Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Cell ; 161(3): 647-660, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25910212

ABSTRACT

How disease-associated mutations impair protein activities in the context of biological networks remains mostly undetermined. Although a few renowned alleles are well characterized, functional information is missing for over 100,000 disease-associated variants. Here we functionally profile several thousand missense mutations across a spectrum of Mendelian disorders using various interaction assays. The majority of disease-associated alleles exhibit wild-type chaperone binding profiles, suggesting they preserve protein folding or stability. While common variants from healthy individuals rarely affect interactions, two-thirds of disease-associated alleles perturb protein-protein interactions, with half corresponding to "edgetic" alleles affecting only a subset of interactions while leaving most other interactions unperturbed. With transcription factors, many alleles that leave protein-protein interactions intact affect DNA binding. Different mutations in the same gene leading to different interaction profiles often result in distinct disease phenotypes. Thus disease-associated alleles that perturb distinct protein activities rather than grossly affecting folding and stability are relatively widespread.


Subject(s)
Disease/genetics , Mutation, Missense , Protein Interaction Maps , Proteins/genetics , Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genome-Wide Association Study , Humans , Open Reading Frames , Protein Folding , Protein Stability
2.
Nature ; 580(7803): 402-408, 2020 04.
Article in English | MEDLINE | ID: mdl-32296183

ABSTRACT

Global insights into cellular organization and genome function require comprehensive understanding of the interactome networks that mediate genotype-phenotype relationships1,2. Here we present a human 'all-by-all' reference interactome map of human binary protein interactions, or 'HuRI'. With approximately 53,000 protein-protein interactions, HuRI has approximately four times as many such interactions as there are high-quality curated interactions from small-scale studies. The integration of HuRI with genome3, transcriptome4 and proteome5 data enables cellular function to be studied within most physiological or pathological cellular contexts. We demonstrate the utility of HuRI in identifying the specific subcellular roles of protein-protein interactions. Inferred tissue-specific networks reveal general principles for the formation of cellular context-specific functions and elucidate potential molecular mechanisms that might underlie tissue-specific phenotypes of Mendelian diseases. HuRI is a systematic proteome-wide reference that links genomic variation to phenotypic outcomes.


Subject(s)
Proteome/metabolism , Extracellular Space/metabolism , Humans , Organ Specificity , Protein Interaction Mapping
3.
Mol Cell Proteomics ; 22(1): 100451, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36423812

ABSTRACT

Dimerization of SRC kinase adaptor phosphoprotein 2 (SKAP2) induces an increase of binding for most SRC kinases suggesting a fine-tuning with transphosphorylation for kinase activation. This work addresses the molecular basis of SKAP2-mediated SRC kinase regulation through the lens of their interaction capacities. By combining a luciferase complementation assay and extensive site-directed mutagenesis, we demonstrated that SKAP2 interacts with SRC kinases through a modular organization depending both on their phosphorylation-dependent activation and subcellular localization. SKAP2 contains three interacting modules consisting in the dimerization domain, the SRC homology 3 (SH3) domain, and the second interdomain located between the Pleckstrin homology and the SH3 domains. Functionally, the dimerization domain is necessary and sufficient to bind to most activated and myristyl SRC kinases. In contrast, the three modules are necessary to bind SRC kinases at their steady state. The Pleckstrin homology and SH3 domains of SKAP2 as well as tyrosines located in the interdomains modulate these interactions. Analysis of mutants of the SRC kinase family member hematopoietic cell kinase supports this model and shows the role of two residues, Y390 and K7, on its degradation following activation. In this article, we show that a modular architecture of SKAP2 drives its interaction with SRC kinases, with the binding capacity of each module depending on both their localization and phosphorylation state activation. This work opens new perspectives on the molecular mechanisms of SRC kinases activation, which could have significant therapeutic impact.


