Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Mol Biol Cell ; 31(12): 1289-1301, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32267207

ABSTRACT

The multi-component cytoplasmic dynein transports cellular cargoes with the help of another multi-component complex dynactin, but we do not know enough about factors that may affect the assembly and functions of these proteins. From a genetic screen for mutations affecting early-endosome distribution in Aspergillus nidulans, we identified the prp40AL438* mutation in Prp40A, a homologue of Prp40, an essential RNA-splicing factor in the budding yeast. Prp40A is not essential for splicing, although it associates with the nuclear splicing machinery. The prp40AL438* mutant is much healthier than the ∆prp40A mutant, but both mutants exhibit similar defects in dynein-mediated early-endosome transport and nuclear distribution. In the prp40AL438* mutant, the frequency but not the speed of dynein-mediated early-endosome transport is decreased, which correlates with a decrease in the microtubule plus-end accumulations of dynein and dynactin. Within the dynactin complex, the actin-related protein Arp1 forms a mini-filament. In a pull-down assay, the amount of Arp1 pulled down with its pointed-end protein Arp11 is lowered in the prp40AL438* mutant. In addition, we found from published interactome data that a mammalian Prp40 homologue PRPF40A interacts with Arp1. Thus, Prp40 homologues may regulate the assembly or function of dynein-dynactin and their mechanisms deserve to be further studied.


Subject(s)
Dynactin Complex/metabolism , Dyneins/metabolism , RNA Splicing Factors/genetics , Actin Cytoskeleton/metabolism , Actins/metabolism , Amino Acid Sequence/genetics , Aspergillus nidulans/genetics , Aspergillus nidulans/metabolism , Cytoskeleton/metabolism , Dynactin Complex/genetics , Dyneins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mutation/genetics , Protein Binding/genetics , RNA Splicing Factors/metabolism , RNA Splicing Factors/physiology , Ribonucleoprotein, U1 Small Nuclear/metabolism , Ribonucleoprotein, U1 Small Nuclear/physiology
2.
Biochim Biophys Acta Gene Regul Mech ; 1862(6): 634-642, 2019 06.
Article in English | MEDLINE | ID: mdl-31042550

ABSTRACT

Removal of introns by pre-mRNA splicing is fundamental to gene function in eukaryotes. However, understanding the mechanism by which exon-intron boundaries are defined remains a challenging endeavor. Published reports support that the recruitment of U1 snRNP at the 5'ss marked by GU dinucleotides defines the 5'ss as well as facilitates 3'ss recognition through cross-exon interactions. However, exceptions to this rule exist as U1 snRNP recruited away from the 5'ss retains the capability to define the splice site, where the cleavage takes place. Independent reports employing exon 7 of Survival Motor Neuron (SMN) genes suggest a long-distance effect of U1 snRNP on splice site selection upon U1 snRNP recruitment at target sequences with or without GU dinucleotides. These findings underscore that sequences distinct from the 5'ss may also impact exon definition if U1 snRNP is recruited to them through partial complementarity with the U1 snRNA. In this review we discuss the expanded role of U1 snRNP in splice-site selection due to U1 ability to be recruited at more sites than predicted solely based on GU dinucleotides.


Subject(s)
RNA Splice Sites , RNA Splicing/physiology , Ribonucleoprotein, U1 Small Nuclear/physiology , Alternative Splicing , Exons/genetics , Humans , Introns/genetics , Mutation , RNA Splicing/genetics , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , Ribonucleoprotein, U1 Small Nuclear/genetics , SMN Complex Proteins , Survival of Motor Neuron 1 Protein
3.
Mol Cell ; 71(6): 1012-1026.e3, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30174293

ABSTRACT

Pre-mRNA splicing is an essential step in the expression of most human genes. Mutations at the 5' splice site (5'ss) frequently cause defective splicing and disease due to interference with the initial recognition of the exon-intron boundary by U1 small nuclear ribonucleoprotein (snRNP), a component of the spliceosome. Here, we use a massively parallel splicing assay (MPSA) in human cells to quantify the activity of all 32,768 unique 5'ss sequences (NNN/GYNNNN) in three different gene contexts. Our results reveal that although splicing efficiency is mostly governed by the 5'ss sequence, there are substantial differences in this efficiency across gene contexts. Among other uses, these MPSA measurements facilitate the prediction of 5'ss sequence variants that are likely to cause aberrant splicing. This approach provides a framework to assess potential pathogenic variants in the human genome and streamline the development of splicing-corrective therapies.


