Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 485
Filter
1.
Nature ; 631(8020): 432-438, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38898279

ABSTRACT

When mRNAs have been transcribed and processed in the nucleus, they are exported to the cytoplasm for translation. This export is mediated by the export receptor heterodimer Mex67-Mtr2 in the yeast Saccharomyces cerevisiae (TAP-p15 in humans)1,2. Interestingly, many long non-coding RNAs (lncRNAs) also leave the nucleus but it is currently unclear why they move to the cytoplasm3. Here we show that antisense RNAs (asRNAs) accelerate mRNA export by annealing with their sense counterparts through the helicase Dbp2. These double-stranded RNAs (dsRNAs) dominate export compared with single-stranded RNAs (ssRNAs) because they have a higher capacity and affinity for the export receptor Mex67. In this way, asRNAs boost gene expression, which is beneficial for cells. This is particularly important when the expression program changes. Consequently, the degradation of dsRNA, or the prevention of its formation, is toxic for cells. This mechanism illuminates the general cellular occurrence of asRNAs and explains their nuclear export.


Subject(s)
Active Transport, Cell Nucleus , Cell Nucleus , Gene Expression Regulation, Fungal , RNA Transport , RNA, Antisense , RNA, Double-Stranded , RNA, Messenger , Saccharomyces cerevisiae , Cell Nucleus/metabolism , Cytoplasm/metabolism , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Nucleocytoplasmic Transport Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , RNA, Antisense/metabolism , RNA, Antisense/genetics , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics
2.
Proc Natl Acad Sci U S A ; 121(22): e2314166121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38768348

ABSTRACT

The nonstructural protein 1 (Nsp1) of SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) is a virulence factor that targets multiple cellular pathways to inhibit host gene expression and antiviral response. However, the underlying mechanisms of the various Nsp1-mediated functions and their contributions to SARS-CoV-2 virulence remain unclear. Among the targets of Nsp1 is the mRNA (messenger ribonucleic acid) export receptor NXF1-NXT1, which mediates nuclear export of mRNAs from the nucleus to the cytoplasm. Based on Nsp1 crystal structure, we generated mutants on Nsp1 surfaces and identified an acidic N-terminal patch that is critical for interaction with NXF1-NXT1. Photoactivatable Nsp1 probe reveals the RNA Recognition Motif (RRM) domain of NXF1 as an Nsp1 N-terminal binding site. By mutating the Nsp1 N-terminal acidic patch, we identified a separation-of-function mutant of Nsp1 that retains its translation inhibitory function but substantially loses its interaction with NXF1 and reverts Nsp1-mediated mRNA export inhibition. We then generated a recombinant (r)SARS-CoV-2 mutant on the Nsp1 N-terminal acidic patch and found that this surface is key to promote NXF1 binding and inhibition of host mRNA nuclear export, viral replication, and pathogenicity in vivo. Thus, these findings provide a mechanistic understanding of Nsp1-mediated mRNA export inhibition and establish the importance of this pathway in the virulence of SARS-CoV-2.


Subject(s)
Active Transport, Cell Nucleus , COVID-19 , Nucleocytoplasmic Transport Proteins , RNA, Messenger , RNA-Binding Proteins , SARS-CoV-2 , Viral Nonstructural Proteins , Humans , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , SARS-CoV-2/genetics , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Animals , COVID-19/virology , COVID-19/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Virus Replication , Cell Nucleus/metabolism , Vero Cells , Virulence , Chlorocebus aethiops , HEK293 Cells
3.
Adv Sci (Weinh) ; 11(24): e2307639, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38626369

