Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Oncogene ; 41(18): 2540-2554, 2022 04.
Article in English | MEDLINE | ID: mdl-35301407

ABSTRACT

Low-risk gestational trophoblastic neoplasia including choriocarcinoma is often effectively treated with Methotrexate (MTX) as a first line therapy. However, MTX resistance (MTX-R) occurs in at least ≈33% of cases. This can sometimes be salvaged with actinomycin-D but often requires more toxic combination chemotherapy. Moreover, additional therapy may be needed and, for high-risk patients, 5% still die from the multidrug-resistant disease. Consequently, new treatments that are less toxic and could reverse MTX-R are needed. Here, we compared the proteome/phosphoproteome of MTX-resistant and sensitive choriocarcinoma cells using quantitative mass-spectrometry to identify therapeutically actionable molecular changes associated with MTX-R. Bioinformatics analysis of the proteomic data identified cell cycle and DNA damage repair as major pathways associated with MTX-R. MTX-R choriocarcinoma cells undergo cell cycle delay in G1 phase that enables them to repair DNA damage more efficiently through non-homologous end joining in an ATR-dependent manner. Increased expression of cyclin-dependent kinase 4 (CDK4) and loss of p16Ink4a in resistant cells suggested that CDK4 inhibition may be a strategy to treat MTX-R choriocarcinoma. Indeed, inhibition of CDK4/6 using genetic silencing or the clinically relevant inhibitor, Palbociclib, induced growth inhibition both in vitro and in an orthotopic in vivo mouse model. Finally, targeting the ATR pathway, genetically or pharmacologically, re-sensitised resistant cells to MTX in vitro and potently prevented the growth of MTX-R tumours in vivo. In short, we identified two novel therapeutic strategies to tackle MTX-R choriocarcinoma that could rapidly be translated into the clinic.


Subject(s)
Choriocarcinoma , Cyclin-Dependent Kinase 6/metabolism , Methotrexate , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Choriocarcinoma/drug therapy , Choriocarcinoma/genetics , Choriocarcinoma/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Dactinomycin , Female , Humans , Methotrexate/pharmacology , Mice , Pregnancy , Proteomics
3.
Mol Microbiol ; 112(6): 1847-1862, 2019 12.
Article in English | MEDLINE | ID: mdl-31562654

ABSTRACT

Mycobacterium tuberculosis (Mtb) is able to persist in the body through months of multi-drug therapy. Mycobacteria possess a wide range of regulatory proteins, including the protein kinase B (PknB) which controls peptidoglycan biosynthesis during growth. Here, we observed that depletion of PknB resulted in specific transcriptional changes that are likely caused by reduced phosphorylation of the H-NS-like regulator Lsr2 at threonine 112. The activity of PknB towards this phosphosite was confirmed with purified proteins, and this site was required for adaptation of Mtb to hypoxic conditions, and growth on solid media. Like H-NS, Lsr2 binds DNA in sequence-dependent and non-specific modes. PknB phosphorylation of Lsr2 reduced DNA binding, measured by fluorescence anisotropy and electrophoretic mobility shift assays, and our NMR structure of phosphomimetic T112D Lsr2 suggests that this may be due to increased dynamics of the DNA-binding domain. Conversely, the phosphoablative T112A Lsr2 had increased binding to certain DNA sites in ChIP-sequencing, and Mtb containing this variant showed transcriptional changes that correspond with the change in DNA binding. In summary, PknB controls Mtb growth and adaptations to the changing host environment by phosphorylating the global transcriptional regulator Lsr2.