Subject(s)
Alternative Splicing/genetics , RNA Splice Sites/genetics , RNA Splice Sites/physiology , Alternative Splicing/physiology , Carrier Proteins/genetics , Conserved Sequence/genetics , Exons , Genes, BRCA2 , HeLa Cells , Humans , Introns , Mutation , RNA Splicing/genetics , RNA Splicing/physiology , RNA, Small Nuclear/physiology , Ribonucleoprotein, U1 Small Nuclear/physiology , Spliceosomes , Survival of Motor Neuron 1 Protein/genetics , Transcriptional Elongation Factors
4.
Proc Natl Acad Sci U S A ; 113(19): 5269-74, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27114556

ABSTRACT

Alternative pre-mRNA splicing (AS) is a critical regulatory mechanism that operates extensively in the nervous system to produce diverse protein isoforms. Fruitless AS isoforms have been shown to influence male courtship behavior, but the underlying mechanisms are unknown. Using genome-wide approaches and quantitative behavioral assays, we show that the P-element somatic inhibitor (PSI) and its interaction with the U1 small nuclear ribonucleoprotein complex (snRNP) control male courtship behavior. PSI mutants lacking the U1 snRNP-interacting domain (PSIΔAB mutant) exhibit extended but futile mating attempts. The PSIΔAB mutant results in significant changes in the AS patterns of ∼1,200 genes in the Drosophila brain, many of which have been implicated in the regulation of male courtship behavior. PSI directly regulates the AS of at least one-third of these transcripts, suggesting that PSI-U1 snRNP interactions coordinate the behavioral network underlying courtship behavior. Importantly, one of these direct targets is fruitless, the master regulator of courtship. Thus, PSI imposes a specific mode of regulatory control within the neuronal circuit controlling courtship, even though it is broadly expressed in the fly nervous system. This study reinforces the importance of AS in the control of gene activity in neurons and integrated neuronal circuits, and provides a surprising link between a pleiotropic pre-mRNA splicing pathway and the precise control of successful male mating behavior.


Subject(s)
Alternative Splicing/physiology , Drosophila Proteins/physiology , Drosophila/physiology , Genes, Insect/physiology , Nuclear Proteins/physiology , RNA-Binding Proteins/physiology , Ribonucleoprotein, U1 Small Nuclear/physiology , Sexual Behavior, Animal/physiology , Animals , Courtship , Female , Male , Nerve Tissue Proteins/physiology , Sex Characteristics
5.
Adv Exp Med Biol ; 848: 51-69, 2015.
Article in English | MEDLINE | ID: mdl-25757615

ABSTRACT

U1 snRNP (U1 small nuclear ribonucleoprotein) is an essential component of the splicing machinery. U1 snRNP also plays an additional role in 3'-end mRNA processing when it binds close to polyadenylation sites (PAS). Cotranscriptionally, U1 snRNP binding close to putative PAS prevents premature cleavage and polyadenylation and consequently safeguards pre-mRNA transcripts and defines promoter directionality. At the 3'-end of mRNAs, U1 snRNP binding to putative PAS may regulate mRNA length or inhibit polyadenylation and, therefore, gene expression. U1 interference (U1i) is a technique to inhibit gene expression based on the property of U1 snRNP to inhibit polyadenylation. It requires the expression of a modified U1 snRNP, which interacts with a target gene upstream of its PAS and inhibits target gene expression. U1i has been used to inhibit the expression of reporter or endogenous genes both in tissue culture and in animal models. In addition, U1i combination with RNA interference (RNAi), another RNA-based gene silencing technology, results in a synergistic increased inhibition. This is of special interest for antiviral therapy, where strong inhibitions may be required to decrease the expression of replicative viral RNAs and impact the replication cycle. Furthermore, the combination of U1i and RNAi-based inhibitors should prevent the appearance of viral variants resistant to the treatment and allows the dose of inhibitors to be decreased and a functional inhibition to be obtained with fewer off target effects. In fact, U1i has been used to inhibit the expression of HIV-1 and HBV, whose viral genomes express mRNAs that must be polyadenylated by the nuclear polyadenylation machinery. In the case of HBV, antiviral U1i has been combined with RNAi to demonstrate a strong inhibition of expression from HBV sequences in vivo. This shows that, although several aspects of U1i technology remain to be addressed, U1i and U1i combined with RNAi have great potential as antivirals.