ABSTRACT

Regulating nuclear export precisely is essential for maintaining mRNA homeostasis and impacts tumor progression. However, the mechanisms governing nuclear mRNA export remain poorly elucidated. Herein, it is revealed that the enhanced hypoxic long no-ncoding RNA (lncRNA prostate cancer associated transcript 6 (PCAT6) in breast cancer (BC) promotes the nuclear export of m6A-modified mRNAs, bolstering breast cancer stem cells (BCSCs) stemness and doxorubicin resistance. Clinically, hypoxic PCAT6 correlates with malignant BC features and poor prognosis. Mechanically, PCAT6 functions as a scaffold between interferon-stimulated gene 15 (ISG15) and heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNPA2B1), leading to ISGylation of hnRNPA2B1, thus protecting hnRNPA2B1 from ubiquitination-mediated proteasomal degradation. Interestingly, as an m6A reader, hnRNPA2B1 selectively mediates m6A-tagged mRNAs nuclear export via the Aly/REF export factor (ALYREF)/ nuclear RNA export factor 1 (NXF1) complex, which promotes stemness-related genes expression. HnRNPA2B1 knockdown or mRNA export inhibition can result in the retention of nuclear m6A-tagged mRNA associated with stemness maintenance, which suppresses BCSCs self-renewal and effectively improves the efficacy of doxorubicin therapy. These findings demonstrate the pivotal role of m6A-modified mRNA nuclear export in BC progression, highlighting that the inhibition of m6A-tagged mRNA and its nuclear export is a potential therapeutic strategy for the amelioration of cancer chemotherapy.


Subject(s)
Breast Neoplasms , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , RNA, Messenger , Humans , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Female , RNA, Messenger/genetics , RNA, Messenger/metabolism , Cell Line, Tumor , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Animals , Nucleocytoplasmic Transport Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Neoplastic Stem Cells/metabolism
4.
Mol Cell ; 84(9): 1764-1782.e10, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38593806

ABSTRACT

mRNAs continually change their protein partners throughout their lifetimes, yet our understanding of mRNA-protein complex (mRNP) remodeling is limited by a lack of temporal data. Here, we present time-resolved mRNA interactome data by performing pulse metabolic labeling with photoactivatable ribonucleoside in human cells, UVA crosslinking, poly(A)+ RNA isolation, and mass spectrometry. This longitudinal approach allowed the quantification of over 700 RNA binding proteins (RBPs) across ten time points. Overall, the sequential order of mRNA binding aligns well with known functions, subcellular locations, and molecular interactions. However, we also observed RBPs with unexpected dynamics: the transcription-export (TREX) complex recruited posttranscriptionally after nuclear export factor 1 (NXF1) binding, challenging the current view of transcription-coupled mRNA export, and stress granule proteins prevalent in aged mRNPs, indicating roles in late stages of the mRNA life cycle. To systematically identify mRBPs with unknown functions, we employed machine learning to compare mRNA binding dynamics with Gene Ontology (GO) annotations. Our data can be explored at chronology.rna.snu.ac.kr.


Subject(s)
RNA, Messenger , RNA-Binding Proteins , Humans , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Protein Binding , Nucleocytoplasmic Transport Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , HeLa Cells , Time Factors , Machine Learning
5.
Arch Biochem Biophys ; 754: 109896, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38417691

ABSTRACT

AIMS: The purpose of this study was to explore the role of RAE1 in the invasion and metastasis of gastric cancer (GC) cells. MATERIALS AND METHODS: RAE1 expression in GC cells was determined by reverse-transcription polymerase chain reaction (qRT-PCR) and Western blotting (WB). Cell models featuring RAE1 gene silencing and overexpression were constructed by lentiviral transfection; The proliferation, migration, and invasion ability of cells were detected by cell counting, colony formation assay, would healing assay, and transwell invasion and migration test. WB analysis of ERK/MAPK signaling pathway (ERK1/2, p-ERK1/2, c-Myc) and EMT-related molecules (ZEB1, E-cadherin, N-cadherin, and Vimentin). RESULTS: The expression level of RAE1 in GC was notably higher than in adjacent tissues. Elevated RAE1 expression correlated with an unfavorable prognosis for GC patients. Knockdown of RAE1, as compared to the control group, resulted in a significant inhibition of proliferation, migration, and invasion abilities in GC cell lines. Furthermore, RAE1 knockdown led to a substantial decrease in the expression of N-cadherin, vimentin, ZEB1, p-ERK1/2, and c-Myc proteins, coupled with a marked increase in E-cadherin expression. The biological effects of RAE1 in GC cells were effectively reversed by the inhibition of the ERK/MAPK signaling pathway using SCH772984. Additionally, RAE1 knockdown demonstrated a suppressive effect on GC tumor size in vivo. Immunohistochemistry (IHC) results revealed significantly lower expression of Ki-67 in RAE1 knockout mice compared to the control group. CONCLUSIONS: RAE1 promotes GC cell migration and invasion through the ERK/MAPK pathway and is a potential therapeutic target for GC therapy.


