Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Oncogene ; 37(1): 107-115, 2018 01 04.
Article in English | MEDLINE | ID: mdl-28892045

ABSTRACT

In acute myeloid leukemia (AML), specific genomic aberrations induce aberrant methylation, thus directly influencing the transcriptional programing of leukemic cells. Therefore, therapies targeting epigenetic processes are advocated as a promising therapeutic tool for AML treatment. However, to develop new therapies, a comprehensive understanding of the mechanism(s) driving the epigenetic changes as a result of acquired genetic abnormalities is necessary. This understanding is still lacking. In this study, we performed genome-wide CpG-island methylation profiling on pediatric AML samples. Six differentially methylated genomic regions within two genes, discriminating inv(16)(p13;q22) from non-inv(16) pediatric AML samples, were identified. All six regions had a hypomethylated phenotype in inv(16) AML samples, and this was most prominent at the regions encompassing the meningioma (disrupted in balanced translocation) 1 (MN1) oncogene. MN1 expression primarily correlated with the methylation level of the 3' end of the MN1 exon-1 locus. Decitabine treatment of different cell lines showed that induced loss of methylation at the MN1 locus can result in an increase of MN1 expression, indicating that MN1 expression is coregulated by DNA methylation. To investigate this methylation-associated mechanism, we determined the expression of DNA methyltransferases in inv(16) AML. We found that DNMT3B expression was significantly lower in inv(16) samples. Furthermore, DNMT3B expression correlated negatively with MN1 expression in pediatric AML samples. Importantly, depletion of DNMT3B impaired remethylation efficiency of the MN1 exon-1 locus in AML cells after decitabine exposure. These findings identify DNMT3B as an important coregulator of MN1 methylation. Taken together, this study shows that the methylation level of the MN1 exon-1 locus regulates MN1 expression levels in inv(16) pediatric AML. This methylation level is dependent on DNMT3B, thus suggesting a role for DNMT3B in leukemogenesis in inv(16) AML, through MN1 methylation regulation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Carcinogenesis/genetics , Cell Line, Tumor , Child , Child, Preschool , CpG Islands/genetics , DNA Methylation/drug effects , Decitabine , Epigenesis, Genetic/genetics , Exons/genetics , Female , Humans , Infant , Infant, Newborn , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/pathology , Male , Nucleic Acid Hybridization/methods , Oligonucleotide Array Sequence Analysis/methods , Oncogene Proteins, Fusion/genetics , Promoter Regions, Genetic/genetics , Trans-Activators , DNA Methyltransferase 3B
2.
Leukemia ; 32(3): 828-836, 2018 03.
Article in English | MEDLINE | ID: mdl-28871137

ABSTRACT

Overexpression of the BRE (brain and reproductive organ-expressed) gene defines a distinct pediatric and adult acute myeloid leukemia (AML) subgroup. Here we identify a promoter enriched for active chromatin marks in BRE intron 4 causing strong biallelic expression of a previously unknown C-terminal BRE transcript. This transcript starts with BRE intron 4 sequences spliced to exon 5 and downstream sequences, and if translated might code for an N terminally truncated BRE protein. Remarkably, the new BRE transcript was highly expressed in over 50% of 11q23/KMT2A (lysine methyl transferase 2A)-rearranged and t(8;16)/KAT6A-CREBBP cases, while it was virtually absent from other AML subsets and normal tissues. In gene reporter assays, the leukemia-specific fusion protein KMT2A-MLLT3 transactivated the intragenic BRE promoter. Further epigenome analyses revealed 97 additional intragenic promoter marks frequently bound by KMT2A in AML with C-terminal BRE expression. The corresponding genes may be part of a context-dependent KMT2A-MLLT3-driven oncogenic program, because they were higher expressed in this AML subtype compared with other groups. C-terminal BRE might be an important contributor to this program because in a case with relapsed AML, we observed an ins(11;2) fusing CHORDC1 to BRE at the region where intragenic transcription starts in KMT2A-rearranged and KAT6A-CREBBP AML.