Subject(s)
DNA-Binding Proteins/metabolism , Mycobacterium tuberculosis/growth & development , Proto-Oncogene Proteins c-akt/metabolism , Bacterial Proteins/metabolism , Chromatin Immunoprecipitation Sequencing/methods , DNA-Binding Proteins/physiology , Electrophoretic Mobility Shift Assay/methods , Gene Expression Regulation, Bacterial/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/physiology , Threonine/metabolism , Transcription Factors/metabolism
4.
Sci Rep ; 9(1): 8337, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31171861

ABSTRACT

Protein phosphatase PstP is conserved throughout the Actinobacteria in a genetic locus related to cell wall synthesis and cell division. In many Actinobacteria it is the sole annotated serine threonine protein phosphatase to counter the activity of multiple serine threonine protein kinases. We used transcriptional knockdown, electron microscopy and comparative phosphoproteomics to investigate the putative dual functions of PstP as a specific regulator of cell division and as a global regulator of protein phosphorylation. Comparative phosphoproteomics in the early stages of PstP depletion showed hyperphosphorylation of protein kinases and their substrates, confirming PstP as a negative regulator of kinase activity and global serine and threonine phosphorylation. Analysis of the 838 phosphorylation sites that changed significantly, suggested that PstP may regulate diverse phosphoproteins, preferentially at phosphothreonine near acidic residues, near the protein termini, and within membrane associated proteins. Increased phosphorylation of the activation loop of protein kinase B (PknB) and of the essential PknB substrate CwlM offer possible explanations for the requirement for pstP for growth and for cell wall defects when PstP was depleted.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Mycobacterium smegmatis/enzymology , Phosphoproteins/metabolism , Cell Wall/metabolism , Computer Simulation , Microscopy, Electron , Multigene Family , Mutation , Mycobacterium smegmatis/genetics , Mycobacterium tuberculosis/genetics , Phosphorylation , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Proteomics
5.
Cell Rep ; 25(1): 57-67.e5, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30282038

ABSTRACT

Tuberculosis claims >1 million lives annually, and its causative agent Mycobacterium tuberculosis is a highly successful pathogen. Protein kinase B (PknB) is reported to be critical for mycobacterial growth. Here, we demonstrate that PknB-depleted M. tuberculosis can replicate normally and can synthesize peptidoglycan in an osmoprotective medium. Comparative phosphoproteomics of PknB-producing and PknB-depleted mycobacteria identify CwlM, an essential regulator of peptidoglycan synthesis, as a major PknB substrate. Our complementation studies of a cwlM mutant of M. tuberculosis support CwlM phosphorylation as a likely molecular basis for PknB being essential for mycobacterial growth. We demonstrate that growing mycobacteria produce two forms of CwlM: a non-phosphorylated membrane-associated form and a PknB-phosphorylated cytoplasmic form. Furthermore, we show that the partner proteins for the phosphorylated and non-phosphorylated forms of CwlM are FhaA, a fork head-associated domain protein, and MurJ, a proposed lipid II flippase, respectively. From our results, we propose a model in which CwlM potentially regulates both the biosynthesis of peptidoglycan precursors and their transport across the cytoplasmic membrane.


Subject(s)
Mycobacterium tuberculosis/enzymology , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Amino Acid Sequence , Cell Wall/enzymology , Mycobacterium tuberculosis/cytology , Mycobacterium tuberculosis/growth & development , Phosphorylation , Proto-Oncogene Proteins c-akt/deficiency
6.
J Cell Physiol ; 232(11): 2985-2995, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28369848

ABSTRACT

Patients with end-stage renal disease (ESRD) have elevated circulating calcium (Ca) and phosphate (Pi), and exhibit accelerated progression of calcific aortic valve disease (CAVD). We hypothesized that matrix vesicles (MVs) initiate the calcification process in CAVD. Ca induced rat valve interstitial cells (VICs) calcification at 4.5 mM (16.4-fold; p < 0.05) whereas Pi treatment alone had no effect. Ca (2.7 mM) and Pi (2.5 mM) synergistically induced calcium deposition (10.8-fold; p < 0.001) in VICs. Ca treatment increased the mRNA of the osteogenic markers Msx2, Runx2, and Alpl (p < 0.01). MVs were harvested by ultracentrifugation from VICs cultured with control or calcification media (containing 2.7 mM Ca and 2.5 mM Pi) for 16 hr. Proteomics analysis revealed the marked enrichment of exosomal proteins, including CD9, CD63, LAMP-1, and LAMP-2 and a concomitant up-regulation of the Annexin family of calcium-binding proteins. Of particular note Annexin VI was shown to be enriched in calcifying VIC-derived MVs (51.9-fold; p < 0.05). Through bioinformatic analysis using Ingenuity Pathway Analysis (IPA), the up-regulation of canonical signaling pathways relevant to cardiovascular function were identified in calcifying VIC-derived MVs, including aldosterone, Rho kinase, and metal binding. Further studies using human calcified valve tissue revealed the co-localization of Annexin VI with areas of MVs in the extracellular matrix by transmission electron microscopy (TEM). Together these findings highlight a critical role for VIC-derived MVs in CAVD. Furthermore, we identify calcium as a key driver of aortic valve calcification, which may directly underpin the increased susceptibility of ESRD patients to accelerated development of CAVD.