Subject(s)
Epithelial-Mesenchymal Transition , Stomach Neoplasms , Animals , Humans , Mice , Cadherins/genetics , Cadherins/metabolism , Carcinogenesis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/genetics , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Vimentin/genetics , Vimentin/metabolism
6.
Cell Rep ; 43(1): 113593, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38113140

ABSTRACT

Nuclear mRNA export via nuclear pore complexes is an essential step in eukaryotic gene expression. Although factors involved in mRNA transport have been characterized, a comprehensive mechanistic understanding of this process and its regulation is lacking. Here, we use single-RNA imaging in yeast to show that cells use mRNA retention to control mRNA export during stress. We demonstrate that, upon glucose withdrawal, the essential RNA-binding factor Nab2 forms RNA-dependent condensate-like structures in the nucleus. This coincides with a reduced abundance of the DEAD-box ATPase Dbp5 at the nuclear pore. Depleting Dbp5, and consequently blocking mRNA export, is necessary and sufficient to trigger Nab2 condensation. The state of Nab2 condensation influences the extent of nuclear mRNA accumulation and can be recapitulated in vitro, where Nab2 forms RNA-dependent liquid droplets. We hypothesize that cells use condensation to regulate mRNA export and control gene expression during stress.


Subject(s)
Nuclear Pore Complex Proteins , Saccharomyces cerevisiae Proteins , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , DEAD-box RNA Helicases/metabolism , Nuclear Pore Complex Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , RNA/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
7.
Adv Biol Regul ; 90: 100990, 2023 12.
Article in English | MEDLINE | ID: mdl-37801910

ABSTRACT

Gle1 regulates gene expression at multiple steps from transcription to mRNA export to translation under stressed and non-stressed conditions. To better understand Gle1 function in stressed human cells, specific antibodies were generated that recognized the phosphorylation of threonine residue 102 (T102) in Gle1. A series of in vitro kinase assays indicated that T102 phosphorylation serves as a priming event for further phosphorylation in Gle1's N-terminal low complexity cluster. Indirect immunofluorescence microscopy with the anti-Gle1-pT102 antibodies revealed that basally phosphorylated Gle1 was pre-dominantly nuclear with punctate distribution; however, under sodium arsenite-induced stress, more cytoplasmic localization was detected. Immunoprecipitation with the anti-Gle1-pT102 antibody resulted in co-isolation of Gle1-pT102 with the DEAD-box protein DDX1 in a phosphatase sensitive manner. This suggested Gle1 phosphorylation might be linked to its role in regulating DDX1 during transcription termination. Notably, whereas the total Gle1-DDX1 association was decreased when Gle1 nucleocytoplasmic shuttling was disrupted, co-isolation of Gle1-pT102 and DDX1 increased under the same conditions. Taken together, these studies demonstrated that Gle1 phosphorylation impacts its cellular distribution and potentially drives nuclear Gle1 functions in transcription termination. We propose a model wherein phosphorylation of Gle1 either reduces its nucleocytoplasmic shuttling capacity or increases its binding affinity with nuclear interaction partners.


