Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 332
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(49): e2208707119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36445968

ABSTRACT

Pathogenic variants in the Retinitis pigmentosa GTPase regulator (RPGR) gene lead to a clinically severe form of X-linked retinal dystrophy. However, it remains unclear why some variants cause a predominant rod, while others result in a cone-dominated phenotype. Post-translational glutamylation of the photoreceptor-specific RPGRORF15 isoform by the TTLL5 enzyme is essential for its optimal function in photoreceptors, and loss of TTLL5 leads to retinal dystrophy with a cone phenotype. Here we show that RPGR retinal disease, studied in a single cohort of 116 male patients, leads to a clear progressive shift from rod- to cone-dominating phenotype as the RPGRORF15 variant location approaches the distal part of the Open Reading Frame 15 (ORF15) region. The rod photoreceptor involvement on the contrary diminishes along the RGPR sequence, and the variants associated with the cone only phenotype are located predominantly in the very distal part, including the C-terminal basic domain. Moreover, these distal truncating RPGRORF15 variants disrupt the interaction with TTLL5 and lead to a significant impairment of RPGR glutamylation. Thus, consistent with the phenotype of TTLL5 pathogenic variants, our study shows that RPGRORF15 variants, which disrupt its basic domain and the interaction with TTLL5, also impair RPGR glutamylation and lead to the cone phenotype. This has implications for ongoing gene therapy clinical trials where the application of RPGR with impaired glutamylation may be less effective in treating RGPR dystrophies and may even convert a rod-cone dystrophy into a cone dystrophy phenotype.


Subject(s)
Cone-Rod Dystrophies , Retinal Dystrophies , Humans , Male , Cone-Rod Dystrophies/genetics , Cone-Rod Dystrophies/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Open Reading Frames/genetics , Open Reading Frames/physiology , Phenotype , Retinal Cone Photoreceptor Cells/metabolism , Retinal Dystrophies/genetics , Retinal Dystrophies/metabolism , Glutamic Acid/metabolism
2.
PLoS Pathog ; 16(5): e1008488, 2020 05.
Article in English | MEDLINE | ID: mdl-32433693

ABSTRACT

There are approximately 20 million events of hepatitis E virus (HEV) infection worldwide annually. The genome of HEV is a single-strand, positive-sense RNA containing 5' and 3' untranslated regions and three open reading frames (ORF). HEV genome has 5' cap and 3' poly(A) tail to mimic host mRNA to escape the host innate immune surveillance and utilize host translational machineries for viral protein translation. The replication mechanism of HEV is poorly understood, especially how the viral polymerase distinguishes viral RNA from host mRNA to synthesize new viral genomes. We hypothesize that the HEV genome contains cis-acting elements that can be recognized by the virally encoded polymerase as "self" for replication. To identify functional cis-acting elements systematically across the HEV genome, we utilized an ORF1 transcomplementation system. Ultimately, we found two highly conserved cis-acting RNA elements within the ORF1 and ORF2 coding regions that are required for viral genome replication in a diverse panel of HEV genotypes. Synonymous mutations in the cis-acting RNA elements, not altering the ORF1 and ORF2 protein sequences, significantly impaired production of infectious viral particles. Mechanistic studies revealed that the cis-acting elements form secondary structures needed to interact with the HEV ORF1 protein to promote HEV replication. Thus, these cis-acting elements function as a scaffold, providing a specific "signal" that recruits viral and host factors to assemble the viral replication complex. Altogether, this work not only facilitates our understanding of the HEV life cycle and provides novel, RNA-directed targets for potential HEV treatments, but also sheds light on the development of HEV as a therapeutic delivery vector.


Subject(s)
Genome, Viral , Hepatitis E virus/physiology , RNA, Viral , Regulatory Sequences, Ribonucleic Acid/physiology , Virus Replication/physiology , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mutation , Open Reading Frames/physiology , RNA, Viral/biosynthesis , RNA, Viral/genetics
3.
J Virol ; 94(24)2020 11 23.
Article in English | MEDLINE | ID: mdl-32967952