Subject(s)
Annexin A6/metabolism , Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , Extracellular Matrix/metabolism , Extracellular Vesicles/metabolism , Hypercalcemia/etiology , Kidney Failure, Chronic/complications , Aged , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Aortic Valve/ultrastructure , Aortic Valve Stenosis/etiology , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/pathology , Calcinosis/etiology , Calcinosis/genetics , Calcinosis/pathology , Calcium/metabolism , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Extracellular Matrix/ultrastructure , Extracellular Vesicles/ultrastructure , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypercalcemia/diagnosis , Kidney Failure, Chronic/diagnosis , Male , Microscopy, Electron, Transmission , Protein Interaction Maps , Proteomics/methods , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Up-Regulation
7.
Mol Cell Proteomics ; 14(9): 2479-92, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26089344

ABSTRACT

Tyrosine kinases (TKs) are central regulators in cellular activities and perturbations of TK signaling contribute to oncogenesis. However, less than half of the TKs have been thoroughly studied and a global functional analysis of their proteomic portrait is lacking. Here we conducted a combined approach of RNA interference (RNAi) and stable isotope labeling with amino acids in cell culture (SILAC)-based quantitative proteomics to decode the TK-regulated proteome and associated signaling dynamics. As a result, a broad proteomic repertoire modulated by TKs was revealed, upon silencing of the 65 TKs expressed in MCF7 breast cancer cells. This yielded 10 new distinctive TK clusters according to similarity in TK-regulated proteome, each characterized by a unique signaling signature in contrast to previous classifications. We provide functional analyses and identify critical pathways for each cluster based on their common downstream targets. Analysis of different breast cancer subtypes showed distinct correlations of each cluster with clinical outcome. From the significantly up- and down-regulated proteins, we identified a number of markers of drug sensitivity and resistance. These data supports the role of TKs in regulating major aspects of cellular activity, but also reveals redundancy in signaling, explaining why kinase inhibitors alone often fail to achieve their clinical aims. The TK-SILACepedia provides a comprehensive resource for studying the global function of TKs in cancer.


Subject(s)
Breast Neoplasms/metabolism , Isotope Labeling/methods , Protein-Tyrosine Kinases/metabolism , Proteome/analysis , Proteomics/methods , RNA Interference , Amino Acids/chemistry , Cell Culture Techniques , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Signal Transduction
8.
Nature ; 515(7526): 279-282, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25119035

ABSTRACT

The mechanisms by which physical forces regulate endothelial cells to determine the complexities of vascular structure and function are enigmatic. Studies of sensory neurons have suggested Piezo proteins as subunits of Ca(2+)-permeable non-selective cationic channels for detection of noxious mechanical impact. Here we show Piezo1 (Fam38a) channels as sensors of frictional force (shear stress) and determinants of vascular structure in both development and adult physiology. Global or endothelial-specific disruption of mouse Piezo1 profoundly disturbed the developing vasculature and was embryonic lethal within days of the heart beating. Haploinsufficiency was not lethal but endothelial abnormality was detected in mature vessels. The importance of Piezo1 channels as sensors of blood flow was shown by Piezo1 dependence of shear-stress-evoked ionic current and calcium influx in endothelial cells and the ability of exogenous Piezo1 to confer sensitivity to shear stress on otherwise resistant cells. Downstream of this calcium influx there was protease activation and spatial reorganization of endothelial cells to the polarity of the applied force. The data suggest that Piezo1 channels function as pivotal integrators in vascular biology.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/physiology , Friction , Ion Channels/metabolism , Stress, Mechanical , Animals , Embryo, Mammalian/blood supply , Embryo, Mammalian/metabolism , Female , Hemorheology , Male , Mice
9.
J Proteome Res ; 12(4): 1580-90, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23413775