Subject(s)
Antiviral Agents/therapeutic use , Molecular Targeted Therapy/methods , RNA Interference , Ribonucleoprotein, U1 Small Nuclear/genetics , Virus Diseases/therapy , Animals , Humans , Molecular Conformation , RNA Interference/physiology , Ribonucleoprotein, U1 Small Nuclear/antagonists & inhibitors , Ribonucleoprotein, U1 Small Nuclear/chemistry , Ribonucleoprotein, U1 Small Nuclear/physiology , Structure-Activity Relationship
6.
J Immunol ; 188(10): 4769-75, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22490866

ABSTRACT

The NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome is a caspase-1-containing cytosolic protein complex that is essential for processing and secretion of IL-1ß. The U1-small nuclear ribonucleoprotein (U1-snRNP) that includes U1-small nuclear RNA is a highly conserved intranuclear molecular complex involved in splicing pre-mRNA. Abs against this self nuclear molecule are characteristically found in autoimmune diseases like systemic lupus erythematosus, suggesting a potential role of U1-snRNP in autoimmunity. Although endogenous DNA and microbial nucleic acids are known to activate the inflammasomes, it is unknown whether endogenous RNA-containing U1-snRNP could activate this molecular complex. In this study, we show that U1-snRNP activates the NLRP3 inflammasome in CD14(+) human monocytes dependently of anti-U1-snRNP Abs, leading to IL-1ß production. Reactive oxygen species and K(+) efflux were responsible for this activation. Knocking down the NLRP3 or inhibiting caspase-1 or TLR7/8 pathway decreased IL-1ß production from monocytes treated with U1-snRNP in the presence of anti-U1-snRNP Abs. Our findings indicate that endogenous RNA-containing U1-snRNP could be a signal that activates the NLRP3 inflammasome in autoimmune diseases like systemic lupus erythematosus where anti-U1-snRNP Abs are present.


Subject(s)
Carrier Proteins/metabolism , Monocytes/immunology , Monocytes/metabolism , Ribonucleoprotein, U1 Small Nuclear/physiology , Adult , Antibodies/physiology , Carrier Proteins/physiology , Humans , Interleukin-1beta/biosynthesis , Lipopolysaccharide Receptors/biosynthesis , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , Reactive Oxygen Species/metabolism , Ribonucleoprotein, U1 Small Nuclear/immunology
7.
Mol Cell ; 43(6): 927-39, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21925381

ABSTRACT

Alternative intronic polyadenylation (IPA) can generate truncated protein isoforms with significantly altered functions. Here, we describe 31 dominant-negative, secreted variant isoforms of receptor tyrosine kinases (RTKs) that are produced by activation of intronic poly(A) sites. We show that blocking U1-snRNP can activate IPA, indicating a larger role for U1-snRNP in RNA surveillance. Moreover, we report the development of an antisense-based method to effectively and specifically activate expression of individual soluble decoy RTKs (sdRTKs) to alter signaling, with potential therapeutic implications. In particular, a quantitative switch from signal transducing full-length vascular endothelial growth factor receptor-2 (VEGFR2/KDR) to a dominant-negative sKDR results in a strong antiangiogenic effect both on directly targeted cells and on naive cells exposed to conditioned media, suggesting a role for this approach in interfering with angiogenic paracrine and autocrine loops.


Subject(s)
Introns , Polyadenylation , Receptor Protein-Tyrosine Kinases/biosynthesis , Humans , Neovascularization, Physiologic/physiology , Poly A/chemistry , Poly A/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/chemistry , Protein Isoforms/physiology , RNA Splicing , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/physiology , Ribonucleoprotein, U1 Small Nuclear/physiology , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/physiology
8.
Dev Biol ; 350(2): 534-47, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21146519

ABSTRACT

The conserved SWI/SNF chromatin remodeling complex uses the energy from ATP hydrolysis to alter local chromatin environments through disrupting DNA-histone contacts. These alterations influence transcription activation, as well as repression. The Drosophila SWI/SNF counterpart, known as the Brahma or Brm complex, has been shown to have an essential role in regulating the proper expression of many developmentally important genes, including those required for eye and wing tissue morphogenesis. A temperature sensitive mutation in one of the core complex subunits, SNR1 (SNF5/INI1/SMARCB1), results in reproducible wing patterning phenotypes that can be dominantly enhanced and suppressed by extragenic mutations. SNR1 functions as a regulatory subunit to modulate chromatin remodeling activities of the Brahma complex on target genes, including both activation and repression. To help identify gene targets and cofactors of the Brahma complex, we took advantage of the weak dominant nature of the snr1(E1) mutation to carry out an unbiased genetic modifier screen. Using a set of overlapping chromosomal deficiencies that removed the majority of the Drosophila genome, we looked for genes that when heterozygous would function to either enhance or suppress the snr1(E1) wing pattern phenotype. Among potential targets of the Brahma complex, we identified components of the Notch, EGFR and DPP signaling pathways important for wing development. Mutations in genes encoding histone demethylase enzymes were identified as cofactors of Brahma complex function. In addition, we found that the Lysine Specific Demethylase 1 gene (lsd1) was important for the proper cell type-specific development of wing patterning.