ABSTRACT

Although astroviruses causes enteric diseases and encephalitis in humans and nephritis and hepatitis in poultry, astrovirus infection is thought to be self-limiting. However, little is known about its molecular mechanism. In this study, we found that a novel goose astrovirus (GAstV), GAstV-GD, and its open reading frame 2 (ORF2) could efficiently activate the innate immune response and induce a high level of OASL in vitro and in vivo The truncation assay for ORF2 further revealed that the P2 domain of ORF2 contributed to stimulating OASL, whereas the acidic C terminus of ORF2 attenuated such activation. Moreover, the overexpression and knockdown of OASL could efficiently restrict and promote the viral replication of GAstV-GD, respectively. Our data not only give novel insights for elucidating self-limiting infection by astrovirus but also provide virus and host targets for fighting against astroviruses.IMPORTANCE Astroviruses cause gastroenteritis and encephalitis in human, and nephritis, hepatitis, and gout disease in poultry. However, the host immune response activated by astrovirus is mostly unknown. Here, we found that a novel goose astrovirus, GAstV-GD, and its ORF2 protein could efficiently induce a high level of OASL in vitro and in vivo, which could feed back to restrict the replication of GAstV-GD, revealing novel innate molecules triggered by astroviruses and highlighting that the ORF2 of GAstV-GD and OASL can be potential antiviral targets for astroviruses.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , Astroviridae/drug effects , Geese/virology , Open Reading Frames/drug effects , Virus Replication/drug effects , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/pharmacology , Animals , Astroviridae/genetics , Astroviridae Infections/immunology , Astroviridae Infections/veterinary , Astroviridae Infections/virology , Cell Line , Gene Knockdown Techniques , Immunity, Innate , Kinetics , Open Reading Frames/physiology , Poultry Diseases/immunology , Poultry Diseases/virology , Transcriptome , Virus Replication/physiology
4.
Parasitology ; 148(11): 1277-1287, 2021 09.
Article in English | MEDLINE | ID: mdl-34099078

ABSTRACT

During their complex life cycles, the Apicomplexan parasites Plasmodium falciparum and Toxoplasma gondii employ several layers of regulation of their gene expression. One such layer is mediated at the level of translation through upstream open reading frames (uORFs). As uORFs are found in the upstream regions of a majority of transcripts in both the parasites, it is essential that their roles in translational regulation be appreciated to a greater extent. This review provides a comprehensive summary of studies that show uORF-mediated gene regulation in these parasites and highlights examples of clinically and physiologically relevant genes, including var2csa in P. falciparum, and ApiAT1 in T. gondii, that exhibit uORF-mediated regulation. In addition to these examples, several studies that use bioinformatics, transcriptomics, proteomics and ribosome profiling also indicate the possibility of widespread translational regulation by uORFs. Further analysis of these genome-wide datasets, taking into account uORFs associated with each gene, will reveal novel genes involved in key biological pathways such as cell-cycle progression, stress-response and pathogenicity. The cumulative evidence from studies presented in this review suggests that uORFs will play crucial roles in regulating gene expression during clinical disease caused by these important human pathogens.


Subject(s)
Open Reading Frames/physiology , Plasmodium falciparum/physiology , Protein Biosynthesis/physiology , Toxoplasma/physiology , Gene Expression Regulation , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics , Toxoplasma/genetics , Toxoplasmosis/genetics , Toxoplasmosis/parasitology
5.
Proc Natl Acad Sci U S A ; 115(30): 7831-7836, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29915080

ABSTRACT

Plants adapt to alterations in light conditions by controlling their gene expression profiles. Expression of light-inducible genes is transcriptionally induced by transcription factors such as HY5. However, few detailed analyses have been carried out on the control of transcription start sites (TSSs). Of the various wavelengths of light, it is blue light (BL) that regulates physiological responses such as hypocotyl elongation and flowering time. To understand how gene expression is controlled not only by transcript abundance but also by TSS selection, we examined genome-wide TSS profiles in Arabidopsis seedlings after exposure to BL irradiation following initial growth in the dark. Thousands of genes use multiple TSSs, and some transcripts have upstream ORFs (uORFs) that take precedence over the main ORF (mORF) encoding proteins. The uORFs often function as translation inhibitors of the mORF or as triggers of nonsense-mediated mRNA decay (NMD). Transcription from TSSs located downstream of the uORFs in 220 genes is enhanced by BL exposure. This type of regulation is found in HY5 and HYH, major regulators of light-dependent gene expression. Translation efficiencies of the genes showing enhanced usage of these TSSs increased upon BL exposure. We also show that transcripts from TSSs upstream of uORFs in 45 of the 220 genes, including HY5, accumulated in a mutant of NMD. These results suggest that BL controls gene expression not only by enhancing transcriptions but also by choosing the TSS, and transcripts from downstream TSSs evade uORF-mediated inhibition to ensure high expression of light-regulated genes.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation, Plant/physiology , Nuclear Proteins/metabolism , Open Reading Frames/physiology , Transcription Initiation Site/physiology , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Carrier Proteins/genetics , DNA-Binding Proteins , Nuclear Proteins/genetics
6.
J Bacteriol ; 203(2)2020 12 18.
Article in English | MEDLINE | ID: mdl-33077634