Subject(s)
src Homology Domains , src-Family Kinases , src-Family Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation
4.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33402530

ABSTRACT

The recent emergence and reemergence of viruses in the human population has highlighted the need to develop broader panels of therapeutic molecules. High-throughput screening assays opening access to untargeted steps of the viral replication cycle will provide powerful leverage to identify innovative antiviral molecules. We report here the development of an innovative protein complementation assay, termed αCentauri, to measure viral translocation between subcellular compartments. As a proof of concept, the Centauri fragment was either tethered to the nuclear pore complex or sequestered in the nucleus, while the complementary α fragment (<16 amino acids) was attached to the integrase proteins of infectious HIV-1. The translocation of viral ribonucleoproteins from the cytoplasm to the nuclear envelope or to the nucleoplasm efficiently reconstituted superfolder green fluorescent protein or NanoLuc αCentauri reporters. These fluorescence- or bioluminescence-based assays offer a robust readout of specific steps of viral infection in a multiwell format that is compatible for high-throughput screening and is validated by a short hairpin RNA-based prototype screen.


Subject(s)
High-Throughput Screening Assays/methods , Virus Diseases/metabolism , Virus Replication/physiology , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Green Fluorescent Proteins/metabolism , HIV Infections/metabolism , HeLa Cells , Humans , Nuclear Envelope/metabolism , Nuclear Pore/metabolism , Ribonucleoproteins/metabolism , Virus Replication/drug effects
5.
J Biol Chem ; 298(6): 101956, 2022 06.
Article in English | MEDLINE | ID: mdl-35452674

ABSTRACT

The signaling pathways and cellular functions regulated by the four Numb-associated kinases are largely unknown. We reported that AAK1 and GAK control intracellular trafficking of RNA viruses and revealed a requirement for BIKE in early and late stages of dengue virus (DENV) infection. However, the downstream targets phosphorylated by BIKE have not yet been identified. Here, to identify BIKE substrates, we conducted a barcode fusion genetics-yeast two-hybrid screen and retrieved publicly available data generated via affinity-purification mass spectrometry. We subsequently validated 19 of 47 putative BIKE interactors using mammalian cell-based protein-protein interaction assays. We found that CLINT1, a cargo-specific adapter implicated in bidirectional Golgi-to-endosome trafficking, emerged as a predominant hit in both screens. Our experiments indicated that BIKE catalyzes phosphorylation of a threonine 294 CLINT1 residue both in vitro and in cell culture. Our findings revealed that CLINT1 phosphorylation mediates its binding to the DENV nonstructural 3 protein and subsequently promotes DENV assembly and egress. Additionally, using live-cell imaging we revealed that CLINT1 cotraffics with DENV particles and is involved in mediating BIKE's role in DENV infection. Finally, our data suggest that additional cellular BIKE interactors implicated in the host immune and stress responses and the ubiquitin proteasome system might also be candidate phosphorylation substrates of BIKE. In conclusion, these findings reveal cellular substrates and pathways regulated by the understudied Numb-associated kinase enzyme BIKE, a mechanism for CLINT1 regulation, and control of DENV infection via BIKE signaling, with potential implications for cell biology, virology, and host-targeted antiviral design.


Subject(s)
Dengue Virus , Dengue , Animals , Dengue/metabolism , Dengue Virus/metabolism , Humans , Phosphorylation , Two-Hybrid System Techniques , Virus Replication
6.
Mol Cell Proteomics ; 20: 100049, 2021.
Article in English | MEDLINE | ID: mdl-33515806

ABSTRACT

Viruses manipulate the central machineries of host cells to their advantage. They prevent host cell antiviral responses to create a favorable environment for their survival and propagation. Measles virus (MV) encodes two nonstructural proteins MV-V and MV-C known to counteract the host interferon response and to regulate cell death pathways. Several molecular mechanisms underlining MV-V regulation of innate immunity and cell death pathways have been proposed, whereas MV-C host-interacting proteins are less studied. We suggest that some cellular factors that are controlled by MV-C protein during viral replication could be components of innate immunity and the cell death pathways. To determine which host factors are targeted by MV-C, we captured both direct and indirect host-interacting proteins of MV-C protein. For this, we used a strategy based on recombinant viruses expressing tagged viral proteins followed by affinity purification and a bottom-up mass spectrometry analysis. From the list of host proteins specifically interacting with MV-C protein in different cell lines, we selected the host targets that belong to immunity and cell death pathways for further validation. Direct protein interaction partners of MV-C were determined by applying protein complementation assay and the bioluminescence resonance energy transfer approach. As a result, we found that MV-C protein specifically interacts with p65-iASPP protein complex that controls both cell death and innate immunity pathways and evaluated the significance of these host factors on virus replication.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factor RelA/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Cell Death , Cell Line , Chlorocebus aethiops , Host-Pathogen Interactions , Humans , Intracellular Signaling Peptides and Proteins/genetics , Measles virus/genetics , Measles virus/physiology , Protein Interaction Maps , Proteomics , Repressor Proteins/genetics , Transcription Factor RelA/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Viral Nonstructural Proteins/genetics , Virus Replication
7.
Int J Mol Sci ; 24(19)2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37834238