Subject(s)
Co-Repressor Proteins/physiology , Drosophila Proteins/physiology , Drosophila/growth & development , Oxidoreductases, N-Demethylating/physiology , Ribonucleoprotein, U1 Small Nuclear/physiology , Wings, Animal/growth & development , Animals , Cell Cycle Proteins/physiology , Cells, Cultured , Transcription Factors/physiology
9.
Nucleic Acids Res ; 35(5): 1391-401, 2007.
Article in English | MEDLINE | ID: mdl-17264129

ABSTRACT

Characterization of spliceosomal complexes in the fission yeast Schizosaccharomyces pombe revealed particles sedimenting in the range of 30-60S, exclusively containing U1 snRNA. Here, we report the tandem affinity purification (TAP) of U1-specific protein complexes. The components of the complexes were identified using (LC-MS/MS) mass spectrometry. The fission yeast U1 snRNP contains 16 proteins, including the 7 Sm snRNP core proteins. In both fission and budding yeast, the U1 snRNP contains 9 and 10 U1 specific proteins, respectively, whereas the U1 particle found in mammalian cells contains only 3. Among the U1-specific proteins in S. pombe, three are homolog to the mammalian and six to the budding yeast Saccharomyces cerevisiae U1-specific proteins, whereas three, called U1H, U1J and U1L, are proteins specific to S. pombe. Furthermore, we demonstrate that the homolog of U1-70K and the three proteins specific to S. pombe are essential for growth. We will discuss the differences between the U1 snRNPs with respect to the organism-specific proteins found in the two yeasts and the resulting effect it has on pre-mRNA splicing.


Subject(s)
Ribonucleoprotein, U1 Small Nuclear/physiology , Ribonucleoproteins, Small Nuclear/physiology , Schizosaccharomyces pombe Proteins/physiology , Chromatography, Affinity , Proteomics , Ribonucleoprotein, U1 Small Nuclear/genetics , Ribonucleoprotein, U1 Small Nuclear/isolation & purification , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/isolation & purification , Schizosaccharomyces/genetics , Schizosaccharomyces/growth & development , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/isolation & purification , Spliceosomes/chemistry , Tandem Mass Spectrometry
10.
Lupus ; 15(3): 132-7, 2006.
Article in English | MEDLINE | ID: mdl-16634365

ABSTRACT

Mixed connective tissue disease (MCTD) was first described in 1972 as a disease syndrome with overlapping features of systemic sclerosis, systemic lupus erythematosus (SLE) and polymyositis associated with antibodies to RNAse sensitive extractable nuclear antigen. When the antigen was subsequently characterized as polypeptides on the U1 ribonuclear protein component of the splicesosome (U1RNP), MCTD became the first rheumatic disease syndrome to be defined by a serologic test. Clinical features include a high frequency of Raynaud's syndrome, swollen hands, sclerodactyly, arthritis, polymyositis and interstitial lung disease. Over the last 30 years there has been a continuing debate as to whether MCTD constitutes a 'distinct clinical entity'. Here, I will review the pathological, immunogenetic and clinical features of MCTD and conclude that the debate remains unresolved. The early misconception that it has a relatively good prognosis has not stood the test of time with long-term follow-up studies. These have identified a tendency for MCTD to evolve into SLE or systemic sclerosis and highlighted pulmonary hypertension and scleroderma renal crisis as important causes of death. Providing it is realized that our appreciation of the clinical features associated with anti-U1RNP have evolved over time, MCTD remains a useful concept in clinical practice. Whether it can be credited with the term 'disease' awaits the demonstration of common etiopathological events underlying the development of antibodies to U1 RNP and their associated clinical features.


