Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Pathog ; 18(9): e1010329, 2022 09.
Article in English | MEDLINE | ID: mdl-36074777

ABSTRACT

Efficient virus replication in Aedes vector mosquitoes is essential for the transmission of arboviral diseases such as dengue virus (DENV) in human populations. Like in vertebrates, virus-host protein-protein interactions are essential for viral replication and immune evasion in the mosquito vector. Here, 79 mosquito host proteins interacting with DENV non-structural proteins NS1 and NS5 were identified by label-free mass spectrometry, followed by a functional screening. We confirmed interactions with host factors previously observed in mammals, such as the oligosaccharyltransferase complex, and we identified protein-protein interactions that seem to be specific for mosquitoes. Among the interactors, the double-stranded RNA (dsRNA) binding protein Loquacious (Loqs), an RNA interference (RNAi) cofactor, was found to be essential for efficient replication of DENV and Zika virus (ZIKV) in mosquito cells. Loqs did not affect viral RNA stability or translation of a DENV replicon and its proviral activity was independent of its RNAi regulatory activity. Interestingly, Loqs colocalized with DENV dsRNA replication intermediates in infected cells and directly interacted with high affinity with DENV RNA in the 3' untranslated region in vitro (KD = 48-62 nM). Our study provides an interactome for DENV NS1 and NS5 and identifies Loqs as a key proviral host factor in mosquitoes. We propose that DENV hijacks a factor of the RNAi mechanism for replication of its own RNA.


Subject(s)
Aedes , Arboviruses , Dengue Virus , Dengue , Zika Virus Infection , Zika Virus , 3' Untranslated Regions , Animals , Arboviruses/genetics , Dengue Virus/genetics , Humans , Mammals , Mosquito Vectors , RNA, Double-Stranded/metabolism , Virus Replication/genetics , Zika Virus/genetics
2.
Sci Rep ; 12(1): 9987, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35705637

ABSTRACT

Bioluminescence resonance energy transfer (BRET) saturation is a method of studying protein-protein interaction (PPI) upon quantification of the dependence of the BRET signal on the acceptor/donor (A:D) expression ratio. In this study, using the very bright Nluc/YFP BRET pair acquired respectively with microplate reader and automated confocal microscopy, we significantly improved BRET saturation assay by extending A:D expression detection range and normalizing A:D expression with a new BRET-free probe. We next found that upon using variable instead of fixed amount of donor molecules co-expressed with increasing acceptor concentrations, BRET saturation assay robustness can be further improved when studying cytosolic protein, although the relative amounts of dimers (BRETmax) and the relative dimer affinity (BRET50) remain similar. Altogether, we show that our method can be applied to many PPI networks, involving the NF-κB pathway, high-affinity nanobody, rabies virus-host interactions, mTOR complex and JAK/STAT signaling. Altogether our approach paves the way for robust PPI validation and characterization in living cells.


Subject(s)
Luminescent Measurements , Protein Interaction Maps , Biological Assay , Energy Transfer , Luminescent Measurements/methods , Signal Transduction
3.
PLoS One ; 15(5): e0232585, 2020.
Article in English | MEDLINE | ID: mdl-32374750

ABSTRACT

Neurotropic flavivirus Japanese encephalitis virus (JEV) and West Nile virus (WNV) are amongst the leading causes of encephalitis. Using label-free quantitative proteomics, we identified proteins differentially expressed upon JEV (gp-3, RP9) or WNV (IS98) infection of human neuroblastoma cells. Data are available via ProteomeXchange with identifier PXD016805. Both viruses were associated with the up-regulation of immune response (IFIT1/3/5, ISG15, OAS, STAT1, IRF9) and the down-regulation of SSBP2 and PAM, involved in gene expression and in neuropeptide amidation respectively. Proteins associated to membranes, involved in extracellular matrix organization and collagen metabolism represented major clusters down-regulated by JEV and WNV. Moreover, transcription regulation and mRNA processing clusters were also heavily regulated by both viruses. The proteome of neuroblastoma cells infected by JEV or WNV was significantly modulated in the presence of mosquito saliva, but distinct patterns were associated to each virus. Mosquito saliva favored modulation of proteins associated with gene regulation in JEV infected neuroblastoma cells while modulation of proteins associated with protein maturation, signal transduction and ion transporters was found in WNV infected neuroblastoma cells.