ABSTRACT

Zebrafish is an important model organism in biological research. One of the least explored tissues of zebrafish is blood, because the existing methods for isolating blood from this organism are tedious and irreproducible. The small volume of blood collected by these methods also prohibits many biochemical and cytological analyses. This technical obstacle limits the utilization of zebrafish in many applications, particularly in hematological research and plasma biomarker discovery. To overcome this limitation, we have established a novel method of extracting blood from zebrafish, based on the use of low centrifugal force to collect blood from a wound. This method consistently recovers more blood than traditional methods. Gel electrophoresis and flow cytometry showed that composition of blood harvested by this method is indistinguishable from traditional methods. The increase in yield enables us to perform biochemical experiments on zebrafish blood. In particular, we have demonstrated that quantitative proteomics can be performed on plasma collected from single zebrafish. Here, we have compared, by using shotgun proteomic analysis, the plasma proteomes of adult male and female zebrafish. Twenty-seven gender-dependent plasma proteins are identified and their biochemical importance discussed. Taken together, this novel technique enables analyses that were previously difficult to perform on zebrafish blood.


Subject(s)
Blood Proteins/analysis , Blood Specimen Collection/methods , Zebrafish Proteins/blood , Zebrafish/blood , Animals , Female , Male , Proteomics/methods , Reproducibility of Results , Sex Factors , Tandem Mass Spectrometry
10.
Nanomedicine ; 9(5): 583-93, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23117048

ABSTRACT

The protein corona of a nanomaterial is a complex layer of proteins spontaneously and stably formed when the material is exposed to body fluids or intracellular environments. In this study, we utilised stable isotope labeling by amino acids in cell culture (SILAC)-based quantitative proteomics to characterise the binding of human cellular proteins to two forms of carbon nanoparticles: namely multi-walled carbon nanotubes (MWCNTs) and carbon black (CB). The relative binding efficiency of over 750 proteins to these materials is measured. The data indicate that MWCNTs and CB bind to vastly different sets of proteins. The molecular basis of selectivity in protein binding is investigated. This study is the first large-scale characterisation of protein corona on CNT, providing the biochemical basis for the assessment of the suitability of CNTs as biomedical tools, and as an emerging pollutant. FROM THE CLINICAL EDITOR: This team of investigators performed the first large-scale characterization of protein corona on carbon nanotubes, studying 750 proteins and assessing the suitability of CNTs as biomedical tools and as an emerging pollutant.


Subject(s)
Amino Acids/chemistry , Carbon/chemistry , Nanotubes, Carbon/chemistry , Proteins/chemistry , Cell Line , Humans , Isotope Labeling , Nanoparticles/chemistry , Protein Binding , Proteomics , Soot/chemistry
11.
PLoS One ; 7(10): e47497, 2012.
Article in English | MEDLINE | ID: mdl-23144703