Subject(s)
Nuclear Pore Complex Proteins , Humans , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , Phosphorylation , Cell Nucleus/metabolism
8.
RNA ; 29(12): 1870-1880, 2023 12.
Article in English | MEDLINE | ID: mdl-37699651

ABSTRACT

The conserved TREX complex has multiple functions in gene expression such as transcription elongation, 3' end processing, mRNP assembly and nuclear mRNA export as well as the maintenance of genomic stability. In Saccharomyces cerevisiae, TREX is composed of the pentameric THO complex, the DEAD-box RNA helicase Sub2, the nuclear mRNA export adaptor Yra1, and the SR-like proteins Gbp2 and Hrb1. Here, we present the structural analysis of the endogenous TREX complex of S. cerevisiae purified from its native environment. To this end, we used cross-linking mass spectrometry to gain structural information on regions of the complex that are not accessible to classical structural biology techniques. We also used negative-stain electron microscopy to investigate the organization of the cross-linked complex used for XL-MS by comparing our endogenous TREX complex with recently published structural models of recombinant THO-Sub2 complexes. According to our analysis, the endogenous yeast TREX complex preferentially assembles into a dimer.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , RNA, Messenger/genetics , RNA Transport , Transcription, Genetic , Nucleocytoplasmic Transport Proteins/genetics , Poly(A)-Binding Proteins/genetics
9.
Biol Chem ; 404(8-9): 845-850, 2023 07 26.
Article in English | MEDLINE | ID: mdl-37436777

ABSTRACT

Cell viability largely depends on the surveillance of mRNA export and translation. Upon pre-mRNA processing and nuclear quality control, mature mRNAs are exported into the cytoplasm via Mex67-Mtr2 attachment. At the cytoplasmic site of the nuclear pore complex, the export receptor is displaced by the action of the DEAD-box RNA helicase Dbp5. Subsequent quality control of the open reading frame requires translation. Our studies suggest an involvement of Dbp5 in cytoplasmic no-go-and non-stop decay. Most importantly, we have also identified a key function for Dbp5 in translation termination, which identifies this helicase as a master regulator of mRNA expression.


Subject(s)
Nucleocytoplasmic Transport Proteins , Saccharomyces cerevisiae Proteins , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Active Transport, Cell Nucleus , Saccharomyces cerevisiae Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Gene Expression
10.
Commun Biol ; 6(1): 664, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37353594

ABSTRACT

Self-renewing somatic tissues rely on progenitors to support the continuous tissue regeneration. The gene regulatory network maintaining progenitor function remains incompletely understood. Here we show that NUP98 and RAE1 are highly expressed in epidermal progenitors, forming a separate complex in the nucleoplasm. Reduction of NUP98 or RAE1 abolishes progenitors' regenerative capacity, inhibiting proliferation and inducing premature terminal differentiation. Mechanistically, NUP98 binds on chromatin near the transcription start sites of key epigenetic regulators (such as DNMT1, UHRF1 and EZH2) and sustains their expression in progenitors. NUP98's chromatin binding sites are co-occupied by HDAC1. HDAC inhibition diminishes NUP98's chromatin binding and dysregulates NUP98 and RAE1's target gene expression. Interestingly, HDAC inhibition further induces NUP98 and RAE1 to localize interdependently to the nucleolus. These findings identified a pathway in progenitor maintenance, where HDAC activity directs the high levels of NUP98 and RAE1 to directly control key epigenetic regulators, escaping from nucleolar aggregation.


Subject(s)
Chromatin , Nucleocytoplasmic Transport Proteins , Nucleocytoplasmic Transport Proteins/chemistry , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , Chromatin/genetics , Nuclear Matrix-Associated Proteins/chemistry , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Binding Sites
11.
Am J Med Genet A ; 191(6): 1614-1618, 2023 06.
Article in English | MEDLINE | ID: mdl-36891747

ABSTRACT

Ethylmalonic encephalopathy (EE) is a rare, severe, autosomal recessive condition caused by pathogenic variants in ETHE1 leading to progressive encephalopathy, hypotonia evolving to dystonia, petechiae, orthostatic acrocyanosis, diarrhea, and elevated ethylmalonic acid in urine. In this case report, we describe a patient with only mild speech and gross motor delays, subtle biochemical abnormalities, and normal brain imaging found to be homozygous for a pathogenic ETHE1 variant (c.586G>A) via whole exome sequencing. This case highlights the clinical heterogeneity of ETHE1 mutations and the utility of whole-exome sequencing in diagnosing mild cases of EE.