Subject(s)
Culicidae/metabolism , Encephalitis, Japanese/metabolism , Neurons/pathology , Proteome/metabolism , West Nile Fever/metabolism , Animals , Cell Line, Tumor , Culicidae/virology , Encephalitis Viruses, Japanese/isolation & purification , Encephalitis, Japanese/pathology , Encephalitis, Japanese/virology , Female , Humans , Neurons/metabolism , Neurons/virology , Proteome/analysis , Saliva/metabolism , Saliva/virology , West Nile Fever/pathology , West Nile Fever/virology , West Nile virus/isolation & purification
4.
Sci Rep ; 9(1): 12171, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31434934

ABSTRACT

Phosphoprotein (P) and matrix protein (M) cooperate to undermine the immune response to rabies virus (RABV) infections. While P is involved in the modulation of the Jak-Stat pathway through the cytoplasmic retention of interferon (IFN)-activated STAT1 (pSTAT1), M interacts with the RelAp43-p105-ABIN2-TPL2 complex, to efficiently inhibit the nuclear factor-κB (NF-κB) pathway. Using transfections, protein-complementation assays, reverse genetics and DNA ChIP, we identified a role of M protein in the control of Jak-Stat signaling pathway, in synergy with the P protein. In unstimulated cells, both M and P proteins were found to interact with JAK1. Upon type-I IFN stimulation, the M switches toward pSTAT1 interaction, which results in an enhanced capacity of P protein to interact with pSTAT1 and restrain it in the cytoplasm. Furthermore, the role for M-protein positions 77, 100, 104 and 110 was also demonstrated in interaction with both JAK1 and pY-STAT1, and confirmed in vivo. Together, these data indicate that M protein cooperates with P protein to restrain in parallel, and sequentially, NF-κB and Jak-Stat pathways.


Subject(s)
Lyssavirus/metabolism , Phosphoproteins/metabolism , Signal Transduction , Viral Matrix Proteins/metabolism , Viral Proteins/metabolism , Animals , Cytoplasm/metabolism , HeLa Cells , Humans , Immunity, Innate , Interferon Type I/metabolism , Janus Kinase 1/metabolism , Lyssavirus/pathogenicity , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , NF-kappa B/metabolism , Phosphoproteins/genetics , Promoter Regions, Genetic , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Viral Matrix Proteins/genetics , Viral Proteins/genetics , Virulence
5.
mSphere ; 4(3)2019 05 22.
Article in English | MEDLINE | ID: mdl-31118297

ABSTRACT

Throughout the rabies virus (RABV) infectious cycle, host-virus interactions define its capacity to replicate, escape the immune response, and spread. As phosphorylation is a key regulatory mechanism involved in most cellular processes, kinases represent a target of choice to identify host factors required for viral replication. A kinase and phosphatase small interfering RNA (siRNA) high-content screening was performed on a fluorescent protein-recombinant field isolate (Tha RABV). We identified 57 high-confidence key host factors important for RABV replication with a readout set at 18 h postinfection and 73 with a readout set at 36 h postinfection, including 24 common factors at all stages of the infection. Amongst them, gene clusters of the most prominent pathways were determined. Up to 15 mitogen-activated protein kinases (MAPKs) and effectors, including MKK7 (associated with Jun N-terminal protein kinase [JNK] signalization) and DUSP5, as well as 17 phosphatidylinositol (PI)-related proteins, including PIP5K1C and MTM1, were found to be involved in the later stage of RABV infection. The importance of these pathways was further validated, as small molecules Ro 31-8820 and PD 198306 inhibited RABV replication in human neurons.IMPORTANCE Rabies virus relies on cellular machinery for its replication while simultaneously evading the host immune response. Despite their importance, little is known about the key host factors required for rabies virus infection. Here, we focused on the human kinome, at the core of many cellular pathways, to unveil a new understanding of the rabies virus infectious cycle and to discover new potential therapeutic targets in a small interfering RNA screening. The mitogen-activated protein kinase pathway and phosphatidylinositol metabolism were identified as prominent factors involved in rabies virus infection, and those findings were further confirmed in human neurons. While bringing a new insight into rabies virus biology, we also provide a new list of host factors involved in rabies virus infection.


Subject(s)
Host Microbial Interactions , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositols/metabolism , RNA Interference , Rabies virus/physiology , Cell Line , High-Throughput Screening Assays , Humans , Metabolic Networks and Pathways , Rabies virus/genetics , Small Molecule Libraries , Virus Replication
6.
PLoS Pathog ; 13(10): e1006697, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29084252

ABSTRACT

At the crossroad between the NF-κB and the MAPK pathways, the ternary complex composed of p105, ABIN2 and TPL2 is essential for the host cell response to pathogens. The matrix protein (M) of field isolates of rabies virus was previously shown to disturb the signaling induced by RelAp43, a NF-κB protein close to RelA/p65. Here, we investigated how the M protein disturbs the NF-κB pathway in a RelAp43-dependant manner and the potential involvement of the ternary complex in this mechanism. Using a tandem affinity purification coupled with mass spectrometry approach, we show that RelAp43 interacts with the p105-ABIN2-TPL2 complex and we observe a strong perturbation of this complex in presence of M protein. M protein interaction with RelAp43 is associated with a wide disturbance of NF-κB signaling, involving a modulation of IκBα-, IκBß-, and IκBε-RelAp43 interaction and a favored interaction of RelAp43 with the non-canonical pathway (RelB and p100/p52). Monitoring the interactions between host and viral proteins using protein-fragment complementation assay and bioluminescent resonance energy transfer, we further show that RelAp43 is associated to the p105-ABIN2-TPL2 complex as RelAp43-p105 interaction stabilizes the formation of a complex with ABIN2 and TPL2. Interestingly, the M protein interacts not only with RelAp43 but also with TPL2 and ABIN2. Upon interaction with this complex, M protein promotes the release of ABIN2, which ultimately favors the production of RelAp43-p50 NF-κB dimers. The use of recombinant rabies viruses further indicates that this mechanism leads to the control of IFNß, TNF and CXCL2 expression during the infection and a high pathogenicity profile in rabies virus infected mice. All together, our results demonstrate the important role of RelAp43 and M protein in the regulation of NF-κB signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Multiprotein Complexes/metabolism , NF-kappa B p50 Subunit/metabolism , Proto-Oncogene Proteins/metabolism , Rabies virus/metabolism , Rabies/metabolism , Adaptor Proteins, Signal Transducing/genetics , HEK293 Cells , HeLa Cells , Humans , MAP Kinase Kinase Kinases/genetics , Multiprotein Complexes/genetics , NF-kappa B p50 Subunit/genetics , Proto-Oncogene Proteins/genetics , Rabies/genetics , Rabies virus/genetics , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
7.
Sci Rep ; 6: 39420, 2016 12 21.
Article in English | MEDLINE | ID: mdl-28000711

ABSTRACT

The matrix (M) protein of wild isolates of rabies virus such as Tha (M-Tha) was previously shown to be able to interact with RelAp43, a protein of the NF-κB family, and to efficiently suppress NF-κB-dependent reporter gene expression, in contrast with the vaccine strain SAD. Here, we analyze the mechanisms involved in RelAp43-M protein interaction. We demonstrate that the central part of M-Tha, and the specific C-terminal region of RelAp43 are required for this interaction. Four differences in the corresponding amino acid sequences of the M-Tha and M-SAD are shown to be crucial for RelAp43 interaction and subsequent modulation of innate immune response. Furthermore, the capacity of M-Tha to interact with RelAp43 was shown to be crucial for the control of the expression of four genes (IFN, TNF, IL8 and CXCL2) during viral infection. These findings reveal that RelAp43 is a potent regulator of transcription of genes involved in innate immune response during rabies virus infection and that the M protein of wild isolates of rabies virus is a viral immune-modulatory factor playing an important role in this RelAp43-mediated host innate immunity response in contrast to M protein of vaccine strains, which have lost this property.


Subject(s)
Gene Expression/immunology , Immunity, Innate/immunology , NF-kappa B/metabolism , Rabies virus/immunology , Rabies virus/metabolism , Rabies/metabolism , Viral Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/immunology , Humans , Mice, Inbred BALB C , Rabies/immunology , Rabies/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...