ABSTRACT

Infection with Ebola virus (EBOV) is responsible for hemorrhagic fever in humans with a high mortality rate. Combined efforts of prevention and therapeutic intervention are required to tackle highly variable RNA viruses, whose infections often lead to outbreaks. Here, we have screened the 2P2I3D chemical library using a nanoluciferase-based protein complementation assay (NPCA) and isolated two compounds that disrupt the interaction of the EBOV protein fragment VP35IID with the N-terminus of the dsRNA-binding proteins PKR and PACT, involved in IFN response and/or intrinsic immunity, respectively. The two compounds inhibited EBOV infection in cell culture as well as infection by measles virus (MV) independently of IFN induction. Consequently, we propose that the compounds are antiviral by restoring intrinsic immunity driven by PACT. Given that PACT is highly conserved across mammals, our data support further testing of the compounds in other species, as well as against other negative-sense RNA viruses.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Humans , Animals , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/metabolism , Ebolavirus/physiology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Mammals
8.
Nucleic Acids Res ; 48(18): 10428-10440, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32960265

ABSTRACT

Cellular exonucleases involved in the processes that regulate RNA stability and quality control have been shown to restrict or to promote the multiplication cycle of numerous RNA viruses. Influenza A viruses are major human pathogens that are responsible for seasonal epidemics, but the interplay between viral proteins and cellular exonucleases has never been specifically studied. Here, using a stringent interactomics screening strategy and an siRNA-silencing approach, we identified eight cellular factors among a set of 75 cellular proteins carrying exo(ribo)nuclease activities or involved in RNA decay processes that support influenza A virus multiplication. We show that the exoribonuclease ERI1 interacts with the PB2, PB1 and NP components of the viral ribonucleoproteins and is required for viral mRNA transcription. More specifically, we demonstrate that the protein-protein interaction is RNA dependent and that both the RNA binding and exonuclease activities of ERI1 are required to promote influenza A virus transcription. Finally, we provide evidence that during infection, the SLBP protein and histone mRNAs co-purify with vRNPs alongside ERI1, indicating that ERI1 is most probably recruited when it is present in the histone pre-mRNA processing complex in the nucleus.


Subject(s)
Exoribonucleases/genetics , Influenza A virus/genetics , Influenza, Human/genetics , Nuclear Proteins/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , Cell Line , Histones/genetics , Host-Pathogen Interactions , Humans , Influenza A virus/pathogenicity , Influenza, Human/virology , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Small Interfering , RNA, Viral/genetics , Ribonucleoproteins/genetics , Transcription, Genetic/genetics , Viral Proteins/genetics , Virus Replication/genetics
9.
Proc Natl Acad Sci U S A ; 116(22): 10968-10977, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31076555

ABSTRACT

New therapeutic strategies targeting influenza are actively sought due to limitations in current drugs available. Host-directed therapy is an emerging concept to target host functions involved in pathogen life cycles and/or pathogenesis, rather than pathogen components themselves. From this perspective, we focused on an essential host partner of influenza viruses, the RED-SMU1 splicing complex. Here, we identified two synthetic molecules targeting an α-helix/groove interface essential for RED-SMU1 complex assembly. We solved the structure of the SMU1 N-terminal domain in complex with RED or bound to one of the molecules identified to disrupt this complex. We show that these compounds inhibiting RED-SMU1 interaction also decrease endogenous RED-SMU1 levels and inhibit viral mRNA splicing and viral multiplication, while preserving cell viability. Overall, our data demonstrate the potential of RED-SMU1 destabilizing molecules as an antiviral therapy that could be active against a wide range of influenza viruses and be less prone to drug resistance.