Subject(s)
Brain Diseases, Metabolic, Inborn , Brain Diseases , Purpura , Humans , Brain Diseases, Metabolic, Inborn/diagnosis , Brain Diseases, Metabolic, Inborn/genetics , Purpura/diagnosis , Purpura/genetics , Brain/pathology , Brain Diseases/diagnosis , Brain Diseases/genetics , Brain Diseases/pathology , Mitochondrial Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics
12.
Life Sci Alliance ; 6(3)2023 03.
Article in English | MEDLINE | ID: mdl-36599624

ABSTRACT

Replication licensing, a prerequisite of DNA replication, helps to ensure once-per-cell-cycle genome duplication. Some DNA replication-initiation proteins are sequentially loaded onto replication origins to form pre-replicative complexes (pre-RCs). ORC and Noc3p bind replication origins throughout the cell cycle, providing a platform for pre-RC assembly. We previously reported that cell cycle-dependent ORC dimerization is essential for the chromatin loading of the symmetric MCM double-hexamers. Here, we used Saccharomyces cerevisiae separation-of-function NOC3 mutants to confirm the separable roles of Noc3p in DNA replication and ribosome biogenesis. We also show that an essential and cell cycle-dependent Noc3p dimerization cycle regulates the ORC dimerization cycle. Noc3p dimerizes at the M-to-G1 transition and de-dimerizes in S-phase. The Noc3p dimerization cycle coupled with the ORC dimerization cycle enables replication licensing, protects nascent sister replication origins after replication initiation, and prevents re-replication. This study has revealed a new mechanism of replication licensing and elucidated the molecular mechanism of Noc3p as a mediator of ORC dimerization in pre-RC formation.


Subject(s)
Protein Multimerization , Saccharomyces cerevisiae Proteins , Cell Cycle/genetics , Dimerization , DNA Replication/genetics , Origin Recognition Complex/genetics , Origin Recognition Complex/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Protein Multimerization/genetics , Protein Multimerization/physiology , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/physiology , Nuclear Proteins/genetics , Nuclear Proteins/physiology
13.
Nat Commun ; 13(1): 5881, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36202822

ABSTRACT

The changes occurring in mRNA organization during nucleo-cytoplasmic transport and export, are not well understood. Moreover, directionality of mRNA passage through the nuclear pore complex (NPC) has not been examined within individual NPCs. Here we find that an mRNP is compact during nucleoplasmic travels compared to a more open structure after transcription and at the nuclear periphery. Compaction levels of nuclear transcripts can be modulated by varying levels of SR proteins and by changing genome organization. Nuclear mRNPs are mostly rod-shaped with distant 5'/3'-ends, although for some, the ends are in proximity. The latter is more abundant in the cytoplasm and can be modified by translation inhibition. mRNAs and lncRNAs exiting the NPC exhibit predominant 5'-first export. In some cases, several adjacent NPCs are engaged in export of the same mRNA suggesting 'gene gating'. Altogether, we show that the mRNP is a flexible structure during travels, with 5'-directionality during export.


Subject(s)
Nuclear Pore , RNA, Long Noncoding , Active Transport, Cell Nucleus/genetics , Cell Nucleus/metabolism , DEAD-box RNA Helicases/metabolism , Nuclear Pore/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , RNA Transport , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
Nucleic Acids Res ; 50(17): 10140-10152, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36099418

ABSTRACT

tRNAs that are transcribed in the nucleus are exported to the cytoplasm to perform their iterative essential function in translation. However, the complex set of tRNA post-transcriptional processing and subcellular trafficking steps are not completely understood. In particular, proteins involved in tRNA nuclear export remain unknown since the canonical tRNA nuclear exportin, Los1/Exportin-t, is unessential in all tested organisms. We previously reported that budding yeast Mex67-Mtr2, a mRNA nuclear exporter, co-functions with Los1 in tRNA nuclear export. Here we employed in vivo co-purification of tRNAs with endogenously expressed nuclear exporters to document that Crm1 also is a bona fide tRNA nuclear exporter. We document that Los1, Mex67-Mtr2 and Crm1 possess individual tRNA preferences for forming nuclear export complexes with members of the 10 families of intron-containing pre-tRNAs. Remarkably, Mex67-Mtr2, but not Los1 or Crm1, is error-prone, delivering tRNAs to the cytoplasm prior to 5' leader removal. tRNA retrograde nuclear import functions to monitor the aberrant leader-containing spliced tRNAs, returning them to the nucleus where they are degraded by 3' to 5' exonucleases. Overall, our work identifies a new tRNA nuclear exporter, uncovers exporter preferences for specific tRNA families, and documents contribution of tRNA nuclear import to tRNA quality control.


Subject(s)
RNA, Transfer , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Active Transport, Cell Nucleus/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Exonucleases/metabolism , Karyopherins/genetics , Karyopherins/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Messenger/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
15.
Biosci Rep ; 42(9)2022 09 30.
Article in English | MEDLINE | ID: mdl-36093993

ABSTRACT

Mitochondrial cytopathies, among which the Leigh syndrome (LS), are caused by variants either in the mitochondrial or the nuclear genome, affecting the oxidative phosphorylation process. The aim of the present study consisted in defining the molecular diagnosis of a group of Tunisian patients with LS. Six children, belonging to five Tunisian families, with clinical and imaging presentations suggestive of LS were recruited. Whole mitochondrial DNA and targeted next-generation sequencing of a panel of 281 nuclear genes involved in mitochondrial physiology were performed. Bioinformatic analyses were achieved in order to identify deleterious variations. A single m.10197G>A (p.Ala47Thr) variant was found in the mitochondrial MT-ND3 gene in one patient, while the others were related to autosomal homozygous variants: two c.1412delA (p.Gln471ArgfsTer42) and c.1264A>G (p.Thr422Ala) in SLC19A3, one c.454C>G (p.Pro152Ala) in SLC25A19 and one c.122G>A (p.Gly41Asp) in ETHE1. Our findings demonstrate the usefulness of genomic investigations to improve LS diagnosis in consanguineous populations and further allow for treating the patients harboring variants in SLC19A3 and SLC25A19 that contribute to thiamine transport, by thiamine and biotin supplementation. Considering the Tunisian genetic background, the newly identified variants could be screened in patients with similar clinical presentation in related populations.


Subject(s)
Leigh Disease , Biotin/genetics , Child , DNA, Mitochondrial/genetics , High-Throughput Nucleotide Sequencing , Humans , Leigh Disease/diagnosis , Leigh Disease/genetics , Leigh Disease/therapy , Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/genetics , Mutation , Nucleocytoplasmic Transport Proteins/genetics , Thiamine
16.
Eur J Med Genet ; 65(10): 104597, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36030004

ABSTRACT

Premature ovarian insufficiency (POI) is a major cause of female subfertility. Although POI affects approximately 1-2% women worldwide, the etiology of a large number of POI patients remains unknown partially due to the genetic heterogeneity of POI. EIF4ENIF1 is one of the known POI-causative genes, and it plays an essential role in inhibiting mRNA translation and regulating mRNA destabilization in ovarian cells. In our study, two EIF4ENIF1 variants, c.9_11delGAG (p.R4del) (rs3834682) and c.2861G > C (p.G954A) (rs766008983) were identified in two sporadic Han Chinese POI patients through whole-exome sequencing. Both variants are rare in the human population. The two patients' mothers don't carry the rare variants and they have regular menstruation. The missense variant c.2861G > C was predicted to be deleterious by multiple bioinformatic tools. Western blot analysis further demonstrated that both of the two variants exhibited reduced mRNA and protein expression levels compared with the wild-type in vitro. Taken together, our findings reported two rare POI-associated EIF4ENIF1 variants, providing insights into genetic counseling and suggesting the contribution of EIF4ENIF1 variants in female infertility.