Subject(s)
Mixed Connective Tissue Disease/therapy , Humans , Mixed Connective Tissue Disease/diagnosis , Mixed Connective Tissue Disease/etiology , Ribonucleoprotein, U1 Small Nuclear/chemistry , Ribonucleoprotein, U1 Small Nuclear/physiology
11.
Genes Dev ; 19(22): 2695-704, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16291644

ABSTRACT

Recent studies have revealed that transcription of noncoding, intergenic DNA is abundant among eukaryotes. However, the functions of this transcription are poorly understood. We have previously shown that in Saccharomyces cerevisiae, expression of an intergenic transcript, SRG1, represses the transcription of the adjacent gene, SER3, by transcription interference. We now show that SRG1 transcription is regulated by serine, thereby conferring regulation of SER3, a serine biosynthetic gene. This regulation requires Cha4, a serine-dependent activator that binds to the SRG1 promoter and is required for SRG1 induction in the presence of serine. Furthermore, two coactivator complexes, SAGA and Swi/Snf, are also directly required for activation of SRG1 and transcription interference of SER3. Taken together, our results elucidate a physiological role for intergenic transcription in the regulation of SER3. Moreover, our results demonstrate a mechanism by which intergenic transcription allows activators to act indirectly as repressors.


Subject(s)
DNA, Intergenic/genetics , Gene Expression Regulation, Fungal/physiology , Saccharomyces cerevisiae/genetics , Transcription, Genetic/physiology , DNA-Binding Proteins/metabolism , Drosophila Proteins/physiology , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Phosphoglycerate Dehydrogenase/biosynthesis , Phosphoglycerate Dehydrogenase/genetics , Promoter Regions, Genetic , RNA-Binding Proteins/physiology , Repressor Proteins/physiology , Ribonucleoprotein, U1 Small Nuclear/physiology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Serine/physiology , Suppression, Genetic/genetics , Trans-Activators/metabolism , Transcription Factors/physiology
12.
Development ; 132(1): 105-15, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15576411

ABSTRACT

Chromatin remodeling complexes play crucial roles in transcription and are implicated in processes including cell proliferation, differentiation and embryonic patterning. Brg1 is the catalytic subunit of the SWI/SNF chromatin remodeling complex and shows neural-enriched expression. Although early lethality of Brg1-null mice reflects its importance in embryogenesis, this phenotype precluded further study of specific Brg1-dependent developmental processes. Here, we have identified a requirement of Brg1 for both Xenopus primary neurogenesis and neuronal differentiation of mammalian P19 embryonic carcinoma cells. In Xenopus, loss of Brg1 function did not affect neural induction or neural cell fate determination. However, the Sox2-positive, proliferating neural progenitor cell population was expanded, and expression of a terminally differentiated neuronal marker, N-tubulin, was diminished upon loss of Brg1 activity, suggesting that Brg1 is required for neuronal differentiation. The ability of the bHLH transcription factors Ngnr1 and NeuroD to drive neuronal differentiation was also abolished by loss of Brg1 function, indicating that Brg1 is essential for the proneural activities of Ngnr1 and NeuroD. Consistent with this, dominant-negative interference with Brg1 function in P19 cells suppressed neuronal differentiation promoted by NeuroD2, showing the requirement of Brg1 for neuronal differentiation is conserved in mammalian cells. Finally, we discovered that Brg1 physically associates with both Ngnr1 and NeuroD and that interference with Brg1 function blocks Neurogenin3- and NeuroD2-mediated reporter gene transactivation. Together, our results demonstrate that Brg1 (and by inference the SWI/SNF complex) is required for neuronal differentiation by mediating the transcriptional activities of proneural bHLH proteins.


Subject(s)
Brain/embryology , Drosophila Proteins/physiology , Gene Expression Regulation, Developmental , Nuclear Proteins/physiology , RNA-Binding Proteins/physiology , Ribonucleoprotein, U1 Small Nuclear/physiology , Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors , Blotting, Western , Cell Cycle , Cell Differentiation , Cell Line, Tumor , Cell Lineage , Cell Proliferation , Chromatin/metabolism , Cloning, Molecular , DNA Helicases , DNA, Complementary/metabolism , Drosophila Proteins/metabolism , Histones/metabolism , Immunohistochemistry , Immunoprecipitation , In Situ Nick-End Labeling , Luciferases/metabolism , Mice , Microscopy, Fluorescence , Models, Genetic , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Nuclear Proteins/metabolism , Oligonucleotides/chemistry , Phenotype , Phylogeny , Plasmids/metabolism , Protein Binding , RNA/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U1 Small Nuclear/metabolism , Time Factors , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Tubulin/metabolism , Xenopus
13.
Arthritis Rheum ; 50(10): 3232-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15476231

