Conformation , RNA, Long Noncoding/chemistry , Xenopus laevis
12.
RNA ; 25(1): 17-22, 2019 01.
Article in English | MEDLINE | ID: mdl-30301832

ABSTRACT

Posttranscriptional modifications of rRNA occur in the nucleolus where rRNA modification guide RNAs, or snoRNAs, concentrate. On the other hand, scaRNAs, the modification guide RNAs for spliceosomal snRNAs, concentrate in the Cajal body (CB). It is generally assumed, therefore, that snRNAs must accumulate in CBs to be modified by scaRNAs. Here we demonstrate that the evidence for the latter postulate is not consistent. In the nucleus, scaRNA localization is not limited to CBs. Furthermore, canonical scaRNAs can modify rRNAs. We suggest that the conventional view that scaRNAs function only in the CB needs revision.


Subject(s)
Coiled Bodies/metabolism , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/metabolism , RNA, Small Nucleolar/chemistry , RNA, Small Nucleolar/metabolism , Animals , Base Sequence , HeLa Cells , Humans , Nucleic Acid Conformation , RNA Processing, Post-Transcriptional , RNA, Guide, Kinetoplastida/genetics , RNA, Ribosomal/chemistry , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , RNA, Small Nucleolar/genetics , Spliceosomes/genetics , Spliceosomes/metabolism , Xenopus/genetics , Xenopus/metabolism
14.
Proc Natl Acad Sci U S A ; 115(34): E7970-E7977, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30082412

ABSTRACT

Most intronic RNAs are degraded within seconds or minutes after their excision from newly formed transcripts. However, stable intronic sequence RNAs (sisRNAs) have been described from oocytes of the frog Xenopus, from Drosophila embryos, and from human cell lines. In Xenopus oocytes, sisRNAs are abundant in both the nucleus and cytoplasm, they occur in the form of lariats, and they are stable for days. In this study we demonstrate that cytoplasmic sisRNAs are also found in human, mouse, chicken, and zebrafish cells. They exist as circular (lariat) molecules, mostly 100-500 nucleotides in length, and are derived from many housekeeping genes. They tend to have an unusual cytosine branchpoint (with the exception of those from the frog). Stable lariats are exported from the nucleus to the cytoplasm by the NXF1/NXT1 system, demonstrating that their presence in the cytoplasm is not due to passive diffusion. Lariats in the cytoplasm are not associated with transcripts of the genes from which they are derived. The biological significance of cytoplasmic sisRNAs remains obscure.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , RNA, Untranslated/metabolism , 3T3 Cells , Active Transport, Cell Nucleus/physiology , Animals , Cell Nucleus/genetics , Chickens , Cytoplasm/genetics , Drosophila , HeLa Cells , Humans , Mice , RNA, Untranslated/genetics , Xenopus , Zebrafish
15.
RNA ; 24(7): 908-914, 2018 07.
Article in English | MEDLINE | ID: mdl-29686135

ABSTRACT

We report that 7SL, the RNA component of the signal recognition particle (SRP), is an abundant noncoding RNA (ncRNA) in mature red blood cells (RBCs) of human, mouse, and the frog Xenopus. 7SL RNA in RBCs is not associated with the canonical proteins of the SRP. Instead, it coimmunoprecipitates from a lysate of RBCs with a number of membrane-binding proteins. Human and mouse RBCs also contain a previously undescribed 68 nt RNA, sRN7SL, derived from the "S domain" of 7SL RNA. We discuss the possibility that 7SL RNA is selectively protected from nucleases by association with the RBC membrane. Because 7SL is not associated with the canonical proteins of the SRP, it could represent a nonfunctional remnant of the protein synthetic machinery. Alternatively, it could play a new, as yet undefined role in RBC metabolism.


Subject(s)
Erythrocytes/metabolism , RNA, Small Cytoplasmic/blood , Signal Recognition Particle/blood , Animals , Humans , Mice , Proteins/metabolism , RNA, Small Cytoplasmic/metabolism , Signal Recognition Particle/metabolism , Xenopus
16.
RNA ; 24(1): 30-42, 2018 01.
Article in English | MEDLINE | ID: mdl-28974555

ABSTRACT

The branch point recognition region of spliceosomal snRNA U2 is heavily modified post-transcriptionally in most eukaryotic species. We focused on this region to learn how nearby positions may interfere with each other when targeted for modification. Using an in vivo yeast Saccharomyces cerevisiae cell system, we tested the modification activity of several guide RNAs from human, mouse, the frog Xenopus tropicalis, the fruit fly Drosophila melanogaster, and the worm Caenorhabditis elegans We experimentally verified predictions for vertebrate U2 modification guide RNAs SCARNA4 and SCARNA15, and identified a C. elegans ortholog of SCARNA15. We observed crosstalk between sites in the heavily modified regions, such that modification at one site may inhibit modification at nearby sites. This is true for the branch point recognition region of U2 snRNA, the 5' loop of U5 snRNA, and certain regions of rRNAs, when tested either in yeast or in HeLa cells. The position preceding a uridine targeted for isomerization by a box H/ACA guide RNA is the most sensitive for noncanonical base-pairing and modification (either pseudouridylation or 2'-O-methylation). Based on these findings, we propose that modification must occur stepwise starting with the most vulnerable positions and ending with the most inhibiting modifications. We discuss possible strategies that cells use to reach complete modification in heavily modified regions.