Subject(s)
Gene Rearrangement , Leukemia, Myeloid, Acute/genetics , Nerve Tissue Proteins/genetics , Protein Interaction Domains and Motifs/genetics , Transcriptional Activation , Translocation, Genetic , Cell Line , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 16 , Epigenesis, Genetic , Exons , Gene Expression Regulation, Leukemic , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Introns , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic
3.
Oncogene ; 35(14): 1822-31, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26165837

ABSTRACT

MicroRNAs (miRNAs) are crucial components of homeostatic and developmental gene regulation. In turn, dysregulation of miRNA expression is a common feature of different types of cancer, which can be harnessed therapeutically. Here we identify miR-139-5p suppression across several cytogenetically defined acute myeloid leukemia (AML) subgroups. The promoter of mir-139 was transcriptionally silenced and could be reactivated by histone deacetylase inhibitors in a dose-dependent manner. Restoration of mir-139 expression in cell lines representing the major AML subgroups (t[8;21], inv[16], mixed lineage leukemia-rearranged and complex karyotype AML) caused cell cycle arrest and apoptosis in vitro and in xenograft mouse models in vivo. During normal hematopoiesis, mir-139 is exclusively expressed in terminally differentiated neutrophils and macrophages. Ectopic expression of mir-139 repressed proliferation of normal CD34(+)-hematopoietic stem and progenitor cells and perturbed myelomonocytic in vitro differentiation. Mechanistically, mir-139 exerts its effects by repressing the translation initiation factor EIF4G2, thereby reducing overall protein synthesis while specifically inducing the translation of cell cycle inhibitor p27(Kip1). Knockdown of EIF4G2 recapitulated the effects of mir-139, whereas restoring EIF4G2 expression rescued the mir-139 phenotype. Moreover, elevated miR-139-5p expression is associated with a favorable outcome in a cohort of 165 pediatric patients with AML. Thus, mir-139 acts as a global tumor suppressor-miR in AML by controlling protein translation. As AML cells are dependent on high protein synthesis rates controlling the expression of mir-139 constitutes a novel path for the treatment of AML.


Subject(s)
Eukaryotic Initiation Factor-4G/genetics , Leukemia, Myeloid/genetics , MicroRNAs/biosynthesis , Protein Biosynthesis , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Eukaryotic Initiation Factor-4G/biosynthesis , Female , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Humans , Leukemia, Myeloid/pathology , Male , Mice , MicroRNAs/genetics , Xenograft Model Antitumor Assays
4.
Leukemia ; 28(5): 1022-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24270738

ABSTRACT

MicroRNAs (miRNAs) play a pivotal role in the regulation of hematopoiesis and development of leukemia. Great interest emerged in modulating miRNA expression for therapeutic purposes. In order to identify miRNAs, which specifically suppress leukemic growth of acute myeloid leukemia (AML) with t(8;21), inv(16) or mixed lineage leukemia (MLL) rearrangement by inducing differentiation, we conducted a miRNA expression profiling in a cohort of 90 cytogenetically characterized, de novo pediatric AML cases. Four miRNAs, specifically downregulated in MLL-rearranged, t(8;21) or inv(16) AMLs, were characterized by their tumor-suppressive properties in cell lines representing those respective cytogenetic groups. Among those, forced expression of miR-9 reduced leukemic growth and induced monocytic differentiation of t(8;21) AML cell lines in vitro and in vivo. The tumor-suppressive functions of miR-9 were specifically restricted to AML cell lines and primary leukemic blasts with t(8;21). On the other hand, these functions were not evident in AML blasts from patients with MLL rearrangements. We showed that miR-9 exerts its effects through the cooperation with let-7 to repress the oncogenic LIN28B/HMGA2 axis. Thus, miR-9 is a tumor suppressor-miR which acts in a stringent cell context-dependent manner.