ABSTRACT

Originally the novel protein Blom7α was identified as novel pre-mRNA splicing factor that interacts with SNEV(Prp19/Pso4), an essential protein involved in extension of human endothelial cell life span, DNA damage repair, the ubiquitin-proteasome system, and pre-mRNA splicing. Blom7α belongs to the heteronuclear ribonucleoprotein K homology (KH) protein family, displaying 2 KH domains, a well conserved and widespread RNA-binding motif. In order to identify specific sequence binding motifs, we here used Systematic Evolution of Ligands by Exponential Enrichment (SELEX) with a synthetic RNA library. Besides sequence motifs like (U/A)(1-4) C(2-6) (U/A)(1-5), we identified an AC-rich RNA-aptamer that we termed AK48 (Aptamer KH-binding 48), binding to Blom7α with high affinity. Addition of AK48 to pre-mRNA splicing reactions in vitro inhibited the formation of mature spliced mRNA and led to a slight accumulation of the H complex of the spliceosome. These results suggest that the RNA binding activity of Blom7α might be required for pre-mRNA splicing catalysis. The inhibition of in-vitro splicing by the small RNA AK48 indicates the potential use of small RNA molecules in targeting the spliceosome complex as a novel target for drug development.


Subject(s)
Aptamers, Nucleotide/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , RNA Precursors/metabolism , RNA Splicing , AT Rich Sequence/genetics , Amino Acid Sequence , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/pharmacology , Base Sequence , Binding Sites/genetics , Electrophoretic Mobility Shift Assay , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Molecular Sequence Data , Mutation , Protein Binding , RNA Precursors/genetics , SELEX Aptamer Technique/methods , Sequence Homology, Amino Acid , Spliceosomes/drug effects , Spliceosomes/genetics , Spliceosomes/metabolism
12.
Proteomics ; 12(9): 1431-6, 2012 May.
Article in English | MEDLINE | ID: mdl-22585751

ABSTRACT

Influenza A virus is one of the world's major uncontrolled pathogens, causing seasonal epidemics as well as global pandemics. This was evidenced by the recent emergence and now prevalence of the 2009 swine origin pandemic H1N1 influenza A virus. In this study, quantitative proteomics using stable isotope labelling with amino acids in cell culture was used to investigate the changes in the host cell proteome in cells infected with pandemic H1N1 influenza A virus. The study was conducted in A549 cells that retain properties similar to alveolar cells. Several global pathways were affected, including cell cycle regulation and lipid metabolism, and these could be correlated with recent microarray analyses of cells infected with influenza A virus. Taken together, both quantitative proteomics and transcriptomic approaches can be used to identify potential cellular proteins whose functions in the virus life cycle could be targeted for chemotherapeutic intervention.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human/metabolism , Lung/metabolism , Lung/virology , Proteome/analysis , Blotting, Western , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/virology , Host-Pathogen Interactions , Humans , Influenza, Human/pathology , Influenza, Human/virology , Isotope Labeling , Lung/pathology , Proteome/chemistry , Proteomics/methods , Reproducibility of Results
13.
Biochem J ; 438(1): 81-91, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21639856

ABSTRACT

The Cdc5L (cell division cycle 5-like) complex is a spliceosomal subcomplex that also plays a role in DNA repair. The complex contains the splicing factor hPrp19, also known as SNEV or hPso4, which is involved in cellular life-span regulation and proteasomal breakdown. In a recent large-scale proteomics analysis for proteins associated with this complex, proteins involved in transcription, cell-cycle regulation, DNA repair, the ubiquitin-proteasome system, chromatin remodelling, cellular aging, the cytoskeleton and trafficking, including four members of the exocyst complex, were identified. In the present paper we report that Exo70 interacts directly with SNEV(hPrp19/hPso4) and shuttles to the nucleus, where it associates with the spliceosome. We mapped the interaction site to the N-terminal 100 amino acids of Exo70, which interfere with pre-mRNA splicing in vitro. Furthermore, Exo70 influences the splicing of a model substrate as well as of its own pre-mRNA in vivo. In addition, we found that Exo70 is alternatively spliced in a cell-type- and cell-age- dependent way. These results suggest a novel and unexpected role of Exo70 in nuclear mRNA splicing, where it might signal membrane events to the splicing apparatus.