ABSTRACT

OBJECTIVE: To analyze spontaneous T cell spreading against determinants of the U1-70K protein in young autoimmune MRL/lpr lupus mice, in comparison with the T cell spreading occurring in normal BALB/c mice immunized with peptide 131-151 of this protein. METHODS: Peripheral blood lymphocytes (PBLs) from both unprimed MRL/lpr mice and immunized BALB/c mice were tested for their ability to proliferate ex vivo in response to 18 overlapping peptides of the U1-70K spliceosomal protein, using assays for lymphocyte proliferation and secretion of interleukin-2. RESULTS: The proliferative response to peptides of the U1-70K protein evolved rapidly in MRL/lpr mice tested at different ages. At least 5 peptides were recognized by PBLs from 8-week-old autoimmune mice, whereas a different peptide was recognized by PBLs from MRL/lpr mice at 12 weeks of age. At 15 weeks, the proliferative response was weak or negative when assessed with any of the test peptides. At least 2 major peptides recognized by MRL/lpr PBLs were also recognized by PBLs generated in the BALB/c mice primed with peptide 131-151. We further demonstrated that, in preautoimmune MRL/lpr mice, repeated administration of phosphorylated peptide 131-151 (called P140), which was shown previously to be protective, transiently abolished T cell intramolecular spreading to other regions of the 70K protein. CONCLUSION: This is the first study to demonstrate that intramolecular T cell spreading effectively occurs in MRL/lpr mice with lupus, and that region 131-151 is important in the cascade of events observed in the murine lupus response. This sequence might originate a mechanism of tolerance spreading that leads to the beneficial effect observed in MRL/lpr mice after treatment with the phosphorylated peptide 131-151.


Subject(s)
Lupus Erythematosus, Systemic/immunology , Lymphocytes/immunology , Ribonucleoprotein, U1 Small Nuclear/physiology , Animals , Cell Division/immunology , Female , Immunization , Mice , Mice, Inbred BALB C , Mice, Inbred MRL lpr , Peptides/physiology , Phosphorylation , Ribonucleoprotein, U1 Small Nuclear/immunology
14.
RNA ; 10(9): 1388-98, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15317975

ABSTRACT

Control of Rous sarcoma virus RNA splicing depends in part on the interaction of U1 and U11 snRNPs with an intronic RNA element called the negative regulator of splicing (NRS). A 23mer RNA hairpin (NRS23) of the NRS directly binds U1 and U11 snRNPs. Mutations that disrupt base-pairing between the loop of NRS23 and U1 snRNA abolish its negative control of splicing. We have determined the solution structure of NRS23 using NOEs, torsion angles, and residual dipolar couplings that were extracted from multidimensional heteronuclear NMR spectra. Our structure showed that the 6-bp stem of NRS23 adopts a nearly A-form duplex conformation. The loop, which consists of 11 residues according to secondary structure probing, was in a closed conformation. U913, the first residue in the loop, was bulged out or dynamic, and loop residues G914-C923, G915-U922, and U916-A921 were base-paired. The remaining UUGU tetraloop sequence did not adopt a stable structure and appears flexible in solution. This tetraloop differs from the well-known classes of tetraloops (GNRA, CUYG, UNCG) in terms of its stability, structure, and function. Deletion of the bulged U913, which is not complementary to U1 snRNA, increased the melting temperature of the RNA hairpin. This hyperstable hairpin exhibited a significant decrease in binding to U1 snRNP. Thus, the structure of the NRS RNA, as well as its sequence, is important for interaction with U1 snRNP and for splicing suppression.


Subject(s)
Avian Sarcoma Viruses/genetics , Nucleic Acid Conformation , RNA Splicing , RNA, Small Nuclear/chemistry , RNA, Viral/chemistry , RNA, Viral/genetics , Base Pairing/genetics , Base Sequence , Binding Sites/genetics , Humans , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Mutation/genetics , Retroviridae/genetics , Ribonucleoprotein, U1 Small Nuclear/physiology , Ribonucleoproteins, Small Nuclear/physiology , Solutions
15.
EMBO J ; 23(2): 376-85, 2004 Jan 28.
Article in English | MEDLINE | ID: mdl-14713954

ABSTRACT

Communication between U1 and U2 snRNPs is critical during pre-spliceosome assembly; yet, direct connections have not been observed. To investigate this assembly step, we focused on Prp5, an RNA-dependent ATPase of the DExD/H family. We identified homologs of Saccharomyces cerevisiae Prp5 in humans (hPrp5) and Schizosaccharomyces pombe (SpPrp5), and investigated their interactions and function. Depletion and reconstitution of SpPrp5 from extracts demonstrate that ATP binding and hydrolysis by Prp5 are required for pre-spliceosome complex A formation. hPrp5 and SpPrp5 are each physically associated with both U1 and U2 snRNPs; Prp5 contains distinct U1- and U2-interacting domains that are required for pre-spliceosome assembly; and, we observe a Prp5-associated U1/U2 complex in S. pombe. Together, these data are consistent with Prp5 being a bridge between U1 and U2 snRNPs at the time of pre-spliceosome formation.