Subject(s)
RNA Processing, Post-Transcriptional , RNA, Small Nuclear/genetics , Animals , Base Sequence , Binding Sites , Caenorhabditis elegans , Drosophila melanogaster , HeLa Cells , Humans , Mice , Pseudouridine/metabolism , RNA, Small Nuclear/metabolism , Saccharomyces cerevisiae , Xenopus
17.
RNA ; 23(7): 1060-1067, 2017 07.
Article in English | MEDLINE | ID: mdl-28432181

ABSTRACT

The pseudouridine at position 43 in vertebrate U2 snRNA is one of the most conserved post-transcriptional modifications of spliceosomal snRNAs; the equivalent position is pseudouridylated in U2 snRNAs in different phyla including fungi, insects, and worms. Pseudouridine synthase Pus1p acts alone on U2 snRNA to form this pseudouridine in yeast Saccharomyces cerevisiae and mouse. Furthermore, in S. cerevisiae, Pus1p is the only pseudouridine synthase for this position. Using an in vivo yeast cell system, we tested enzymatic activity of Pus1p from the fission yeast Schizosaccharomyces pombe, the worm Caenorhabditis elegans, the fruit fly Drosophila melanogaster, and the frog Xenopus tropicalis We demonstrated that Pus1p from C. elegans has no enzymatic activity on U2 snRNA when expressed in yeast cells, whereas in similar experiments, position 44 in yeast U2 snRNA (equivalent to position 43 in vertebrates) is a genuine substrate for Pus1p from S. cerevisiae, S. pombe, Drosophila, Xenopus, and mouse. However, when we analyzed U2 snRNAs from Pus1 knockout mice and the pus1Δ S. pombe strain, we could not detect any changes in their modification patterns when compared to wild-type U2 snRNAs. In S. pombe, we found a novel box H/ACA RNA encoded downstream from the RPC10 gene and experimentally verified its guide RNA activity for positioning Ψ43 and Ψ44 in U2 snRNA. In vertebrates, we showed that SCARNA8 (also known as U92 scaRNA) is a guide for U2-Ψ43 in addition to its previously established targets U2-Ψ34/Ψ44.


Subject(s)
Intramolecular Transferases/metabolism , RNA, Guide, Kinetoplastida/metabolism , RNA, Small Nuclear/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Knockout Techniques , Intramolecular Transferases/genetics , Mice , Pseudouridine/metabolism , RNA, Fungal/metabolism , RNA, Small Nuclear/chemistry , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Xenopus/genetics , Xenopus/metabolism
18.
Proc Natl Acad Sci U S A ; 114(7): E1111-E1117, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28069952

ABSTRACT

The toxic proline:arginine (PRn) poly-dipeptide encoded by the (GGGGCC)n repeat expansion in the C9orf72 form of heritable amyotrophic lateral sclerosis (ALS) binds to the central channel of the nuclear pore and inhibits the movement of macromolecules into and out of the nucleus. The PRn poly-dipeptide binds to polymeric forms of the phenylalanine:glycine (FG) repeat domain, which is shared by several proteins of the nuclear pore complex, including those in the central channel. A method of chemical footprinting was used to characterize labile, cross-ß polymers formed from the FG domain of the Nup54 protein. Mutations within the footprinted region of Nup54 polymers blocked both polymerization and binding by the PRn poly-dipeptide. The aliphatic alcohol 1,6-hexanediol melted FG domain polymers in vitro and reversed PRn-mediated enhancement of the nuclear pore permeability barrier. These data suggest that toxicity of the PRn poly-dipeptide results in part from its ability to lock the FG repeats of nuclear pore proteins in the polymerized state. Our study offers a mechanistic interpretation of PRn poly-dipeptide toxicity in the context of a prominent form of ALS.


Subject(s)
Active Transport, Cell Nucleus , C9orf72 Protein/pharmacology , DNA Repeat Expansion/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Biopolymers , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Dipeptides/genetics , Dipeptides/metabolism , Dipeptides/pharmacology , Female , Glycols/pharmacology , Humans , Microscopy, Confocal , Nuclear Pore/chemistry , Nuclear Pore/drug effects , Nuclear Pore/ultrastructure , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/ultrastructure , Oocytes/ultrastructure , Permeability/drug effects , Protein Binding , Protein Domains , Wheat Germ Agglutinins/metabolism , Wheat Germ Agglutinins/pharmacology , Xenopus laevis
20.
Genetica ; 144(1): 107-24, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26790663

ABSTRACT

This paper reports genome sizes of one Hawaiian Scaptomyza and 16 endemic Hawaiian Drosophila species that include five members of the antopocerus species group, one member of the modified mouthpart group, and ten members of the picture wing clade. Genome size expansions have occurred independently multiple times among Hawaiian Drosophila lineages, and have resulted in an over 2.3-fold range of genome sizes among species, with the largest observed in Drosophila cyrtoloma (1C = 0.41 pg). We find evidence that these repeated genome size expansions were likely driven by the addition of significant amounts of heterochromatin and satellite DNA. For example, our data reveal that the addition of seven heterochromatic chromosome arms to the ancestral haploid karyotype, and a remarkable proportion of ~70 % satellite DNA, account for the greatly expanded size of the D. cyrtoloma genome. Moreover, the genomes of 13/17 Hawaiian picture wing species are composed of substantial proportions (22-70 %) of detectable satellites (all but one of which are AT-rich). Our results suggest that in this tightly knit group of recently evolved species, genomes have expanded, in large part, via evolutionary amplifications of satellite DNA sequences in centric and pericentric domains (especially of the X and dot chromosomes), which have resulted in longer acrocentric chromosomes or metacentrics with an added heterochromatic chromosome arm. We discuss possible evolutionary mechanisms that may have shaped these patterns, including rapid fixation of novel expanded genomes during founder-effect speciation.


Subject(s)
Biological Evolution , Drosophila/genetics , Genome Size , Genome, Insect , Animals , Body Size , DNA, Satellite/genetics , Female , Hawaii , Heterochromatin , Karyotyping , Male , Wings, Animal
SELECTION OF CITATIONS
SEARCH DETAIL
...