ABSTRACT

Previous work identified gene product 56 (gp56), encoded by the lytic bacteriophage SP01, as being responsible for inhibition of Bacillus subtilis cell division during its infection. Assembly of the essential tubulin-like protein FtsZ into a ring-shaped structure at the nascent site of cytokinesis determines the timing and position of division in most bacteria. This FtsZ ring serves as a scaffold for recruitment of other proteins into a mature division-competent structure permitting membrane constriction and septal cell wall synthesis. Here, we show that expression of the predicted 9.3-kDa gp56 of SP01 inhibits later stages of B. subtilis cell division without altering FtsZ ring assembly. Green fluorescent protein-tagged gp56 localizes to the membrane at the site of division. While its localization does not interfere with recruitment of early division proteins, gp56 interferes with the recruitment of late division proteins, including Pbp2b and FtsW. Imaging of cells with specific division components deleted or depleted and two-hybrid analyses suggest that gp56 localization and activity depend on its interaction with FtsL. Together, these data support a model in which gp56 interacts with a central part of the division machinery to disrupt late recruitment of the division proteins involved in septal cell wall synthesis.IMPORTANCE Studies over the past decades have identified bacteriophage-encoded factors that interfere with host cell shape or cytokinesis during viral infection. The phage factors causing cell filamentation that have been investigated to date all act by targeting FtsZ, the conserved prokaryotic tubulin homolog that composes the cytokinetic ring in most bacteria and some groups of archaea. However, the mechanisms of several phage factors that inhibit cytokinesis, including gp56 of bacteriophage SP01 of Bacillus subtilis, remain unexplored. Here, we show that, unlike other published examples of phage inhibition of cytokinesis, gp56 blocks B. subtilis cell division without targeting FtsZ. Rather, it utilizes the assembled FtsZ cytokinetic ring to localize to the division machinery and to block recruitment of proteins needed for septal cell wall synthesis.


Subject(s)
Bacillus Phages/chemistry , Bacillus subtilis/virology , Bacterial Proteins/physiology , Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Penicillin-Binding Proteins/metabolism , Bacillus Phages/genetics , Bacillus subtilis/cytology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Count , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Green Fluorescent Proteins , Luminescent Agents , Open Reading Frames/physiology , Stem Cells/cytology
7.
J Virol ; 93(24)2019 12 15.
Article in English | MEDLINE | ID: mdl-31554685

ABSTRACT

The herpesvirus nuclear egress complex (NEC) is composed of two viral proteins. They play key roles in mediating the translocation of capsids from the nucleus to the cytoplasm by facilitating the budding of capsids into the perinuclear space (PNS). The NEC of alphaherpesvirus can induce the formation of virion-like vesicles from the nuclear membrane in the absence of other viral proteins. However, whether the NEC of gammaherpesvirus harbors the ability to do so in mammalian cells remains to be determined. In this study, we first constructed open reading frame 67 (ORF67)-null and ORF69-null mutants of murine gammaherpesvirus 68 (MHV-68) and demonstrated that both ORF67 and ORF69 play critical roles in nuclear egress and hence viral lytic replication. Biochemical and bioimaging analyses showed that ORF67 and ORF69 interacted with each other and were sufficient to induce the formation of virion-like vesicles from the nuclear membrane in mammalian cells. Thus, we designated ORF67 and ORF69 components of MHV-68 NEC. Furthermore, we identified amino acids critical for mediating the interaction between ORF67 and ORF69 through homology modeling and verified their function in nuclear egress, providing insights into the molecular basis of NEC formation in gammaherpesviruses.IMPORTANCE Increasing amounts of knowledge indicate that the nuclear egress complex (NEC) is critical for the nuclear egress of herpesvirus capsids, which can be viewed as a vesicle-mediated transport pathway through the nuclear membrane. In this study, we identified open reading frame 67 (ORF67) and ORF69 as components of the NEC in murine gammaherpesvirus 68 (MHV-68) and demonstrated that they efficiently induce virion-like vesicles from the nuclear membrane in mammalian cells. This is the first time that the NEC of a gammaherpesvirus has been found to demonstrate such an essential characteristic. In addition, we identified amino acids critical for mediating the interaction between ORF67 and ORF69 as well as nuclear egress. Notably, these amino acids are conserved in Kaposi's sarcoma-associated herpesvirus (KSHV) and Epstein-Barr virus (EBV), providing a structural basis to design antigammaherpesvirus drugs.