Subject(s)
Primary Ovarian Insufficiency , Asian People , Female , Humans , Male , Mutation, Missense , Nucleocytoplasmic Transport Proteins/genetics , Primary Ovarian Insufficiency/genetics , RNA, Messenger/genetics , Exome Sequencing
17.
J Virol ; 96(18): e0090022, 2022 09 28.
Article in English | MEDLINE | ID: mdl-36040180

ABSTRACT

Many negative-sense RNA viruses, including the highly pathogenic Ebola virus (EBOV), use cytoplasmic inclusion bodies (IBs) for viral RNA synthesis. However, it remains unclear how viral mRNAs are exported from these IBs for subsequent translation. We recently demonstrated that the nuclear RNA export factor 1 (NXF1) is involved in a late step in viral protein expression, i.e., downstream of viral mRNA transcription, and proposed it to be involved in this mRNA export process. We now provide further evidence for this function by showing that NXF1 is not required for translation of viral mRNAs, thus pinpointing its function to a step between mRNA transcription and translation. We further show that RNA binding of both NXF1 and EBOV NP is necessary for export of NXF1 from IBs, supporting a model in which NP hands viral mRNA over to NXF1 for export. Mapping of NP-NXF1 interactions allowed refinement of this model, revealing two separate interaction sites, one of them directly involving the RNA binding cleft of NP, even though these interactions are RNA-independent. Immunofluorescence analyses demonstrated that individual NXF1 domains are sufficient for its recruitment into IBs, and complementation assays helped to define NXF1 domains important for its function in the EBOV life cycle. Finally, we show that NXF1 is also required for protein expression of other viruses that replicate in cytoplasmic IBs, including Lloviu and Junín virus. These data suggest a role for NXF1 in viral mRNA export from IBs for various viruses, making it a potential target for broadly active antivirals. IMPORTANCE Filoviruses such as the Ebola virus (EBOV) cause severe hemorrhagic fevers with high case fatality rates and limited treatment options. The identification of virus-host cell interactions shared among several viruses would represent promising targets for the development of broadly active antivirals. In this study, we reveal the mechanistic details of how EBOV usurps the nuclear RNA export factor 1 (NXF1) to export viral mRNAs from viral inclusion bodies (IBs). We further show that NXF1 is not only required for the EBOV life cycle but also necessary for other viruses known to replicate in cytoplasmic IBs, including the filovirus Lloviu virus and the highly pathogenic arenavirus Junín virus. This suggests NXF1 as a promising target for the development of broadly active antivirals.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Nucleocytoplasmic Transport Proteins , RNA, Viral , RNA-Binding Proteins , Antiviral Agents , Ebolavirus/genetics , Ebolavirus/metabolism , Humans , Inclusion Bodies, Viral/metabolism , Inclusion Bodies, Viral/virology , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
18.
Genes Cells ; 27(10): 621-628, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35950937