Subject(s)
Alternative Splicing , Cell Nucleus/genetics , DNA Repair Enzymes/metabolism , Nuclear Proteins/metabolism , RNA Precursors/genetics , Spliceosomes/genetics , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Blotting, Western , Cell Nucleus/metabolism , DNA Repair Enzymes/genetics , Fluorescent Antibody Technique , HeLa Cells , Humans , Molecular Sequence Data , Nuclear Proteins/genetics , Protein Binding , RNA Splicing Factors , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Two-Hybrid System Techniques
14.
Cell Commun Signal ; 8: 28, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20920157

ABSTRACT

Prion diseases or transmissible spongiform encephalopathies (TSEs) are fatal diseases associated with the conversion of the cellular prion protein (PrPC) to the abnormal prion protein (PrPSc). Since the molecular mechanisms in pathogenesis are widely unclear, we analyzed the global phospho-proteome and detected a differential pattern of tyrosine- and threonine phosphorylated proteins in PrPSc-replicating and pentosan polysulfate (PPS)-rescued N2a cells in two-dimensional gel electrophoresis. To quantify phosphorylated proteins, we performed a SILAC (stable isotope labeling by amino acids in cell culture) analysis and identified 105 proteins, which showed a regulated phosphorylation upon PrPSc infection. Among those proteins, we validated the dephosphorylation of stathmin and Cdc2 and the induced phosphorylation of cofilin in PrPSc-infected N2a cells in Western blot analyses. Our analysis showed for the first time a differentially regulated phospho-proteome in PrPSc infection, which could contribute to the establishment of novel protein markers and to the development of novel therapeutic intervention strategies in targeting prion-associated disease.

15.
Mol Cell Proteomics ; 9(9): 1920-36, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20467043

ABSTRACT

Virus-host interactions involve complex interplay between viral and host factors, rendering them an ideal target for proteomic analysis. Here we detail a high throughput quantitative proteomics analysis of Vero cells infected with the coronavirus infectious bronchitis virus (IBV), a positive strand RNA virus that replicates in the cytoplasm. Stable isotope labeling with amino acids in cell culture (SILAC) was used in conjunction with LC-MS/MS to identify and quantify 1830 cellular and two viral proteins from IBV-infected cells. Fractionation of cells into cytoplasmic, nuclear, and nucleolar extracts was used to reduce sample complexity and provide information on the trafficking of proteins between the different compartments. Each fraction showed a proportion of proteins exhibiting >or=2-fold changes in abundance. Ingenuity Pathway Analysis revealed that proteins that changed in response to infection could be grouped into different functional categories. These included proteins regulated by NF-kappaB- and AP-1-dependent pathways and proteins involved in the cytoskeleton and molecular motors. A luciferase-based reporter gene assay was used to validate the up-regulation of AP-1- and NF-kappaB-dependent transcription in IBV-infected cells and confirmed using immunofluorescence. Immunofluorescence was used to validate changes in the subcellular localization of vimentin and myosin VI in IBV-infected cells. The proteomics analysis also confirmed the presence of the viral nucleocapsid protein as localizing in the cytoplasm, nucleus, and nucleolus and the viral membrane protein in the cytoplasmic fraction. This research is the first application of SILAC to study total host cell proteome changes in response to positive sense RNA virus infection and illustrates the versatility of this technique as applied to infectious disease research.


Subject(s)
Cell Nucleolus/chemistry , Cell Nucleus/chemistry , Cytoplasm/chemistry , Infectious bronchitis virus/pathogenicity , Isotope Labeling/methods , Proteomics , Viral Proteins/chemistry , Amino Acid Sequence , Animals , Chlorocebus aethiops , Chromatography, Liquid , Electrophoresis, Polyacrylamide Gel , Microscopy, Confocal , Molecular Sequence Data , Tandem Mass Spectrometry/methods , Vero Cells
16.
EMBO Rep ; 11(6): 445-51, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20467437

ABSTRACT

Heterogeneous nuclear ribonucleoprotein-M (hnRNP-M) is an abundant nuclear protein that binds to pre-mRNA and is a component of the spliceosome complex. A direct interaction was detected in vivo between hnRNP-M and the human spliceosome proteins cell division cycle 5-like (CDC5L) and pleiotropic regulator 1 (PLRG1) that was inhibited during the heat-shock stress response. A central region in hnRNP-M is required for interaction with CDC5L/PLRG1. hnRNP-M affects both 5' and 3' alternative splice site choices, and an hnRNP-M mutant lacking the CDC5L/PLRG1 interaction domain is unable to modulate alternative splicing of an adeno-E1A mini-gene substrate.