Subject(s)
Adenosine Triphosphatases/physiology , Introns , RNA Helicases/physiology , RNA, Messenger/metabolism , Ribonucleoprotein, U1 Small Nuclear/metabolism , Ribonucleoprotein, U2 Small Nuclear/metabolism , Schizosaccharomyces pombe Proteins/physiology , Spliceosomes/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , DEAD-box RNA Helicases , Exons , Models, Genetic , Protein Structure, Tertiary , RNA Helicases/chemistry , RNA Precursors/metabolism , RNA, Messenger/chemistry , Ribonucleoprotein, U1 Small Nuclear/physiology , Ribonucleoprotein, U2 Small Nuclear/physiology , Saccharomyces cerevisiae Proteins/chemistry , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Sequence Homology, Amino Acid
16.
J Biol Chem ; 278(21): 18997-9007, 2003 May 23.
Article in English | MEDLINE | ID: mdl-12649279

ABSTRACT

Mutations in the human tau gene leading to aberrant splicing have been identified in FTDP-17, an autosomal dominant hereditary neurodegenerative disorder. Molecular mechanisms by which such mutations cause tau aberrant splicing were not understood. We characterized two mutations in exon 10 of the tau gene, N279K and Del280K. Our results revealed an exonic splicing enhancer element located in exon 10. The activity of this AG-rich splicing enhancer was altered by N279K and Del280K mutations. This exonic enhancer element interacts with human Tra2 beta protein. The interaction between Tra2 beta and the exonic splicing enhancer correlates with the activity of this enhancer element in stimulating splicing. Biochemical studies including in vitro splicing and RNA interference experiments in transfected cells support a role for Tra2 beta protein in regulating alternative splicing of human tau gene. Our results implicate the human tau gene as a target gene for the alternative splicing regulator Tra2 beta, suggesting that Tra2 beta may play a role in aberrant tau exon 10 alternative splicing and in the pathogenesis of tauopathies.


Subject(s)
Enhancer Elements, Genetic/genetics , Exons/genetics , Microtubule-Associated Proteins/genetics , Mutation , Nerve Tissue Proteins/physiology , RNA-Binding Proteins/physiology , tau Proteins/genetics , Alternative Splicing , Biotinylation , Enhancer Elements, Genetic/physiology , HeLa Cells , Humans , Mutagenesis, Site-Directed , Neuroblastoma , RNA Precursors/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoprotein, U1 Small Nuclear/antagonists & inhibitors , Ribonucleoprotein, U1 Small Nuclear/physiology , Serine-Arginine Splicing Factors , Transfection , Tumor Cells, Cultured
17.
EMBO J ; 22(7): 1620-31, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12660168

ABSTRACT

We have investigated use of a conserved non-canonical GA 5' splice site present in vertebrate fibroblast growth factor receptor (FGFR) genes. Despite previous studies suggesting that GA at the beginning of an intron is incompatible with splicing, we observe efficient utilization of this splice site for human FGFR1 gene constructs. We show that use of the GA splice site is dependent on both a conventional splice site six nucleotides upstream and sequence elements within the downstream intron. Furthermore, our results are consistent with competition between the tandem 5' splice sites being mediated by U6 snRNP, rather than U1 snRNP. Thus the GA 5' splice site represents an extension of the adjacent conventional 5' splice site, the first natural example of such a composite 5' splice site.


Subject(s)
Alternative Splicing , Receptors, Fibroblast Growth Factor/genetics , Ribonucleoprotein, U1 Small Nuclear/physiology , Ribonucleoprotein, U4-U6 Small Nuclear/physiology , Animals , Base Sequence , Cell Line , DNA , Exons , Humans , Introns , Molecular Sequence Data , Mutagenesis , Regulatory Sequences, Nucleic Acid , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
18.
Oncogene ; 21(20): 3112-20, 2002 May 09.
Article in English | MEDLINE | ID: mdl-12082626