ABSTRACT

TAP is a general mRNA export receptor and is highly conserved among eukaryotes. The nematode Caenorhabditis elegans has another TAP-like protein, NXF-2, but little is known about its function. In this study, we show that NXF-2 is specifically expressed in germ cells and forms a novel granular structure that is different from that of P granules and that NXF-2 granules are anchored to the nuclear periphery in the mitotic region of the hermaphrodite gonad. In contrast, NXF-2 granules are released within the whole cytoplasm in the meiotic region, where the feminization gene tra-2 starts to function. Both inhibition of XPO-1 (an ortholog of the export receptor CRM1) and mutation of the nuclear export signal of NXF-2 caused the release of NXF-2 granules from the nuclear periphery, indicating that anchoring of NXF-2 granules depends on XPO-1 function. Moreover, inhibition of NXF-2 resulted in a substantial nuclear accumulation of the reporter mRNA carrying the tra-2 3'UTR. These results suggest that, together with XPO-1, NXF-2 exports and anchors tra-2 mRNA to the nuclear periphery to avoid precocious translation until the germ cells reach the meiotic region, thereby contributing to the regulation of tra-2 mRNA expression.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , 3' Untranslated Regions , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Carrier Proteins/metabolism , Germ Cells/metabolism , Membrane Proteins/metabolism , Nuclear Export Signals/genetics , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
19.
PLoS Pathog ; 18(8): e1010349, 2022 08.
Article in English | MEDLINE | ID: mdl-36007063

ABSTRACT

SARS-CoV-2 is a betacoronavirus and the etiological agent of COVID-19, a devastating infectious disease. Due to its far-reaching effect on human health, there is an urgent and growing need to understand the viral molecular biology of SARS-CoV-2 and its interaction with the host cell. SARS-CoV-2 encodes 9 predicted accessory proteins, which are presumed to be dispensable for in vitro replication, most likely having a role in modulating the host cell environment to aid viral replication. Here we show that the ORF6 accessory protein interacts with cellular Rae1 to inhibit cellular protein production by blocking mRNA export. We utilised cell fractionation coupled with mRNAseq to explore which cellular mRNA species are affected by ORF6 expression and show that ORF6 can inhibit the export of many mRNA including those encoding antiviral factors such as IRF1 and RIG-I. We also show that export of these mRNA is blocked in the context of SARS-CoV-2 infection. Together, our studies identify a novel mechanism by which SARS-CoV-2 can manipulate the host cell environment to supress antiviral responses, providing further understanding to the replication strategies of a virus that has caused an unprecedented global health crisis.


Subject(s)
COVID-19 , SARS-CoV-2 , Viral Proteins/metabolism , Antiviral Agents , COVID-19/genetics , Humans , Immunity, Innate , Nuclear Matrix-Associated Proteins , Nucleocytoplasmic Transport Proteins/genetics , RNA, Messenger/genetics
20.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 39(7): 694-697, 2022 Jul 10.
Article in Chinese | MEDLINE | ID: mdl-35810423

ABSTRACT

OBJECTIVE: To explore the genetic etiology of a child with suspected propionic acidemia. METHODS: Genomic DNA was extracted from peripheral blood sample of the child and subjected to high-throughput sequencing to screen pathogenic variants of genes associated with methylmalonic acidemia and propionic acidemia, including MUT, MMACHC, MMAA, MMAB, MMADHC, LMBRD1, PCCA, PCCB and SLC22A5. Candidate variants were verified by Sanger sequencing of the proband, her parents and sister. RESULTS: The proband was found to harbor two pathogenic variants of the MUT gene, namely c.1560+2T>C and c.729_730insTT (p.Asp244fs), but not in genes associated with propionic acidemia. Her sister and father had carried c.1560+2T>C, and her mother had carried c.729_730insTT (p.Asp244fs). CONCLUSION: The proband was diagnosed as methylmalonic acidemia due to compound heterozygous variants of c.1560+2T>C and c.729_730insTT (p.Asp244fs) of the MUT gene. Her elder sister and parents were all carriers. Genetic testing has facilitated differential diagnosis of methylmalonic acidemia and propionic acidemia in this pedigree.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Propionic Acidemia , Aged , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/genetics , Child , China , Diagnosis, Differential , Female , High-Throughput Nucleotide Sequencing , Humans , Mutation , Nucleocytoplasmic Transport Proteins/genetics , Oxidoreductases/genetics , Pedigree , Propionic Acidemia/diagnosis , Propionic Acidemia/genetics , Solute Carrier Family 22 Member 5/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...