Subject(s)
Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Genes, Tumor Suppressor , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , Translocation, Genetic , Animals , Cell Division , Child , Female , Flow Cytometry , Heterografts , Humans , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Nude
5.
FEBS Lett ; 586(8): 1101-10, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22575643

ABSTRACT

RNA-binding proteins play a crucial role in the post-transcriptional regulation of gene expression. Polypyrimidine tract binding protein (PTB in humans) has been extensively characterized as an important splicing factor, and has additional functions in 3' end processing and translation. ROD1 is a PTB paralog containing four RRM (RNA recognition motif) domains. Here, we discover a function of ROD1 in nonsense-mediated mRNA decay (NMD). We show that ROD1 and the core NMD factor UPF1 interact and co-regulate an extensive number of target genes. Using a reporter system, we demonstrate that ROD1, similarly to UPF1 and UPF2, is required for the destabilization of a known NMD substrate. Finally, we show through RIP-seq that ROD1 and UPF1 associate with a significant number of common transcripts.


Subject(s)
Nonsense Mediated mRNA Decay , Polypyrimidine Tract-Binding Protein/metabolism , RNA, Messenger/metabolism , Codon, Nonsense , HEK293 Cells , Humans , Polypyrimidine Tract-Binding Protein/genetics , RNA Stability
6.
Curr Top Microbiol Immunol ; 285: 109-38, 2005.
Article in English | MEDLINE | ID: mdl-15609502

ABSTRACT

The separation of transcription in the nucleus and translation in the cytoplasm requires nucleo-cytoplasmic exchange of proteins and RNAs. Viruses have evolved strategies to capitalize on the nucleo-cytoplasmic trafficking machinery of the cell. Here, we first discuss the principal mechanisms of receptor-mediated nuclear import of proteinaceous cargo through the nuclear pore complex, the gate keeper of the cell nucleus. We then focus on viral strategies leading to nuclear import of genomes and subgenomic particles. Nucleo-cytoplasmic transport is directly important for those viruses that are replicating in the nucleus, such as DNA tumor viruses and RNA viruses, including parvoviruses, the DNA retroviruses hepadnaviruses, RNA-retrotransposons and retroviruses, adenoviruses, herpesviruses, papovaviruses, and particular negative-sense RNA viruses, such as the orthomyxovirus influenza virus. The viral strategies of nuclear import turn out to be surprisingly diverse. Their investigation continues to give insight into how nucleic acids pass in and out of the nucleus.


Subject(s)
DNA Virus Infections/metabolism , DNA Viruses/physiology , Nuclear Pore/metabolism , RNA Virus Infections/metabolism , RNA Viruses/physiology , Virus Physiological Phenomena , Animals , Humans , Nuclear Proteins/metabolism , Receptors, Virus/metabolism , Viral Proteins/metabolism
7.
EMBO J ; 20(20): 5703-14, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11598013

ABSTRACT

The nuclear pore complex (NPC) is a large proteinaceous structure through which bidirectional transport of macromolecules across the nuclear envelope (NE) takes place. Nup153 is a peripheral NPC component that has been implicated in protein and RNP transport and in the interaction of NPCs with the nuclear lamina. Here, Nup153 is localized by immunogold electron microscopy to a position on the nuclear ring of the NPC. Nuclear reconstitution is used to investigate the role of Nup153 in nucleo- cytoplasmic transport and NPC architecture. NPCs assembled in the absence of Nup153 lacked several nuclear basket components, were unevenly distributed in the NE and, unlike wild-type NPCs, were mobile within the NE. Importin alpha/beta-mediated protein import into the nucleus was strongly reduced in the absence of Nup153, while transportin-mediated import was unaffected. This was due to a reduction in import complex translocation rather than to defective receptor recycling. Our results therefore reveal functions for Nup153 in NPC assembly, in anchoring NPCs within the NE and in mediating specific nuclear import events.