Subject(s)
Cell Cycle Proteins/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group M/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , RNA Splice Sites , RNA-Binding Proteins/metabolism , Binding Sites , Cell Cycle Proteins/chemistry , Green Fluorescent Proteins , HeLa Cells , Heat-Shock Response , Heterogeneous-Nuclear Ribonucleoprotein Group M/chemistry , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Nuclear Proteins/chemistry , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , RNA-Binding Proteins/chemistry , Recombinant Fusion Proteins
17.
J Biol Chem ; 284(42): 29193-204, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19641227

ABSTRACT

The removal of introns from pre-mRNA is performed by the spliceosome that stepwise assembles on the pre-mRNA before performing two catalytic steps. The spliceosome-associated CDC5L-SNEV(Prp19-Pso4) complex is implicated in activation of the second catalytic step of pre-mRNA splicing, and one of its members, SNEV(Prp19-Pso4), is also implicated in spliceosome assembly. To identify interaction partners of SNEVPrp19-Pso4, we have performed yeast two-hybrid screenings. Among the putative binding partners was a so far uncharacterized protein carrying two heterogeneous nuclear ribonucleoprotein K homology domains that we termed Blom7alpha. Blom7alpha is expressed in all tissues tested, and at least three splice variants exist. After confirming direct and physical interaction of SNEV and Blom7alpha, we investigated if it plays a functional role during pre-mRNA splicing. Indeed, Blom7alpha co-localizes and co-precipitates with splicing factors and pre-mRNA and is present in affinity-purified spliceosomes. More importantly, addition of Blom7alpha to HeLa nuclear extracts increased splicing activity in a dose-dependent manner. Furthermore, we tested if Blom7alpha influences splice site selection using two different minigene constructs. Indeed, both 5'- as well as 3'-site selection was altered upon Blom7alpha overexpression. Thus we suggest that Blom7alpha is a novel splicing factor of the K homology domain family that might be implicated in alternative splicing by helping to position the CDC5L-SNEV(Prp19-Pso4) complex at the splice sites.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein K/chemistry , Heterogeneous-Nuclear Ribonucleoproteins/chemistry , Alternative Splicing , Animals , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , Dose-Response Relationship, Drug , Escherichia coli/genetics , HeLa Cells , Humans , Introns , Protein Binding , Protein Structure, Tertiary , RNA Precursors/metabolism , Saccharomyces cerevisiae/genetics , Two-Hybrid System Techniques
18.
Cancer Res ; 68(18): 7621-8, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794151

ABSTRACT

Cyclin D1 expression represents one of the key mitogen-regulated events during the G(1) phase of the cell cycle, whereas Cyclin D1 overexpression is frequently associated with human malignancy. Here, we describe a novel mechanism regulating Cyclin D1 levels. We find that SNIP1, previously identified as a regulator of Cyclin D1 expression, does not, as previously thought, primarily function as a transcriptional coactivator for this gene. Rather, SNIP1 plays a critical role in cotranscriptional or posttranscriptional Cyclin D1 mRNA stability. Moreover, we show that the majority of nucleoplasmic SNIP1 is present within a previously undescribed complex containing SkIP, THRAP3, BCLAF1, and Pinin, all proteins with reported roles in RNA processing and transcriptional regulation. We find that this complex, which we have termed the SNIP1/SkIP-associated RNA-processing complex, is coordinately recruited to both the 3' end of the Cyclin D1 gene and Cyclin D1 RNA. Significantly, SNIP1 is required for the further recruitment of the RNA processing factor U2AF65 to both the Cyclin D1 gene and RNA. This study shows a novel mechanism regulating Cyclin D1 expression and offers new insight into the role of SNIP1 and associated proteins as regulators of proliferation and cancer.


Subject(s)
Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , RNA, Neoplasm/genetics , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Cycle/physiology , Cell Line, Tumor , Cyclin D1/biosynthesis , HeLa Cells , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Osteosarcoma/genetics , Osteosarcoma/metabolism , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA-Binding Proteins , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Splicing Factor U2AF , Transcription, Genetic , Transfection
19.
Nucleic Acids Res ; 33(21): 6868-83, 2005.
Article in English | MEDLINE | ID: mdl-16332694

ABSTRACT

We have isolated the human protein SNEV as downregulated in replicatively senescent cells. Sequence homology to the yeast splicing factor Prp19 suggested that SNEV might be the orthologue of Prp19 and therefore might also be involved in pre-mRNA splicing. We have used various approaches including gene complementation studies in yeast using a temperature sensitive mutant with a pleiotropic phenotype and SNEV immunodepletion from human HeLa nuclear extracts to determine its function. A human-yeast chimera was indeed capable of restoring the wild-type phenotype of the yeast mutant strain. In addition, immunodepletion of SNEV from human nuclear extracts resulted in a decrease of in vitro pre-mRNA splicing efficiency. Furthermore, as part of our analysis of protein-protein interactions within the CDC5L complex, we found that SNEV interacts with itself. The self-interaction domain was mapped to amino acids 56-74 in the protein's sequence and synthetic peptides derived from this region inhibit in vitro splicing by surprisingly interfering with spliceosome formation and stability. These results indicate that SNEV is the human orthologue of yeast PRP19, functions in splicing and that homo-oligomerization of SNEV in HeLa nuclear extract is essential for spliceosome assembly and that it might also be important for spliceosome stability.


Subject(s)
RNA Splicing , Spliceosomes/metabolism , Ubiquitin-Protein Ligases/physiology , Amino Acid Sequence , Cell Nucleus/chemistry , Conserved Sequence , DNA Repair Enzymes , Evolution, Molecular , HeLa Cells , Humans , Molecular Sequence Data , Mutation , Nuclear Proteins , Peptides/pharmacology , Phenotype , Protein Structure, Tertiary , RNA Precursors/metabolism , RNA Splicing/drug effects , RNA Splicing Factors , RNA, Messenger/metabolism , RNA-Binding Proteins/analysis , Recombinant Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid , Spliceosomes/drug effects , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics
20.
RNA ; 11(8): 1201-14, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16043505

ABSTRACT

We have analyzed the interaction between the U2AF subunits U2AF35 and U2AF65 in vivo using fluorescence resonance energy transfer (FRET) microscopy. U2 snRNP Auxiliary Factor (U2AF) is an essential pre-mRNA splicing factor complex, comprising 35-kDa (U2AF35) and 65-kDa (U2AF65) subunits. U2AF65 interacts directly with the polypyrimidine tract and promotes binding of U2 snRNP to the pre-mRNA branchpoint, while U2AF35 associates with the conserved AG dinucleotide at the 3' end of the intron and has multiple functions in the splicing process. Using two different approaches for measuring FRET, we have identified and spatially localized sites of direct interaction between U2AF35 and U2AF65 in vivo in live cell nuclei. While U2AF is thought to function as a heterodimeric complex, the FRET data have also revealed a novel U2AF35 self-interaction in vivo, which is confirmed in vitro using biochemical assays. These results suggest that the stoichiometry of the U2AF complex may, at least in part, differ in vivo from the expected heterodimeric complex. The data show that FRET studies offer a valuable approach for probing interactions between pre-mRNA splicing factors in vivo.


Subject(s)
Nuclear Proteins/metabolism , Ribonucleoproteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Cell Nucleus/metabolism , DNA Primers , Flow Cytometry , Fluorescence Resonance Energy Transfer/methods , HeLa Cells , Humans , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Polymerase Chain Reaction , Protein Binding , RNA Splicing , RNA, Messenger/genetics , Recombinant Fusion Proteins/metabolism , Splicing Factor U2AF , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...