Subject(s)
Antiviral Agents/pharmacology , Chromosomal Proteins, Non-Histone/metabolism , Cytokines/metabolism , Orthomyxoviridae/drug effects , RNA Splicing Factors/metabolism , A549 Cells , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Cytokines/chemistry , Cytokines/genetics , HEK293 Cells , Host-Pathogen Interactions/drug effects , Humans , Molecular Docking Simulation , Orthomyxoviridae/pathogenicity , Protein Binding/drug effects , Protein Stability/drug effects , RNA Splicing , RNA Splicing Factors/chemistry , RNA Splicing Factors/genetics , Spliceosomes/drug effects
10.
Genes Dev ; 28(17): 1957-75, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25184681

ABSTRACT

BRCA1 is a breast and ovarian tumor suppressor. Given its numerous incompletely understood functions and the possibility that more exist, we performed complementary systematic screens in search of new BRCA1 protein-interacting partners. New BRCA1 functions and/or a better understanding of existing ones were sought. Among the new interacting proteins identified, genetic interactions were detected between BRCA1 and four of the interactors: TONSL, SETX, TCEANC, and TCEA2. Genetic interactions were also detected between BRCA1 and certain interactors of TONSL, including both members of the FACT complex. From these results, a new BRCA1 function in the response to transcription-associated DNA damage was detected. Specifically, new roles for BRCA1 in the restart of transcription after UV damage and in preventing or repairing damage caused by stabilized R loops were identified. These roles are likely carried out together with some of the newly identified interactors. This new function may be important in BRCA1 tumor suppression, since the expression of several interactors, including some of the above-noted transcription proteins, is repeatedly aberrant in both breast and ovarian cancers.


Subject(s)
BRCA1 Protein/metabolism , DNA Damage/genetics , DNA Repair/genetics , Transcription, Genetic/genetics , BRCA1 Protein/genetics , Cell Line, Tumor , HeLa Cells , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Binding , Protein Interaction Mapping , Ultraviolet Rays
11.
Chemistry ; 27(6): 2112-2123, 2021 Jan 26.
Article in English | MEDLINE | ID: mdl-33137225

ABSTRACT

In this work on the design and studies of luciferins related to the blue-hued coelenterazine, the synthesis of heterocyclic analogues susceptible to produce a photon, possibly at a different wavelength, is undertaken. Here, the synthesis of O-acetylated derivatives of imidazo[1,2-b]pyridazin-3(5 H)-one, imidazo[2,1-f][1,2,4]triazin-7(1 H)-one, imidazo[1,2-a]pyridin-3-ol, imidazo[1,2-a]quinoxalin-1(5 H)-one, benzo[f]imidazo[1,2-a]quinoxalin-3(11 H)-one, imidazo[1',2':1,6]pyrazino[2,3-c]quinolin-3(11 H)-one, and 5,11-dihydro-3 H-chromeno[4,3-e]imidazo[1,2-a]pyrazin-3-one is described thanks to extensive use of the Buchwald-Hartwig N-arylation reaction. The acidic hydrolysis of these derivatives then gave solutions of the corresponding luciferin analogues, which were studied. Not too unexpectedly, even if these were "dressed" with substituents found in actual substrates of the nanoKAZ/NanoLuc luciferase, no bioluminescence was observed with these compounds. However, in a phosphate buffer, all produced a light signal, by chemiluminescence, with extensive variations in their respective intensity and this could be increased by adding a quaternary ammonium salt in the buffer. This aspect was actually instrumental to determine the emission spectra of many of these luciferin analogues.

12.
Chemistry ; 26(4): 948-958, 2020 Jan 16.
Article in English | MEDLINE | ID: mdl-31765054

ABSTRACT

We describe here an extensive structure-bioluminescence relationship study of a chemical library of analogues of coelenterazine, using nanoKAZ/NanoLuc, a mutated luciferase originated from the catalytic subunit of the deep-sea shrimp Oplophorus gracilirostris. Out of the 135 O-acetylated precursors that were prepared by using our recently reported synthesis and following their hydrolysis to give solutions of the corresponding luciferins, notable bioluminescence improvements were achieved in comparison with furimazine, which is currently amongst the best substrates of nanoKAZ/NanoLuc. For instance, the rather more lipophilic analogue 8-(2,3-difluorobenzyl)-2-((5-methylfuran-2-yl)methyl)-6-phenylimidazo[1,2-a]pyrazin-3(7H)-one provided a 1.5-fold improvement of the total light output over a 2 h period, a close to threefold increase of the initial signal intensity and a signal-to-background ratio five times greater than furimazine. The kinetic parameters for the enzymatic reaction were obtained for a selection of luciferin analogues and provided unexpected insights into the luciferase activity. Most prominently, along with a general substrate-dependent and irreversible inactivation of this enzyme, in the case of the optimized luciferin mentioned above, the consumption of 2664 molecules was found to be required for the detection of a single Relative Light Unit (RLU; a luminometer-dependent fraction of a photon).