Subject(s)
Nuclear Pore Complex Proteins/physiology , Nuclear Pore/physiology , Nuclear Proteins/metabolism , Protein Transport/physiology , Animals , Cattle , Female , Heterogeneous-Nuclear Ribonucleoproteins , Humans , Immunohistochemistry , Karyopherins/metabolism , Macromolecular Substances , Male , Microscopy, Immunoelectron , Nuclear Pore/ultrastructure , Nuclear Proteins/genetics , Nucleoplasmins , Oocytes , Phosphoproteins/genetics , Phosphoproteins/metabolism , Recombinant Fusion Proteins/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Serum Albumin, Bovine/genetics , Serum Albumin, Bovine/metabolism , Xenopus laevis
8.
EMBO Rep ; 2(10): 926-32, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11571268

ABSTRACT

We investigated the role of RanBP3, a nuclear member of the Ran-binding protein 1 family, in CRM1-mediated protein export in higher eukaryotes. RanBP3 interacts directly with CRM1 and also forms a trimeric complex with CRM1 and RanGTP. However, RanBP3 does not bind to CRM1 like an export substrate. Instead, it can stabilize CRM1-export substrate interaction. Nuclear RanBP3 stimulates CRM1-dependent protein export in permeabilized cells. These data indicate that RanBP3 functions by a novel mechanism as a cofactor in recognition and export of certain CRM1 substrates. In vitro, RanBP3 binding to CRM1 affects the relative affinity of CRM1 for different substrates.


Subject(s)
Carrier Proteins/metabolism , Carrier Proteins/physiology , Cell Nucleus/metabolism , Karyopherins/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Nucleocytoplasmic Transport Proteins , Receptors, Cytoplasmic and Nuclear , Dose-Response Relationship, Drug , GTP Phosphohydrolases/metabolism , Glutathione Transferase/metabolism , HeLa Cells , Humans , Karyopherins/chemistry , Kinetics , Plasmids/metabolism , Protein Binding , Substrate Specificity , Time Factors , ran GTP-Binding Protein/metabolism , Exportin 1 Protein
9.
Nat Cell Biol ; 3(12): 1092-100, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11781571

ABSTRACT

Adenovirus type 2 (Ad2) imports its DNA genome through the nuclear pore complex (NPC) of cells in interphase for viral production. Here we identify the NPC-filament protein CAN/Nup214 as a docking site for incoming Ad2 capsids. Binding to CAN is independent of cytosolic factors. Capsids disassemble at NPCs to free their DNA for import. This process requires binding of nuclear histone H1 to the stably docked capsids and involves H1-import factors, restricting this irreversible process to the proximity of the nucleus. Our results provide a molecular mechanism for disassembly of Ad2 and reveal an unexpected function of histone H1 in virus-mediated DNA import.


Subject(s)
Adenoviridae/genetics , Capsid Proteins , DNA, Viral/pharmacokinetics , Histones/metabolism , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore/metabolism , Active Transport, Cell Nucleus/physiology , Amino Acid Sequence , Antibodies/pharmacology , Capsid/genetics , Capsid/immunology , Capsid/metabolism , Histones/immunology , Humans , Lung Neoplasms , Molecular Sequence Data , Nuclear Pore Complex Proteins/immunology , Protein Binding/physiology , Tumor Cells, Cultured , alpha Karyopherins/metabolism , beta Karyopherins/metabolism
10.
Chem Biol ; 7(5): 345-54, 2000 May.
Article in English | MEDLINE | ID: mdl-10801471

ABSTRACT

BACKGROUND: Anti-idiotype approaches are based on the assumption that an antibody recognising a ligand can be structurally related to the receptor. Recently we have generated anti-idiotype RNA aptamers designed to mimic the human immunodeficiency virus-1 (HIV-1) Rev nuclear export signal (NES). Nuclear injection of either NES-peptide conjugates or aptamer causes the inhibition of Rev-mediated export. This implied that NES mimics and export substrate might compete for binding to the NES receptor. The mechanism of inhibition, however, is unknown. RESULTS: The interaction between the export aptamer and CRM1 was characterised in vitro. The aptamer binds specifically to CRM1 and this interaction is sensitive to competition by Rev NES-peptide conjugates. The recognition domain of CRM1 has been mapped and includes residues found previously to affect binding of leptomycin B, a fungicide interfering with nuclear export. CONCLUSIONS: Inhibition of Rev-mediated export in vivo by export aptamers appears to result from the binding of the aptamers to the NES-recognition domain of CRM1. This observation demonstrates that anti-idiotype RNA can mimic faithfully structural and functional properties of a protein and can be used to map ligand-binding domains of receptors.