ABSTRACT

Ini1/hsnf5 gene encodes INI1 protein, a chromatin remodeling factor associated with the SWI/SNF complex. In yeast, this complex modifies chromatin condensation to coactivate various transcriptional factors. However, in human, little is known about the SWI/SNF complex and INI1. To elucidate cellular functions of ini1, we constructed a recombinant adenovirus (AdexHA-INI1) capable of overexpressing INI1 in ini1-deficient cells. AdexHA-INI1 produced intranuclear INI1 in three ini1-deficient cell lines, changed their morphology, and decreased the proportion of viable cells. Flow cytometry and a BrdU incorporation assay showed that after the infection, growth of these cells was partially arrested at G1. In two of the three ini1-deficient cell lines, apoptosis was found to occur after the infection, as detected by the presence of cleaved poly (ADP-ribose) polymerase. To determine functional domains of INI1, we constructed plasmids expressing INI1 and its deletion mutants, which were used for a colony formation assay. Repeats 1 and 2 of INI1 were found to be required to suppress the growth of the three ini1-deficient cell lines. The results support the hypothesis that ini1 is a tumor suppressor gene and suggest a novel link between human SWI/SNF chromatin remodeling complex and apoptosis.


Subject(s)
Apoptosis/physiology , Chromatin/metabolism , DNA-Binding Proteins/physiology , Drosophila Proteins , G1 Phase/physiology , Genes, Tumor Suppressor , RNA-Binding Proteins , Adenoviridae/genetics , Apoptosis/genetics , Child , Chromosomal Proteins, Non-Histone , Colony-Forming Units Assay , DNA Replication , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Flow Cytometry , G1 Phase/genetics , Genetic Vectors/genetics , HeLa Cells/metabolism , HeLa Cells/ultrastructure , Humans , Plasmids/genetics , Poly(ADP-ribose) Polymerases/metabolism , Recombinant Fusion Proteins/physiology , Rhabdoid Tumor/pathology , Rhabdomyosarcoma/pathology , Ribonucleoprotein, U1 Small Nuclear/physiology , SMARCB1 Protein , Sequence Deletion , Transcription Factors/physiology , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/ultrastructure
19.
Curr Opin Struct Biol ; 12(1): 82-8, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11839494

ABSTRACT

Polyadenylation at the 3' ends of mRNAs is critical to the translation and stability of the messages. Recently determined structures of poly(A) polymerase, U1A and domains of the poly(A)-binding protein provide a framework for understanding the synthesis and regulation of the poly(A) tail.


Subject(s)
Escherichia coli Proteins , Poly A/biosynthesis , RNA, Messenger/biosynthesis , Animals , Humans , Nucleic Acid Conformation , Poly A/chemistry , Poly(A)-Binding Proteins , Polynucleotide Adenylyltransferase/antagonists & inhibitors , Polynucleotide Adenylyltransferase/metabolism , Polynucleotide Adenylyltransferase/physiology , Protein Conformation , RNA, Messenger/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/physiology , Ribonucleoprotein, U1 Small Nuclear/chemistry , Ribonucleoprotein, U1 Small Nuclear/physiology , Structure-Activity Relationship , Yeasts
20.
J Biol Chem ; 276(33): 31004-15, 2001 Aug 17.
Article in English | MEDLINE | ID: mdl-11425851

ABSTRACT

The mammalian 70K protein, a component of the U1 small nuclear ribonucleoprotein involved in pre-mRNA splicing, interacts with a number of proteins important for regulating constitutive and alternative splicing. Similar proteins that interact with the yeast homolog of the 70K protein, Snp1p, have yet to be identified. We used the two-hybrid system to find four U1-Snp1 associating (Usa) proteins. Two of these proteins physically associate with Snp1p as assayed by coimmunoprecipitation. One is Prp8p, a known, essential spliceosomal component. This interaction suggests some novel functions for Snp1p and the U1 small nuclear ribonucleoprotein late in spliceosome development. The other, Exo84p, is a conserved subunit of the exocyst, an eight-protein complex functioning in secretion. We show here that Exo84p is also involved in pre-mRNA splicing. A temperature-sensitive exo84 mutation caused increased ratios of pre-mRNA to mRNA for the Rpl30 and actin transcripts in cells incubated at the non-permissive temperature. The mutation also led to a defect in splicing and prespliceosome formation in vitro; an indication that Exo84p has a direct role in splicing. The results elucidate a surprising link between splicing and secretion.


Subject(s)
Fungal Proteins/physiology , Membrane Proteins , Peptides , RNA Precursors/metabolism , RNA Splicing , Ribonucleoprotein, U1 Small Nuclear/physiology , Saccharomyces cerevisiae Proteins , Yeasts/genetics , Actins/genetics , Carrier Proteins , Fungal Proteins/chemistry , Intercellular Signaling Peptides and Proteins , Ribonucleoprotein, U4-U6 Small Nuclear , Ribonucleoprotein, U5 Small Nuclear
SELECTION OF CITATIONS
SEARCH DETAIL
...