Subject(s)
Active Transport, Cell Nucleus/physiology , Cell Nucleus/metabolism , Gammaherpesvirinae/metabolism , Herpesviridae Infections/metabolism , Open Reading Frames/physiology , Viral Envelope Proteins/metabolism , Viral Proteins/metabolism , Animals , Capsid/metabolism , Cytoplasm/virology , DNA, Viral , Gammaherpesvirinae/genetics , HEK293 Cells , HeLa Cells , Herpesviridae/metabolism , Herpesviridae Infections/virology , Herpesvirus 4, Human/metabolism , Herpesvirus 8, Human/metabolism , Humans , Loss of Function Mutation , Mice , Nuclear Envelope/metabolism , Open Reading Frames/genetics , Virion/metabolism , Virus Replication
8.
Dev Biol ; 434(1): 96-107, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29208373

ABSTRACT

In early zebrafish development, the program for dorsal axis formation begins soon after fertilization. Previous studies suggested that dorsal determinants (DDs) localize to the vegetal pole, and are transported to the dorsal blastomeres in a microtubule-dependent manner. The DDs activate the canonical Wnt pathway and induce dorsal-specific genes that are required for dorsal axis formation. Among wnt-family genes, only the wnt8a mRNA is reported to localize to the vegetal pole in oocytes and to induce the dorsal axis, suggesting that Wnt8a is a candidate DD. Here, to reveal the roles of maternal wnt8a, we generated wnt8a mutants by transcription activator-like effector nucleases (TALENs), and established zygotic, maternal, and maternal zygotic wnt8a mutants by germ-line replacement. Zebrafish wnt8a has two open reading frames (ORF1 and ORF2) that are tandemly located in the genome. Although the zygotic ORF1 or ORF2 wnt8a mutants showed little or no axis-formation defects, the ORF1/2 compound mutants showed antero-dorsalized phenotypes, indicating that ORF1 and ORF2 have redundant roles in ventrolateral and posterior tissue formation. Unexpectedly, the maternal wnt8a ORF1/2 mutants showed no axis-formation defects. The maternal-zygotic wnt8a ORF1/2 mutants showed more severe antero-dorsalized phenotypes than the zygotic mutants. These results indicated that maternal wnt8a is dispensable for the initial dorsal determination, but cooperates with zygotic wnt8a for ventrolateral and posterior tissue formation. Finally, we re-examined the maternal wnt genes and found that Wnt6a is an alternative candidate DD.


Subject(s)
Cytoskeletal Proteins/metabolism , Embryo, Nonmammalian/embryology , Open Reading Frames/physiology , RNA, Messenger/metabolism , Wnt Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Cytoskeletal Proteins/genetics , RNA, Messenger/genetics , Wnt Proteins/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
9.
Plant Cell ; 28(11): 2830-2849, 2016 11.
Article in English | MEDLINE | ID: mdl-27760805

ABSTRACT

Upstream open reading frames (uORFs) are often translated ahead of the main ORF of a gene and regulate gene expression, sometimes in a condition-dependent manner, but such a role for the minimum uORF (hereafter referred to as AUG-stop) in living organisms is currently unclear. Here, we show that AUG-stop plays an important role in the boron (B)-dependent regulation of NIP5;1, encoding a boric acid channel required for normal growth under low B conditions in Arabidopsis thaliana High B enhanced ribosome stalling at AUG-stop, which was accompanied by the suppression of translation and mRNA degradation. This mRNA degradation was promoted by an upstream conserved sequence present near the 5'-edge of the stalled ribosome. Once ribosomes translate a uORF, reinitiation of translation must take place in order for the downstream ORF to be translated. Our results suggest that reinitiation of translation at the downstream NIP5;1 ORF is enhanced under low B conditions. A genome-wide analysis identified two additional B-responsive genes, SKU5 and the transcription factor gene ABS/NGAL1, which were regulated by B-dependent ribosome stalling through AUG-stop. This regulation was reproduced in both plant and animal transient expression and cell-free translation systems. These findings suggest that B-dependent AUG-stop-mediated regulation is common in eukaryotes.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Boron/metabolism , Gene Expression Regulation, Plant , Open Reading Frames/genetics , RNA Stability/physiology , Ribosomes/metabolism , Aquaporins/genetics , Aquaporins/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Open Reading Frames/physiology , RNA Stability/genetics , Ribosomes/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Nitric Oxide ; 88: 50-60, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31004763

ABSTRACT

The human inducible nitric oxide synthase (iNOS) gene contains an upstream open reading frame (uORF) in its 5'-untranslated region (5'-UTR) implying a translational regulation of iNOS expression. Transfection experiments in human DLD-1 cells revealed that the uORF although translatable seems not to inhibit the translation start at the bona fide ATG. Our data clearly show that human iNOS translation is cap-dependent and that the 5'-UTR of the iNOS mRNA contains no internal ribosome entry site. Translation of the bona fide coding sequence is most likely mediated by a leaky scanning mechanism. The 5'-UTR is encoded by exon 1 and exon 2 of the iNOS gene with the uORF stop codon located in front of the first intron indicating an involvement of the nonsense mediated RNA decay (NMD) in iNOS regulation. SiRNA-mediated down-regulation of Upf1 resulted in enhanced endogenous cytokine iNOS expression in human DLD-1 cells. Transfection of constructs containing iNOS exon 1, intron 1 and exon 2 in front of a luciferase gene showed a clear effect of the mutation of the uORF-ATG on luciferase reportergene expression. Our data indicate that the uORF in the 5'-UTR sequence of human iNOS gene reduces its expression via the NMD mechanism.


Subject(s)
Gene Expression Regulation/physiology , Nitric Oxide Synthase Type II/metabolism , Open Reading Frames/physiology , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Down-Regulation , Exons , Humans , Introns , Mutation , Nitric Oxide Synthase Type II/genetics , Nonsense Mediated mRNA Decay/physiology , RNA Helicases/genetics , RNA Helicases/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
11.
J Proteome Res ; 17(1): 1-11, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29188713

ABSTRACT

Peptides encoded by short open reading frames (sORFs) are usually defined as peptides ≤100 aa long. Usually sORFs were ignored by automatic genome annotation programs due to the high probability of false discovery. However, improved computational tools along with a high-throughput RIBO-seq approach identified a myriad of translated sORFs. Their importance becomes evident as we are gaining experimental validation of their diverse cellular functions. This Review examines various computational and experimental approaches of sORFs identification as well as provides the summary of our current knowledge of their functional roles in cells.


Subject(s)
Open Reading Frames/genetics , Peptides/genetics , Data Mining/methods , Open Reading Frames/physiology , Peptides/physiology
12.
J Biol Chem ; 292(23): 9613-9626, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28432120

ABSTRACT

Chemokines are essential for antimicrobial host defenses and tissue repair. Herpesviruses and poxviruses also encode chemokines, copied from their hosts and repurposed for multiple functions, including immune evasion. The CC chemokine MCK-2 encoded by mouse CMV (MCMV) has an atypical structure consisting of a classic chemokine domain N-terminal to a second unique domain, resulting from the splicing of MCMV ORFs m131 and m129 MCK-2 is essential for full MCMV infectivity in macrophages and for persistent infection in the salivary gland. However, information about its mechanism of action and specific biochemical roles for the two domains has been lacking. Here, using genetic, chemical, and enzymatic analyses of multiple mouse cell lines as well as primary mouse fibroblasts from salivary gland and lung, we demonstrate that MCK-2 binds glycosaminoglycans (GAGs) with affinities in the following order: heparin > heparan sulfate > chondroitin sulfate = dermatan sulfate. Both MCK-2 domains bound these GAGs independently, and computational analysis together with site-directed mutagenesis identified five basic residues distributed across the N terminus and the 30s and 50s loops of the chemokine domain that are important GAG binding determinants. Both domains were required for GAG-dependent oligomerization of full-length MCK-2. Thus, MCK-2 is an atypical viral chemokine consisting of a CC chemokine domain and a unique non-chemokine domain, both of which bind GAGs and are critical for GAG-dependent oligomerization of the full-length protein.


Subject(s)
Chemokines, CC/chemistry , Chemokines, CC/metabolism , Muromegalovirus/chemistry , Muromegalovirus/metabolism , Protein Multimerization/physiology , Viral Proteins/chemistry , Viral Proteins/metabolism , Animals , Chemokines, CC/genetics , Glycosaminoglycans/chemistry , Glycosaminoglycans/genetics , Glycosaminoglycans/metabolism , HEK293 Cells , Humans , Mice , Muromegalovirus/genetics , NIH 3T3 Cells , Open Reading Frames/physiology , Protein Domains , Protein Structure, Secondary , Viral Proteins/genetics
13.
Neurobiol Dis ; 117: 203-210, 2018 09.
Article in English | MEDLINE | ID: mdl-29908326

ABSTRACT

Mitochondrial encephalomyopathies (MEs) result from mutations in mitochondrial genes critical to oxidative phosphorylation. Severe and untreatable ME results from mutations affecting each endogenous mitochondrial encoded gene, including all 13 established protein coding genes. Effective techniques to manipulate mitochondrial genome are limited and targeted mitochondrial protein expression is currently unavailable. Here we report the development of a mitochondrial-targeted RNA expression (mtTRES) vector capable of protein expression within mitochondria (mtTRESPro). We demonstrate that mtTRESPro expressed RNAs are targeted to mitochondria and are capable of being translated using EGFP encoded constructs in vivo. We additionally test mtTRESPro constructs encoding wild type ATP6 for their ability to rescue an established ATP61Drosophila model of ME. Genetic rescue is examined including tests with co-expression of mitochondrial targeted translational inhibitors TLI-NCL::ATP6 RNAs that function to reduce expression of the endogenous mutant protein. The data demonstrate allotopic RNA expression of mitochondrial targeted wild type ATP6 coding RNAs are sufficient to partially rescue a severe and established animal model of ME but only when combined with a method to inhibit mutant protein expression, which likely competes for incorporation into complex V.


Subject(s)
Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Open Reading Frames/physiology , RNA, Mitochondrial/genetics , Animals , Animals, Genetically Modified , Cells, Cultured , Drosophila , HeLa Cells , Humans , Mitochondrial Diseases/metabolism , Mitochondrial Proteins/metabolism , RNA, Mitochondrial/metabolism
14.
Mol Biol Evol ; 34(5): 1110-1126, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28138077

ABSTRACT

While the principal force directing coding sequence (CDS) evolution is selection on protein function, to ensure correct gene expression CDSs must also maintain interactions with RNA-binding proteins (RBPs). Understanding how our genes are shaped by these RNA-level pressures is necessary for diagnostics and for improving transgenes. However, the evolutionary impact of the need to maintain RBP interactions remains unresolved. Are coding sequences constrained by the need to specify RBP binding motifs? If so, what proportion of mutations are affected? Might sequence evolution also be constrained by the need not to specify motifs that might attract unwanted binding, for instance because it would interfere with exon definition? Here, we have scanned human CDSs for motifs that have been experimentally determined to be recognized by RBPs. We observe two sets of motifs-those that are enriched over nucleotide-controlled null and those that are depleted. Importantly, the depleted set is enriched for motifs recognized by non-CDS binding RBPs. Supporting the functional relevance of our observations, we find that motifs that are more enriched are also slower-evolving. The net effect of this selection to preserve is a reduction in the over-all rate of synonymous evolution of 2-3% in both primates and rodents. Stronger motif depletion, on the other hand, is associated with stronger selection against motif gain in evolution. The challenge faced by our CDSs is therefore not only one of attracting the right RBPs but also of avoiding the wrong ones, all while also evolving under selection pressures related to protein structure.


Subject(s)
Open Reading Frames/genetics , Open Reading Frames/physiology , RNA-Binding Proteins/genetics , Animals , Biological Evolution , Databases, Nucleic Acid , Evolution, Molecular , Exons , Humans , Mutation/genetics , RNA , RNA-Binding Proteins/metabolism
15.
Appl Microbiol Biotechnol ; 102(16): 6841-6845, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29909570

ABSTRACT

A large number of DNAs in eukaryote genomes can code for atypical transcripts, and their functions are controversial. It has been reported that the transcripts contain many small open reading frames (sORFs), which were originally considered as non-translatable RNAs. However, increasing evidence has suggested that some of these sORFs can encode for small peptides and some are conserved across large evolutionary distances. It has been reported that the small peptides have functions and may be involved in varieties of cellular processes, playing important roles in development, physiology, and metabolism. Among the sORFs, studies of the non-canonical gene polished rice/tarsal-less (pri/tal) in Drosophila and mille-pattes(mlpt) in Tribolium have been more thoroughly studied. The genes similar to pri/tal in other species have been defined as the tarsal-less-related gene family, tal-like gene. In this review, we described recent progress in the discovery and functional characterization of the small peptides encoded by the tal-like gene and their possible functional potentials.


Subject(s)
Drosophila Proteins/genetics , Open Reading Frames/genetics , Open Reading Frames/physiology , Peptides/physiology , Transaldolase/genetics , Animals
16.
Mol Cell ; 39(2): 234-46, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20670892

ABSTRACT

Methylation of histone H3 by Set1 and Set2 is required for deacetylation of nucleosomes in coding regions by histone deacetylase complexes (HDACs) Set3C and Rpd3C(S), respectively. We report that Set3C and Rpd3C(S) are cotranscriptionally recruited in the absence of Set1 and Set2, but in a manner stimulated by Pol II CTD kinase Cdk7/Kin28. Consistently, Rpd3C(S) and Set3C interact with Ser5-phosphorylated Pol II and histones in extracts, but only the histone interactions require H3 methylation. Moreover, reconstituted Rpd3C(S) binds specifically to Ser5-phosphorylated CTD peptides in vitro. Hence, whereas interaction with methylated H3 residues is required for Rpd3C(S) and Set3C deacetylation activities, their cotranscriptional recruitment is stimulated by the phosphorylated CTD. We further demonstrate that Rpd3, Hos2, and Hda1 have overlapping functions in deacetylating histones and suppressing cotranscriptional histone eviction. A strong correlation between increased acetylation and lower histone occupancy in HDA mutants implies that histone acetylation is important for nucleosome eviction.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Histone Deacetylases/metabolism , Nucleosomes/metabolism , Open Reading Frames/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cyclin-Dependent Kinases/genetics , Histone Deacetylases/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Nucleosomes/genetics , Phosphorylation/physiology , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
17.
J Biol Chem ; 291(33): 16927-35, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27358398

ABSTRACT

Translation regulation largely occurs during initiation, which features ribosome assembly onto mRNAs and selection of the translation start site. Short, upstream ORFs (uORFs) located in the 5'-leader of the mRNA can be selected for translation. Multiple transcripts associated with stress amelioration are preferentially translated through uORF-mediated mechanisms during activation of the integrated stress response (ISR) in which phosphorylation of the α subunit of eIF2 results in a coincident global reduction in translation initiation. This review presents key features of uORFs that serve to optimize translational control that is essential for regulation of cell fate in response to environmental stresses.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Open Reading Frames/physiology , Peptide Chain Initiation, Translational/physiology , RNA, Messenger/metabolism , Stress, Physiological , Animals , Humans
18.
PLoS Pathog ; 11(2): e1004664, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25679389

ABSTRACT

RNA viruses exhibit substantial structural, ecological and genomic diversity. However, genome size in RNA viruses is likely limited by a high mutation rate, resulting in the evolution of various mechanisms to increase complexity while minimising genome expansion. Here we conduct a large-scale analysis of the genome sequences of 99 animal rhabdoviruses, including 45 genomes which we determined de novo, to identify patterns of genome expansion and the evolution of genome complexity. All but seven of the rhabdoviruses clustered into 17 well-supported monophyletic groups, of which eight corresponded to established genera, seven were assigned as new genera, and two were taxonomically ambiguous. We show that the acquisition and loss of new genes appears to have been a central theme of rhabdovirus evolution, and has been associated with the appearance of alternative, overlapping and consecutive ORFs within the major structural protein genes, and the insertion and loss of additional ORFs in each gene junction in a clade-specific manner. Changes in the lengths of gene junctions accounted for as much as 48.5% of the variation in genome size from the smallest to the largest genome, and the frequency with which new ORFs were observed increased in the 3' to 5' direction along the genome. We also identify several new families of accessory genes encoded in these regions, and show that non-canonical expression strategies involving TURBS-like termination-reinitiation, ribosomal frame-shifts and leaky ribosomal scanning appear to be common. We conclude that rhabdoviruses have an unusual capacity for genomic plasticity that may be linked to their discontinuous transcription strategy from the negative-sense single-stranded RNA genome, and propose a model that accounts for the regular occurrence of genome expansion and contraction throughout the evolution of the Rhabdoviridae.


Subject(s)
Evolution, Molecular , Genome, Viral/physiology , Open Reading Frames/physiology , RNA, Viral/genetics , Rhabdoviridae/genetics , Base Sequence , Molecular Sequence Data
19.
Proc Natl Acad Sci U S A ; 111(38): E4043-52, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25192933

ABSTRACT

Human norovirus (HuNoV) is the leading cause of gastroenteritis worldwide. HuNoV replication studies have been hampered by the inability to grow the virus in cultured cells. The HuNoV genome is a positive-sense single-stranded RNA (ssRNA) molecule with three open reading frames (ORFs). We established a reverse genetics system driven by a mammalian promoter that functions without helper virus. The complete genome of the HuNoV genogroup II.3 U201 strain was cloned downstream of an elongation factor-1α (EF-1α) mammalian promoter. Cells transfected with plasmid containing the full-length genome (pHuNoVU201F) expressed the ORF1 polyprotein, which was cleaved by the viral protease to produce the mature nonstructural viral proteins, and the capsid proteins. Progeny virus produced from the transfected cells contained the complete NoV genomic RNA (VP1, VP2, and VPg) and exhibited the same density in isopycnic cesium chloride gradients as native infectious NoV particles from a patient's stool. This system also was applied to drive murine NoV RNA replication and produced infectious progeny virions. A GFP reporter construct containing the GFP gene in ORF1 produced complete virions that contain VPg-linked RNA. RNA from virions containing the encapsidated GFP-genomic RNA was successfully transfected back into cells producing fluorescent puncta, indicating that the encapsidated RNA is replication-competent. The EF-1α mammalian promoter expression system provides the first reverse genetics system, to our knowledge, generalizable for human and animal NoVs that does not require a helper virus. Establishing a complete reverse genetics system expressed from cDNA for HuNoVs now allows the manipulation of the viral genome and production of reporter virions.


Subject(s)
Genes, Reporter , Genome, Viral , Norovirus , Plasmids , RNA, Viral , Viral Proteins , Animals , COS Cells , Chlorocebus aethiops , DNA, Complementary/genetics , DNA, Complementary/metabolism , Gene Expression Regulation/genetics , Humans , Norovirus/genetics , Norovirus/metabolism , Open Reading Frames/physiology , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism , Plasmids/genetics , Plasmids/metabolism , Promoter Regions, Genetic , RNA, Viral/biosynthesis , RNA, Viral/genetics , Viral Proteins/biosynthesis , Viral Proteins/genetics , Virion/genetics , Virion/metabolism
20.
RNA ; 20(4): 559-67, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24570481

ABSTRACT

Candida albicans is a major fungal pathogen that responds to various environmental cues as part of its infection mechanism. We show here that the expression of C. albicans GCN4, which encodes a transcription factor that regulates morphogenetic and metabolic responses, is translationally regulated in response to amino acid starvation induced by exposure to the histidine analog 3-aminotriazole (3AT). However, in contrast to the well-known translational control mechanisms that regulate yeast GCN4 and mammalian ATF4 expression via multiple upstream open reading frames (uORFs) in their 5'-leader sequences, a single inhibitory uORF is necessary and sufficient for C. albicans GCN4 translational control. The 5'-leader sequence of GCN4 contains three uORFs, but uORF3 alone is sufficient for translational regulation. Under nonstress conditions, uORF3 inhibits GCN4 translation. Amino acid starvation conditions promote Gcn2-mediated phosphorylation of eIF2α and leaky ribosomal scanning to bypass uORF3, inducing GCN4 translation. GCN4 expression is also transcriptionally regulated, although maximal induction is observed at higher concentrations of 3AT compared with translational regulation. C. albicans GCN4 expression is therefore highly regulated by both transcriptional and translational control mechanisms. We suggest that it is particularly important that Gcn4 levels are tightly controlled since Gcn4 regulates morphogenetic changes during amino acid starvation conditions, which are important determinants of virulence in this fungus.


Subject(s)
Amino Acids/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Candida albicans/metabolism , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Open Reading Frames/physiology , Protein Biosynthesis , Base Sequence , Blotting, Western , Candida albicans/genetics , Candida albicans/growth & development , Fungal Proteins/genetics , Molecular Sequence Data , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Starvation
SELECTION OF CITATIONS
SEARCH DETAIL