13.
Org Biomol Chem ; 17(15): 3709-3713, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30882838

ABSTRACT

An original gram-scale synthesis of O-acetylated forms of coelenterazine, furimazine or hydroxy-bearing analogues of luciferins is described. The comparison over two hours of their bioluminescence, using the nanoKAZ/NanoLuc luciferase, provides remarkable insights useful for the selection of a substrate adapted for a given application.


Subject(s)
Firefly Luciferin/chemical synthesis , Imidazoles/chemical synthesis , Pyrazines/chemical synthesis , Acetylation , Animals , Fireflies , Firefly Luciferin/analogs & derivatives , Firefly Luciferin/chemistry , Imidazoles/chemistry , Luciferases, Firefly/metabolism , Luminescent Measurements , Molecular Structure , Pyrazines/chemistry
14.
Nat Methods ; 12(8): 787-93, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26053890

ABSTRACT

Many protein interactions are mediated by small linear motifs interacting specifically with defined families of globular domains. Quantifying the specificity of a motif requires measuring and comparing its binding affinities to all its putative target domains. To this end, we developed the high-throughput holdup assay, a chromatographic approach that can measure up to 1,000 domain-motif equilibrium binding affinities per day. After benchmarking the approach on 210 PDZ-peptide pairs with known affinities, we determined the affinities of two viral PDZ-binding motifs derived from human papillomavirus E6 oncoproteins for 209 PDZ domains covering 79% of the human 'PDZome'. We obtained sharply sequence-dependent binding profiles that quantitatively describe the PDZome recognition specificity of each motif. This approach, applicable to many categories of domain-ligand interactions, has wide potential for quantifying the specificities of interactomes.


Subject(s)
High-Throughput Screening Assays , PDZ Domains , Protein Interaction Mapping/methods , Proteins/chemistry , Amino Acid Motifs , Chromatography , DNA-Binding Proteins/chemistry , Humans , Kinetics , Ligands , Oncogene Proteins, Viral/chemistry , Protein Conformation , Proteome , Repressor Proteins/chemistry , Systems Biology
15.
Mol Syst Biol ; 12(4): 863, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27107012

ABSTRACT

High-throughput binary protein interaction mapping is continuing to extend our understanding of cellular function and disease mechanisms. However, we remain one or two orders of magnitude away from a complete interaction map for humans and other major model organisms. Completion will require screening at substantially larger scales with many complementary assays, requiring further efficiency gains in proteome-scale interaction mapping. Here, we report Barcode Fusion Genetics-Yeast Two-Hybrid (BFG-Y2H), by which a full matrix of protein pairs can be screened in a single multiplexed strain pool. BFG-Y2H uses Cre recombination to fuse DNA barcodes from distinct plasmids, generating chimeric protein-pair barcodes that can be quantified via next-generation sequencing. We applied BFG-Y2H to four different matrices ranging in scale from ~25 K to 2.5 M protein pairs. The results show that BFG-Y2H increases the efficiency of protein matrix screening, with quality that is on par with state-of-the-art Y2H methods.


Subject(s)
Centrosome/metabolism , Protein Interaction Mapping/methods , Proteome/metabolism , Saccharomyces cerevisiae/genetics , Chromosomes, Human/metabolism , Gene Library , High-Throughput Nucleotide Sequencing , Humans , Protein Binding , Two-Hybrid System Techniques
16.
J Biol Chem ; 290(8): 4631-4646, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25505242