Subject(s)
Antibodies, Anti-Idiotypic/chemistry , Binding Sites, Antibody , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Karyopherins , RNA/chemistry , Receptors, Cytoplasmic and Nuclear , Transcription Factors/metabolism , Antibiotics, Antineoplastic/metabolism , Antibiotics, Antineoplastic/pharmacology , Antibodies, Anti-Idiotypic/metabolism , Biological Transport , Carrier Proteins/chemistry , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , Gene Products, rev/chemistry , Gene Products, rev/metabolism , HIV-1 , Humans , Leucine/metabolism , Molecular Mimicry , Nucleic Acid Conformation , Peptides/metabolism , Peptides/pharmacology , RNA/metabolism , RNA/pharmacology , Structure-Activity Relationship , Transcription Factors/chemistry , rev Gene Products, Human Immunodeficiency Virus , Exportin 1 Protein
11.
Mol Cell Biol ; 19(10): 6543-53, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10490594

ABSTRACT

Yeast strains lacking the yeast nuclear cap-binding complex (yCBC) are viable, although impaired in growth. We have taken advantage of this observation to carry out a genetic screen for components that show synthetic lethality (SL) with a cbp20-Delta cbp80-Delta double mutation. One set of SL interactions was due to mutations that were complemented by components of U1 small nuclear RNP (snRNP) and the yeast splicing commitment complex. These interactions confirm the role of yCBC in commitment complex formation. Physical interaction of yCBC with the commitment complex components Mud10p and Mud2p, which may directly mediate yCBC function, was demonstrated. Unexpectedly, we identified multiple SL mutations that were complemented by Cbf5p and Nop58p. These are components of the two major classes of yeast small nucleolar RNPs, which function in the maturation of rRNA precursors. Mutants lacking yCBC were found to be defective in rRNA processing. Analysis of the yCBC deletion phenotype suggests that this is likely to be due to a defect in the splicing of a subset of ribosomal protein mRNA precursors.


Subject(s)
Nuclear Proteins/metabolism , RNA Caps/metabolism , RNA Splicing , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/genetics , Gene Deletion , Genes, Lethal , Genetic Complementation Test , Nuclear Proteins/genetics , Protein Binding , RNA Cap-Binding Proteins , RNA Precursors/metabolism , RNA, Messenger/biosynthesis , RNA, Ribosomal/biosynthesis , RNA-Binding Proteins/genetics
12.
Genes Dev ; 13(18): 2425-38, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10500099

ABSTRACT

The characterization of a novel yeast-splicing factor, Luc7p, is presented. The LUC7 gene was identified by a mutation that causes lethality in a yeast strain lacking the nuclear cap-binding complex (CBC). Luc7p is similar in sequence to metazoan proteins that have arginine-serine and arginine-glutamic acid repeat sequences characteristic of a family of splicing factors. We show that Luc7p is a component of yeast U1 snRNP and is essential for vegetative growth. The composition of yeast U1 snRNP is altered in luc7 mutant strains. Extracts of these strains are unable to support any of the defined steps of splicing unless recombinant Luc7p is added. Although the in vivo defect in splicing wild-type reporter introns in a luc7 mutant strain is comparatively mild, splicing of introns with nonconsensus 5' splice site or branchpoint sequences is more defective in the mutant strain than in wild-type strains. By use of reporters that have two competing 5' splice sites, a loss of efficient splicing to the cap proximal splice site is observed in luc7 cells, analogous to the defect seen in strains lacking CBC. CBC can be coprecipitated with U1 snRNP from wild-type, but not from luc7, yeast strains. These data suggest that the loss of Luc7p disrupts U1 snRNP-CBC interaction, and that this interaction contributes to normal 5' splice site recognition.