ABSTRACT

After cell entry, HIV undergoes rapid transport toward the nucleus using microtubules and microfilaments. Neither the cellular cytoplasmic components nor the viral proteins that interact to mediate transport have yet been identified. Using a yeast two-hybrid screen, we identified four cytoskeletal components as putative interaction partners for HIV-1 p24 capsid protein: MAP1A, MAP1S, CKAP1, and WIRE. Depletion of MAP1A/MAP1S in indicator cell lines and primary human macrophages led to a profound reduction in HIV-1 infectivity as a result of impaired retrograde trafficking, demonstrated by a characteristic accumulation of capsids away from the nuclear membrane, and an overall defect in nuclear import. MAP1A/MAP1S did not impact microtubule network integrity or cell morphology but contributed to microtubule stabilization, which was shown previously to facilitate infection. In addition, we found that MAP1 proteins interact with HIV-1 cores both in vitro and in infected cells and that interaction involves MAP1 light chain LC2. Depletion of MAP1 proteins reduced the association of HIV-1 capsids with both dynamic and stable microtubules, suggesting that MAP1 proteins help tether incoming viral capsids to the microtubular network, thus promoting cytoplasmic trafficking. This work shows for the first time that following entry into target cells, HIV-1 interacts with the cytoskeleton via its p24 capsid protein. Moreover, our results support a role for MAP1 proteins in promoting efficient retrograde trafficking of HIV-1 by stimulating the formation of stable microtubules and mediating the association of HIV-1 cores with microtubules.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleus/metabolism , HIV-1/metabolism , Macrophages/metabolism , Microtubule-Associated Proteins/metabolism , Active Transport, Cell Nucleus/genetics , Carrier Proteins/genetics , Cell Line , Cell Nucleus/genetics , Cell Nucleus/virology , HIV Core Protein p24/genetics , HIV Core Protein p24/metabolism , HIV-1/genetics , Humans , Macrophages/pathology , Macrophages/virology , Microfilament Proteins , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Microtubules/metabolism , Microtubules/pathology
17.
J Biol Chem ; 290(30): 18609-20, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26085104

ABSTRACT

Amyloid precursor-like protein 2 (APLP2) and sortilin were reported to individually bind the proprotein convertase subtilisin/kexin type 9 (PCSK9) and regulate its activity on the low-density lipoprotein receptor (LDLR). The data presented herein demonstrate that mRNA knockdowns of APLP2, sortilin, or both in the human hepatocyte cell lines HepG2 and Huh7 do not affect the ability of extracellular PCSK9 to enhance the degradation of the LDLR. Furthermore, mice deficient in APLP2 or sortilin do not exhibit significant changes in liver LDLR or plasma total cholesterol levels. Moreover, cellular overexpression of one or both proteins does not alter PCSK9 secretion, or its activity on the LDLR. We conclude that PCSK9 enhances the degradation of the LDLR independently of either APLP2 or sortilin both ex vivo and in mice. Interestingly, when co-expressed with PCSK9, both APLP2 and sortilin were targeted for lysosomal degradation. Using chemiluminescence proximity and co-immunoprecipitation assays, as well as biosynthetic analysis, we discovered that sortilin binds and stabilizes APLP2, and hence could regulate its intracellular functions on other targets.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Amyloid beta-Protein Precursor/metabolism , Nerve Tissue Proteins/metabolism , Proprotein Convertases/metabolism , Proteolysis , Receptors, LDL/biosynthesis , Serine Endopeptidases/metabolism , Adaptor Proteins, Vesicular Transport/biosynthesis , Adaptor Proteins, Vesicular Transport/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Gene Expression Regulation , Hep G2 Cells , Hepatocytes/metabolism , Humans , Liver/metabolism , Mice , Nerve Tissue Proteins/genetics , Proprotein Convertase 9 , Proprotein Convertases/genetics , Receptors, LDL/genetics , Serine Endopeptidases/genetics
18.
PLoS Pathog ; 10(6): e1004164, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24945353

ABSTRACT

Influenza A viruses are major pathogens in humans and in animals, whose genome consists of eight single-stranded RNA segments of negative polarity. Viral mRNAs are synthesized by the viral RNA-dependent RNA polymerase in the nucleus of infected cells, in close association with the cellular transcriptional machinery. Two proteins essential for viral multiplication, the exportin NS2/NEP and the ion channel protein M2, are produced by splicing of the NS1 and M1 mRNAs, respectively. Here we identify two human spliceosomal factors, RED and SMU1, that control the expression of NS2/NEP and are required for efficient viral multiplication. We provide several lines of evidence that in infected cells, the hetero-trimeric viral polymerase recruits a complex formed by RED and SMU1 through interaction with its PB2 and PB1 subunits. We demonstrate that the splicing of the NS1 viral mRNA is specifically affected in cells depleted of RED or SMU1, leading to a decreased production of the spliced mRNA species NS2, and to a reduced NS2/NS1 protein ratio. In agreement with the exportin function of NS2, these defects impair the transport of newly synthesized viral ribonucleoproteins from the nucleus to the cytoplasm, and strongly reduce the production of infectious influenza virions. Overall, our results unravel a new mechanism of viral subversion of the cellular splicing machinery, by establishing that the human splicing factors RED and SMU1 act jointly as key regulators of influenza virus gene expression. In addition, our data point to a central role of the viral RNA polymerase in coupling transcription and alternative splicing of the viral mRNAs.