Subject(s)
RNA Precursors/metabolism , RNA Splicing/physiology , Ribonucleoprotein, U1 Small Nuclear/physiology , Alternative Splicing/physiology , Amino Acid Sequence , Evolution, Molecular , Fungal Proteins/genetics , Humans , Models, Genetic , Molecular Sequence Data , Mutation , RNA Caps/metabolism , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/genetics , Sequence Homology, Amino Acid , Temperature , Time Factors
13.
Mol Cell Biol ; 19(9): 6276-85, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10454574

ABSTRACT

CRM1 is an export receptor mediating rapid nuclear exit of proteins and RNAs to the cytoplasm. CRM1 export cargoes include proteins with a leucine-rich nuclear export signal (NES) that bind directly to CRM1 in a trimeric complex with RanGTP. Using a quantitative CRM1-NES cargo binding assay, significant differences in affinity for CRM1 among natural NESs are demonstrated, suggesting that the steady-state nucleocytoplasmic distribution of shuttling proteins could be determined by the relative strengths of their NESs. We also show that a trimeric CRM1-NES-RanGTP complex is disassembled by RanBP1 in the presence of RanGAP, even though RanBP1 itself contains a leucine-rich NES. Selection of CRM1-binding proteins from Xenopus egg extract leads to the identification of an NES-containing DEAD-box helicase, An3, that continuously shuttles between the nucleus and the cytoplasm. In addition, we identify the Xenopus homologue of the nucleoporin CAN/Nup214 as a RanGTP- and NES cargo-specific binding site for CRM1, suggesting that this nucleoporin plays a role in export complex disassembly and/or CRM1 recycling.


Subject(s)
Carrier Proteins/metabolism , Karyopherins , Nuclear Pore Complex Proteins , Nuclear Proteins/metabolism , RNA Helicases/metabolism , Receptors, Cytoplasmic and Nuclear , Amino Acid Sequence , Animals , Base Sequence , Biological Transport, Active , Cell Nucleus/metabolism , DNA Primers/genetics , Female , GTP-Binding Proteins/metabolism , Humans , In Vitro Techniques , Models, Biological , Molecular Sequence Data , Oocytes/metabolism , Protein Binding , RNA Helicases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Xenopus , ran GTP-Binding Protein , Exportin 1 Protein
14.
J Cell Biol ; 145(2): 237-54, 1999 Apr 19.
Article in English | MEDLINE | ID: mdl-10209021

ABSTRACT

Gle2p is implicated in nuclear export of poly(A)+ RNA and nuclear pore complex (NPC) structure and distribution in Saccharomyces cerevisiae. Gle2p is anchored at the nuclear envelope (NE) via a short Gle2p-binding motif within Nup116p called GLEBS. The molecular mechanism by which Gle2p and the Gle2p-Nup116p interaction function in mRNA export is unknown. Here we show that RAE1, the mammalian homologue of Gle2p, binds to a GLEBS-like NUP98 motif at the NPC through multiple domains that include WD-repeats and a COOH-terminal non-WD-repeat extension. This interaction is direct, as evidenced by in vitro binding studies and chemical cross-linking. Microinjection experiments performed in Xenopus laevis oocytes demonstrate that RAE1 shuttles between the nucleus and the cytoplasm and is exported from the nucleus in a temperature-dependent and RanGTP-independent manner. Docking of RAE1 to the NE is highly dependent on new mRNA synthesis. Overexpression of the GLEBS-like motif also inhibits NE binding of RAE1 and induces nuclear accumulation of poly(A)+ RNA. Both effects are abrogated either by the introduction of point mutations in the GLEBS-like motif or by overexpression of RAE1, indicating a direct role for RAE1 and the NUP98-RAE1 interaction in mRNA export. Together, our data suggest that RAE1 is a shuttling transport factor that directly contributes to nuclear export of mRNAs through its ability to anchor to a specific NUP98 motif at the NPC.


Subject(s)
Fungal Proteins/metabolism , Membrane Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Matrix-Associated Proteins , Nuclear Pore Complex Proteins , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces pombe Proteins , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cricetinae , Cross-Linking Reagents , Dactinomycin/pharmacology , Female , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Nuclear Envelope/physiology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Oocytes/physiology , RNA Polymerase II/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Repetitive Sequences, Amino Acid , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Xenopus laevis
15.
Curr Biol ; 8(6): 305-14, 1998 Mar 12.
Article in English | MEDLINE | ID: mdl-9512417

ABSTRACT

BACKGROUND: Transport of macromolecules between the nucleus and cytoplasm of eukaryotic cells is mediated by nuclear import and export receptors. The receptors identified to date are members of a family of Ran GTPase-binding proteins whose founding member is importin-beta. Interaction between these receptors and their cargo is regulated by the GTP-bound form of Ran. Export complexes form and import complexes disassemble on binding of RanGTP to the receptor. Yeast Los 1 p is a member of the importin-beta family with a poorly defined role in tRNA production. RESULTS: A human member of the importin-beta family that is distantly related to Los 1 p (21% identity) has been characterized. The protein shuttled between the nucleus and cytoplasm and interacts with tRNA in a RanGTP-dependent manner. Injection of the protein into the nuclei of Xenopus oocytes resulted in a specific stimulation of the export of tRNA from the nucleus and in relief of the competitive inhibition of tRNA export caused by the introduction of saturating amounts of nuclear tRNA. CONCLUSIONS: The human protein has the functional properties expected of a transport receptor that mediates export of tRNA from the nucleus. We therefore name the protein Exportin(tRNA).


Subject(s)
Carrier Proteins/chemistry , Nuclear Proteins/chemistry , Nucleocytoplasmic Transport Proteins , RNA, Transfer/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Animals , Blotting, Western , Carrier Proteins/isolation & purification , Electrophoresis, Polyacrylamide Gel , Humans , Nuclear Proteins/isolation & purification , Polymerase Chain Reaction , Receptors, Cytoplasmic and Nuclear/isolation & purification , Sequence Homology, Amino Acid , Xenopus laevis
17.
Cell ; 90(6): 1051-60, 1997 Sep 19.
Article in English | MEDLINE | ID: mdl-9323133

ABSTRACT

CRM1 is distantly related to receptors that mediate nuclear protein import and was previously shown to interact with the nuclear pore complex. Overexpression of CRM1 in Xenopus oocytes stimulates Rev and U snRNA export from the nucleus. Conversely, leptomycin B, a cytotoxin that is shown to bind to CRM1 protein, specifically inhibits the nuclear export of Rev and U snRNAs. In vitro, CRM1 forms a leptomycin B-sensitive complex involving cooperative binding of both RanGTP and the nuclear export signal (NES) from either the Rev or PKI proteins. We conclude that CRM1 is an export receptor for leucine-rich nuclear export signals and discuss a model for the role of RanGTP in CRM1 function and in nuclear export in general.


Subject(s)
Carrier Proteins/metabolism , Fungal Proteins/metabolism , Karyopherins , Protein Sorting Signals/physiology , Receptors, Cytoplasmic and Nuclear , Animals , Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Biological Transport/drug effects , Biological Transport/physiology , Carrier Proteins/chemistry , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , Fungal Proteins/chemistry , GTP-Binding Proteins/metabolism , Gene Products, rev/metabolism , Leucine/analysis , Nuclear Proteins/metabolism , Oocytes/physiology , Protein Sorting Signals/drug effects , RNA, Small Nuclear/metabolism , Transcription Factors/metabolism , Xenopus , ran GTP-Binding Protein , Exportin 1 Protein
19.
EMBO J ; 16(4): 807-16, 1997 Feb 17.
Article in English | MEDLINE | ID: mdl-9049309

ABSTRACT

The oncogenic nucleoporin CAN/Nup214 is essential in vertebrate cells. Its depletion results in defective nuclear protein import, inhibition of messenger RNA export and cell cycle arrest. We recently found that CAN associates with proteins of 88 and 112 kDa, which we have now cloned and characterized. The 88 kDa protein is a novel nuclear pore complex (NPC) component, which we have named Nup88. Depletion of CAN from the NPC results in concomitant loss of Nup88, indicating that the localization of Nup88 to the NPC is dependent on CAN binding. The 112 kDa protein is the human homologue of yeast CRM1, a protein known to be required for maintenance of correct chromosome structure. This human CRM1 (hCRM1) localized to the NPC as well as to the nucleoplasm. Nuclear overexpression of the FG-repeat region of CAN, containing its hCRM1-interaction domain, resulted in depletion of hCRM1 from the NPC. In CAN-/- mouse embryos lacking CAN, hCRM1 remained in the nuclear envelope, suggesting that this protein can also bind to other repeat-containing nucleoporins. Lastly, hCRM1 shares a domain of significant homology with importin-beta, a cytoplasmic transport factor that interacts with nucleoporin repeat regions. We propose that hCRM1 is a soluble nuclear transport factor that interacts with the NPC.


Subject(s)
Carrier Proteins/genetics , Fungal Proteins/genetics , Karyopherins , Membrane Proteins/genetics , Nuclear Pore Complex Proteins , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear , Amino Acid Sequence , Animals , Base Sequence , Blastocyst , Carrier Proteins/analysis , Carrier Proteins/chemistry , Cell Line , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cloning, Molecular , Dactinomycin/pharmacology , Fungal Proteins/analysis , Fungal Proteins/isolation & purification , Humans , Membrane Proteins/analysis , Membrane Proteins/chemistry , Membrane Proteins/isolation & purification , Mice , Molecular Sequence Data , Molecular Weight , Nuclear Envelope/chemistry , Nuclear Envelope/metabolism , Nuclear Proteins/analysis , Nuclear Proteins/chemistry , Nuclear Proteins/isolation & purification , Protein Binding , RNA Polymerase I/antagonists & inhibitors , Saccharomyces cerevisiae , Sequence Analysis, DNA , Sequence Homology, Amino Acid , beta Karyopherins , Exportin 1 Protein
20.
Genes Dev ; 10(24): 3156-69, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8985184

ABSTRACT

Neuraminidases (sialidases) have an essential role in the removal of terminal sialic acid residues from sialoglycoconjugates and are distributed widely in nature. The human lysosomal enzyme occurs in complex with beta-galactosidase and protective protein/cathepsin A (PPCA), and is deficient in two genetic disorders: sialidosis, caused by a structural defect in the neuraminidase gene, and galactosialidosis, in which the loss of neuraminidase activity is secondary to a deficiency of PPCA. We identified a full-length cDNA clone in the dbEST data base, of which the predicted amino acid sequence has extensive homology to other mammalian and bacterial neuraminidases, including the F(Y)RIP domain and "Asp-boxes." In situ hybridization localized the human neuraminidase gene to chromosome band 6p21, a region known to contain the HLA locus. Transient expression of the cDNA in deficient human fibroblasts showed that the enzyme is compartmentalized in lysosomes and restored neuraminidase activity in a PPCA-dependent manner. The authenticity of the cDNA was verified by the identification of three independent mutations in the open reading frame of the mRNA from clinically distinct sialidosis patients. Coexpression of the mutant cDNAs with PPCA failed to generate neuraminidase activity, confirming the inactivating effect of the mutations. These results establish the molecular basis of sialidosis in these patients, and clearly identify the cDNA-encoded protein as lysosomal neuraminidase.


Subject(s)
Lysosomal Storage Diseases/genetics , Lysosomes/enzymology , Neuraminidase/genetics , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 6 , DNA, Complementary/genetics , Gene Expression , Gene Expression Regulation, Enzymologic , Genetic Complementation Test , Humans , In Situ Hybridization, Fluorescence , Lysosomal Storage Diseases/enzymology , Molecular Sequence Data , Neuraminidase/deficiency , RNA, Messenger/genetics , Sequence Alignment , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...