Subject(s)
Alternative Splicing , Chromosomal Proteins, Non-Histone/metabolism , Cytokines/metabolism , Influenza A virus/physiology , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Active Transport, Cell Nucleus , Cell Line , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Cytokines/antagonists & inhibitors , Cytokines/chemistry , Cytokines/genetics , Gene Silencing , Host-Pathogen Interactions , Humans , Influenza A virus/enzymology , Karyopherins/genetics , Karyopherins/metabolism , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , RNA-Dependent RNA Polymerase/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Spliceosomes/enzymology , Spliceosomes/metabolism , Two-Hybrid System Techniques , Viral Proteins/genetics , Virus Replication
19.
PLoS Pathog ; 9(10): e1003678, 2013.
Article in English | MEDLINE | ID: mdl-24098125

ABSTRACT

Searching for stimulators of the innate antiviral response is an appealing approach to develop novel therapeutics against viral infections. Here, we established a cell-based reporter assay to identify compounds stimulating expression of interferon-inducible antiviral genes. DD264 was selected out of 41,353 compounds for both its immuno-stimulatory and antiviral properties. While searching for its mode of action, we identified DD264 as an inhibitor of pyrimidine biosynthesis pathway. This metabolic pathway was recently identified as a prime target of broad-spectrum antiviral molecules, but our data unraveled a yet unsuspected link with innate immunity. Indeed, we showed that DD264 or brequinar, a well-known inhibitor of pyrimidine biosynthesis pathway, both enhanced the expression of antiviral genes in human cells. Furthermore, antiviral activity of DD264 or brequinar was found strictly dependent on cellular gene transcription, nuclear export machinery, and required IRF1 transcription factor. In conclusion, the antiviral property of pyrimidine biosynthesis inhibitors is not a direct consequence of pyrimidine deprivation on the virus machinery, but rather involves the induction of cellular immune response.


Subject(s)
Alphavirus Infections/metabolism , Antiviral Agents/pharmacology , Chikungunya virus/metabolism , Immunity, Innate/drug effects , Pyrimidines/biosynthesis , Alphavirus Infections/drug therapy , Alphavirus Infections/genetics , Alphavirus Infections/immunology , Animals , Antiviral Agents/chemistry , Chikungunya Fever , Chikungunya virus/genetics , Chikungunya virus/immunology , Chlorocebus aethiops , HeLa Cells , Humans , Immunity, Innate/immunology , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/immunology , Interferon Regulatory Factor-1/metabolism , Pyrimidines/immunology , Vero Cells
20.
EMBO Rep ; 14(6): 534-44, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23619093

ABSTRACT

Chikungunya virus (CHIKV) is a recently re-emerged arbovirus that triggers autophagy. Here, we show that CHIKV interacts with components of the autophagy machinery during its replication cycle, inducing a cytoprotective effect. The autophagy receptor p62 protects cells from death by binding ubiquitinated capsid and targeting it to autophagolysosomes. By contrast, the human autophagy receptor NDP52--but not its mouse orthologue--interacts with the non-structural protein nsP2, thereby promoting viral replication. These results highlight the distinct roles of p62 and NDP52 in viral infection, and identify NDP52 as a cellular factor that accounts for CHIKV species specificity.


Subject(s)
Alphavirus Infections/virology , Autophagy , Chikungunya virus/physiology , Virus Replication , Adaptor Proteins, Signal Transducing/metabolism , Animals , Capsid/metabolism , Chikungunya Fever , HeLa Cells , Host-Pathogen Interactions , Humans , Immunity, Innate , Mice , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Phagosomes/metabolism , Phagosomes/virology , Protein Binding , Protein Transport , Sequestosome-1 Protein , Sirolimus/pharmacology , Species Specificity , Viral Nonstructural Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL