Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.506
Filtrer
1.
ACS Nano ; 18(24): 15545-15556, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38838261

RÉSUMÉ

Deterministic formation of membrane scission necks by protein machinery with multiplexed functions is critical in biology. A microbial example is M2 viroporin, a proton pump from the influenza A virus that is multiplexed with membrane remodeling activity to induce budding and scission in the host membrane during viral maturation. In comparison, the dynamin family constitutes a class of eukaryotic proteins implicated in mitochondrial fission, as well as various budding and endocytosis pathways. In the case of Dnm1, the mitochondrial fission protein in yeast, the membrane remodeling activity is multiplexed with mechanoenzyme activity to create fission necks. It is not clear why these functions are combined in these scission processes, which occur in drastically different compositions and solution conditions. In general, direct experimental access to changing neck sizes induced by individual proteins or peptide fragments is challenging due to the nanoscale dimensions and influence of thermal fluctuations. Here, we use a mechanical model to estimate the size of scission necks by leveraging small-angle X-ray scattering structural data of protein-lipid systems under different conditions. The influence of interfacial tension, lipid composition, and membrane budding morphology on the size of the induced scission necks is systematically investigated using our data and molecular dynamic simulations. We find that the M2 budding protein from the influenza A virus has robust pH-dependent membrane activity that induces nanoscopic necks within the range of spontaneous hemifission for a broad range of lipid compositions. In contrast, the sizes of scission necks generated by mitochondrial fission proteins strongly depend on lipid composition, which suggests a role for mechanical constriction.


Sujet(s)
Membrane cellulaire , Membrane cellulaire/métabolisme , Membrane cellulaire/composition chimique , Protéines de la matrice virale/métabolisme , Protéines de la matrice virale/composition chimique , Dynamines/métabolisme , Dynamines/composition chimique , Virus de la grippe A/métabolisme , Diffusion aux petits angles , Protéines viroporines
2.
Virus Res ; 345: 199387, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38719025

RÉSUMÉ

Influenza A virus can infect respiratory tracts and may cause severe illness in humans. Proteins encoded by influenza A virus can interact with cellular factors and dysregulate host biological processes to support viral replication and cause pathogenicity. The influenza viral PA protein is not only a subunit of influenza viral polymerase but also a virulence factor involved in pathogenicity during infection. To explore the role of the influenza virus PA protein in regulating host biological processes, we performed immunoprecipitation and LC‒MS/MS to globally identify cellular factors that interact with the PA proteins of the influenza A H1N1, 2009 pandemic H1N1, and H3N2 viruses. The results demonstrated that proteins located in the mitochondrion, proteasome, and nucleus are associated with the PA protein. We further discovered that the PA protein is partly located in mitochondria by immunofluorescence and mitochondrial fractionation and that overexpression of the PA protein reduces mitochondrial respiration. In addition, our results revealed the interaction between PA and the mitochondrial matrix protein PYCR2 and the antiviral role of PYCR2 during influenza A virus replication. Moreover, we found that the PA protein could also trigger autophagy and disrupt mitochondrial homeostasis. Overall, our research revealed the impacts of the influenza A virus PA protein on mitochondrial function and autophagy.


Sujet(s)
Mitochondries , Protéines virales , Réplication virale , Humains , Mitochondries/métabolisme , Mitochondries/virologie , Protéines virales/métabolisme , Protéines virales/génétique , RNA replicase/métabolisme , RNA replicase/génétique , Virus de la grippe A/physiologie , Virus de la grippe A/génétique , Virus de la grippe A/pathogénicité , Virus de la grippe A/métabolisme , Interactions hôte-pathogène , Sous-type H3N2 du virus de la grippe A/génétique , Sous-type H3N2 du virus de la grippe A/physiologie , Sous-type H3N2 du virus de la grippe A/métabolisme , Autophagie , Sous-type H1N1 du virus de la grippe A/génétique , Sous-type H1N1 du virus de la grippe A/physiologie , Sous-type H1N1 du virus de la grippe A/pathogénicité , Cellules HEK293 , Grippe humaine/virologie , Grippe humaine/métabolisme , Cellules A549 , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Spectrométrie de masse en tandem
3.
PLoS Pathog ; 20(5): e1012279, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38814988

RÉSUMÉ

The influenza A virus (IAV) consists of 8 single-stranded, negative-sense viral RNA (vRNA) segments. After infection, vRNA is transcribed, replicated, and wrapped by viral nucleoprotein (NP) to form viral ribonucleoprotein (vRNP). The transcription, replication, and nuclear export of the viral genome are regulated by the IAV protein, NS2, which is translated from spliced mRNA transcribed from viral NS vRNA. This splicing is inefficient, explaining why NS2 is present in low abundance after IAV infection. The levels of NS2 and its subsequent accumulation are thought to influence viral RNA replication and vRNP nuclear export. Here we show that NS2 is ubiquitinated at the K64 and K88 residues by K48-linked and K63-linked polyubiquitin (polyUb) chains, leading to the degradation of NS2 by the proteasome. Additionally, we show that a host deubiquitinase, OTUB1, can remove polyUb chains conjugated to NS2, thereby stabilizing NS2. Accordingly, knock down of OTUB1 by siRNA reduces the nuclear export of vRNP, and reduces the overall production of IAV. These results collectively demonstrate that the levels of NS2 in IAV-infected cells are regulated by a ubiquitination-deubiquitination system involving OTUB1 that is necessary for optimal IAV replication.


Sujet(s)
Cysteine endopeptidases , Virus de la grippe A , Protéines virales non structurales , Réplication virale , Animaux , Chiens , Humains , Cysteine endopeptidases/métabolisme , Cysteine endopeptidases/génétique , Enzymes de désubiquitinylation/métabolisme , Cellules HEK293 , Virus de la grippe A/métabolisme , Grippe humaine/métabolisme , Grippe humaine/virologie , ARN viral/métabolisme , ARN viral/génétique , Ubiquitination , Protéines virales non structurales/métabolisme , Protéines virales non structurales/génétique , Réplication virale/physiologie , Lignée cellulaire , Cellules Vero , Chlorocebus aethiops
4.
Protein Expr Purif ; 221: 106506, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38772430

RÉSUMÉ

Influenza poses a substantial health risk, with infants and the elderly being particularly susceptible to its grave impacts. The primary challenge lies in its rapid genetic evolution, leading to the emergence of new Influenza A strains annually. These changes involve punctual mutations predominantly affecting the two main glycoproteins: Hemagglutinin (HA) and Neuraminidase (NA). Our existing vaccines target these proteins, providing short-term protection, but fall short when unexpected pandemics strike. Delving deeper into Influenza's genetic makeup, we spotlight the nucleoprotein (NP) - a key player in the transcription, replication, and packaging of RNA. An intriguing characteristic of the NP is that it is highly conserved across all Influenza A variants, potentially paving the way for a more versatile and broadly protective vaccine. We designed and synthesized a novel NP-Hoc fusion protein combining Influenza A nucleoprotein and T4 phage Hoc, cloned using Gibson assembly in E. coli, and purified via ion affinity chromatography. Simultaneously, we explore the T4 coat protein Hoc, typically regarded as inconsequential in controlled viral replication. Yet, it possesses a unique ability: it can link with another protein, showcasing it on the T4 phage coat. Fusing these concepts, our study designs, expresses, and purifies a novel fusion protein named NP-Hoc. We propose this protein as the basis for a new generation of vaccines, engineered to guard broadly against Influenza A. The excitement lies not just in the immediate application, but the promise this holds for future pandemic resilience, with NP-Hoc marking a significant leap in adaptive, broad-spectrum influenza prevention.


Sujet(s)
Bactériophage T4 , Escherichia coli , Protéines de fusion recombinantes , Bactériophage T4/génétique , Bactériophage T4/composition chimique , Bactériophage T4/métabolisme , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/isolement et purification , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/biosynthèse , Escherichia coli/génétique , Escherichia coli/métabolisme , Expression des gènes , Protéines nucléocapside/génétique , Protéines nucléocapside/composition chimique , Protéines nucléocapside/métabolisme , Virus de la grippe A/génétique , Virus de la grippe A/métabolisme , Vaccins antigrippaux/génétique , Vaccins antigrippaux/biosynthèse , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/composition chimique , Humains , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/composition chimique , Protéines de liaison à l'ARN/isolement et purification
5.
J Virol ; 98(5): e0013824, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38563748

RÉSUMÉ

Influenza A viruses, causing seasonal epidemics and occasional pandemics, rely on interactions with host proteins for their RNA genome transcription and replication. The viral RNA polymerase utilizes host RNA polymerase II (Pol II) and interacts with the serine 5 phosphorylated (pS5) C-terminal domain (CTD) of Pol II to initiate transcription. Our study, using single-particle electron cryomicroscopy (cryo-EM), reveals the structure of the 1918 pandemic influenza A virus polymerase bound to a synthetic pS5 CTD peptide composed of four heptad repeats mimicking the 52 heptad repeat mammalian Pol II CTD. The structure shows that the CTD peptide binds at the C-terminal domain of the PA viral polymerase subunit (PA-C) and reveals a previously unobserved position of the 627 domain of the PB2 subunit near the CTD. We identify crucial residues of the CTD peptide that mediate interactions with positively charged cavities on PA-C, explaining the preference of the viral polymerase for pS5 CTD. Functional analysis of mutants targeting the CTD-binding site within PA-C reveals reduced transcriptional function or defects in replication, highlighting the multifunctional role of PA-C in viral RNA synthesis. Our study provides insights into the structural and functional aspects of the influenza virus polymerase-host Pol II interaction and identifies a target for antiviral development.IMPORTANCEUnderstanding the intricate interactions between influenza A viruses and host proteins is crucial for developing targeted antiviral strategies. This study employs advanced imaging techniques to uncover the structural nuances of the 1918 pandemic influenza A virus polymerase bound to a specific host protein, shedding light on the vital process of viral RNA synthesis. The study identifies key amino acid residues in the influenza polymerase involved in binding host polymerase II (Pol II) and highlights their role in both viral transcription and genome replication. These findings not only deepen our understanding of the influenza virus life cycle but also pinpoint a potential target for antiviral development. By elucidating the structural and functional aspects of the influenza virus polymerase-host Pol II interaction, this research provides a foundation for designing interventions to disrupt viral replication and transcription, offering promising avenues for future antiviral therapies.


Sujet(s)
Cryomicroscopie électronique , Virus de la grippe A , RNA polymerase II , RNA replicase , Protéines virales , Humains , Virus de la grippe A/métabolisme , Virus de la grippe A/génétique , Virus de la grippe A/enzymologie , Grippe humaine/virologie , Modèles moléculaires , Phosphorylation , Liaison aux protéines , Domaines protéiques , RNA polymerase II/métabolisme , RNA polymerase II/composition chimique , ARN viral/métabolisme , ARN viral/génétique , RNA replicase/métabolisme , RNA replicase/composition chimique , Transcription génétique , Protéines virales/métabolisme , Protéines virales/composition chimique , Protéines virales/génétique , Réplication virale
6.
J Virol ; 98(5): e0041124, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38567952

RÉSUMÉ

Influenza A virus infection activates the NLRP3 inflammasome, a multiprotein signaling complex responsible for the proteolytic activation and release of the proinflammatory cytokine IL-1ß from monocytes and macrophages. Some influenza A virus (IAV) strains encode a short 90-amino acid peptide (PB1-F2) on an alternative open reading frame of segment 2, with immunomodulatory activity. We recently demonstrated that contemporary IAV PB1-F2 inhibits the activation of NLRP3, potentially by NEK7-dependent activation. PB1-F2 binds to NLRP3 with its C-terminal 50 amino acids, but the exact binding motif was unknown. On the NLRP3 side, the interface is formed through the leucine-rich-repeat (LRR) domain, potentially in conjunction with the pyrin domain. Here, we took advantage of PB1-F2 sequences from IAV strains with either weak or strong NLRP3 interaction. Sequence comparison and structure prediction using Alphafold2 identified a short four amino acid sequence motif (TQGS) in PB1-F2 that defines NLRP3-LRR binding. Conversion of this motif to that of the non-binding PB1-F2 suffices to lose inhibition of NLRP3 dependent IL-1ß release. The TQGS motif further alters the subcellular localization of PB1-F2 and its colocalization with NLRP3 LRR and pyrin domain. Structural predictions suggest the establishment of additional hydrogen bonds between the C-terminus of PB1-F2 and the LRR domain of NLRP3, with two hydrogen bonds connecting to threonine and glutamine of the TQGS motif. Phylogenetic data show that the identified NLRP3 interaction motif in PB1-F2 is widely conserved among recent IAV-infecting humans. Our data explain at a molecular level the specificity of NLRP3 inhibition by influenza A virus. IMPORTANCE: Influenza A virus infection is accompanied by a strong inflammatory response and high fever. The human immune system facilitates the swift clearance of the virus with this response. An essential signal protein in the proinflammatory host response is IL-1b. It is released from inflammatory macrophages, and its production and secretion depend on the function of NLRP3. We had previously shown that influenza A virus blocks NLRP3 activation by the expression of a viral inhibitor, PB1-F2. Here, we demonstrate how this short peptide binds to NLRP3 and provide evidence that a four amino acid stretch in PB1-F2 is necessary and sufficient to mediate this binding. Our data identify a new virus-host interface required to block one signaling path of the innate host response against influenza A virus.


Sujet(s)
Virus de la grippe A , Protéine-3 de la famille des NLR contenant un domaine pyrine , Protéines virales , Humains , Motifs d'acides aminés , Séquence d'acides aminés , Cellules HEK293 , Inflammasomes/métabolisme , Virus de la grippe A/génétique , Virus de la grippe A/métabolisme , Grippe humaine/virologie , Grippe humaine/immunologie , Interleukine-1 bêta/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/composition chimique , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Liaison aux protéines , Protéines virales/métabolisme , Protéines virales/génétique , Protéines virales/composition chimique
7.
Chemistry ; 30(32): e202401108, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38567703

RÉSUMÉ

Sialyl-Lewisx (SLex) is involved in immune regulation, human fertilization, cancer, and bacterial and viral diseases. The influence of the complex glycan structures, which can present SLex epitopes, on binding is largely unknown. We report here a chemoenzymatic strategy for the preparation of a panel of twenty-two isomeric asymmetrical tri-antennary N-glycans presenting SLex-Lex epitopes on either the MGAT4 or MGAT5 arm that include putative high-affinity ligands for E-selectin. The N-glycans were prepared starting from a sialoglycopeptide isolated from egg yolk powder and took advantage of inherent substrate preferences of glycosyltransferases and the use of 5'-diphospho-N-trifluoracetylglucosamine (UDP-GlcNHTFA) that can be transferred by branching N-acetylglucosaminyltransferases to give, after base treatment, GlcNH2-containing glycans that temporarily disable an antenna from enzymatic modification. Glycan microarray binding studies showed that E-selectin bound equally well to linear glycans and tri-antennary N-glycans presenting SLex-Lex. On the other hand, it was found that hemagglutinins (HA) of H5 influenza A viruses (IAV) preferentially bound the tri-antennary N-glycans. Furthermore, several H5 HAs preferentially bound to N-glycan presenting SLex on the MGAT4 arm. SLex is displayed in the respiratory tract of several avian species, demonstrating the relevance of investigating the binding of, among others IAVs, to complex N-glycans presenting SLex.


Sujet(s)
Sélectine E , Virus de la grippe A , Polyosides , Antigène sialyl Lewis X , Polyosides/composition chimique , Polyosides/métabolisme , Virus de la grippe A/métabolisme , Antigène sialyl Lewis X/métabolisme , Antigène sialyl Lewis X/composition chimique , Sélectine E/métabolisme , Sélectine E/composition chimique , Humains , Oligosaccharides/composition chimique , Oligosaccharides/synthèse chimique , Oligosaccharides/métabolisme , Récepteurs viraux/métabolisme , Récepteurs viraux/composition chimique , Épitopes/composition chimique , Épitopes/métabolisme , Animaux
8.
Nucleic Acids Res ; 52(12): 7188-7210, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38686810

RÉSUMÉ

Genome-wide approaches have significantly advanced our knowledge of the repertoire of RNA-binding proteins (RBPs) that associate with cellular polyadenylated mRNAs within eukaryotic cells. Recent studies focusing on the RBP interactomes of viral mRNAs, notably SARS-Cov-2, have revealed both similarities and differences between the RBP profiles of viral and cellular mRNAs. However, the RBPome of influenza virus mRNAs remains unexplored. Herein, we identify RBPs that associate with the viral mRNA encoding the nucleoprotein (NP) of an influenza A virus. Focusing on TDP-43, we show that it binds several influenza mRNAs beyond the NP-mRNA, and that its depletion results in lower levels of viral mRNAs and proteins within infected cells, and a decreased yield of infectious viral particles. We provide evidence that the viral polymerase recruits TDP-43 onto viral mRNAs through a direct interaction with the disordered C-terminal domain of TDP-43. Notably, other RBPs found to be associated with influenza virus mRNAs also interact with the viral polymerase, which points to a role of the polymerase in orchestrating the assembly of viral messenger ribonucleoproteins.


Sujet(s)
Protéines de liaison à l'ADN , Virus de la grippe A , ARN messager , ARN viral , Protéines de liaison à l'ARN , Réplication virale , Humains , Réplication virale/génétique , ARN viral/métabolisme , ARN viral/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , ARN messager/métabolisme , ARN messager/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Virus de la grippe A/génétique , Virus de la grippe A/physiologie , Virus de la grippe A/métabolisme , Protéines nucléocapside/métabolisme , Protéines nucléocapside/génétique , Cellules HEK293 , Protéines du core viral/métabolisme , Protéines du core viral/génétique , Liaison aux protéines , Animaux
9.
J Virol ; 98(4): e0194123, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38470143

RÉSUMÉ

Influenza A viruses (IAVs) can overcome species barriers by adaptation of the receptor-binding site of the hemagglutinin (HA). To initiate infection, HAs bind to glycan receptors with terminal sialic acids, which are either N-acetylneuraminic acid (NeuAc) or N-glycolylneuraminic acid (NeuGc); the latter is mainly found in horses and pigs but not in birds and humans. We investigated the influence of previously identified equine NeuGc-adapting mutations (S128T, I130V, A135E, T189A, and K193R) in avian H7 IAVs in vitro and in vivo. We observed that these mutations negatively affected viral replication in chicken cells but not in duck cells and positively affected replication in horse cells. In vivo, the mutations reduced virus virulence and mortality in chickens. Ducks excreted high viral loads longer than chickens, although they appeared clinically healthy. To elucidate why these viruses infected chickens and ducks despite the absence of NeuGc, we re-evaluated the receptor binding of H7 HAs using glycan microarray and flow cytometry studies. This re-evaluation demonstrated that mutated avian H7 HAs also bound to α2,3-linked NeuAc and sialyl-LewisX, which have an additional fucose moiety in their terminal epitope, explaining why infection of ducks and chickens was possible. Interestingly, the α2,3-linked NeuAc and sialyl-LewisX epitopes were only bound when presented on tri-antennary N-glycans, emphasizing the importance of investigating the fine receptor specificities of IAVs. In conclusion, the binding of NeuGc-adapted H7 IAV to tri-antennary N-glycans enables viral replication and shedding by chickens and ducks, potentially facilitating interspecies transmission of equine-adapted H7 IAVs.IMPORTANCEInfluenza A viruses (IAVs) cause millions of deaths and illnesses in birds and mammals each year. The viral surface protein hemagglutinin initiates infection by binding to host cell terminal sialic acids. Hemagglutinin adaptations affect the binding affinity to these sialic acids and the potential host species targeted. While avian and human IAVs tend to bind to N-acetylneuraminic acid (sialic acid), equine H7 viruses prefer binding to N-glycolylneuraminic acid (NeuGc). To better understand the function of NeuGc-specific adaptations in hemagglutinin and to elucidate interspecies transmission potential NeuGc-adapted viruses, we evaluated the effects of NeuGc-specific mutations in avian H7 viruses in chickens and ducks, important economic hosts and reservoir birds, respectively. We also examined the impact on viral replication and found a binding affinity to tri-antennary N-glycans containing different terminal epitopes. These findings are significant as they contribute to the understanding of the role of receptor binding in avian influenza infection.


Sujet(s)
Poulets , Canards , Equus caballus , Virus de la grippe A , Grippe chez les oiseaux , Acides neuraminiques , Animaux , Humains , Poulets/génétique , Poulets/métabolisme , Poulets/virologie , Canards/génétique , Canards/métabolisme , Canards/virologie , Épitopes/composition chimique , Épitopes/métabolisme , Glycoprotéine hémagglutinine du virus influenza/composition chimique , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/métabolisme , Equus caballus/génétique , Equus caballus/métabolisme , Equus caballus/virologie , Virus de la grippe A/composition chimique , Virus de la grippe A/classification , Virus de la grippe A/métabolisme , Grippe chez les oiseaux/génétique , Grippe chez les oiseaux/transmission , Grippe chez les oiseaux/virologie , Mutation , Acide N-acétyl-neuraminique/composition chimique , Acide N-acétyl-neuraminique/métabolisme , Acides neuraminiques/composition chimique , Acides neuraminiques/métabolisme , Récepteurs viraux/composition chimique , Récepteurs viraux/génétique , Récepteurs viraux/métabolisme , Suidae/virologie , Zoonoses virales/métabolisme , Zoonoses virales/transmission , Zoonoses virales/virologie
10.
J Virol ; 98(4): e0010224, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38470058

RÉSUMÉ

The transmembrane serine protease 2 (TMPRSS2) activates the outer structural proteins of a number of respiratory viruses including influenza A virus (IAV), parainfluenza viruses, and various coronaviruses for membrane fusion. Previous studies showed that TMPRSS2 interacts with the carboxypeptidase angiotensin-converting enzyme 2 (ACE2), a cell surface protein that serves as an entry receptor for some coronaviruses. Here, by using protease activity assays, we determine that ACE2 increases the enzymatic activity of TMPRSS2 in a non-catalytic manner. Furthermore, we demonstrate that ACE2 knockdown inhibits TMPRSS2-mediated cleavage of IAV hemagglutinin (HA) in Calu-3 human airway cells and suppresses virus titers 100- to 1.000-fold. Transient expression of ACE2 in ACE2-deficient cells increased TMPRSS2-mediated HA cleavage and IAV replication. ACE2 knockdown also reduced titers of MERS-CoV and prevented S cleavage by TMPRSS2 in Calu-3 cells. By contrast, proteolytic activation and multicycle replication of IAV with multibasic HA cleavage site typically cleaved by furin were not affected by ACE2 knockdown. Co-immunoprecipitation analysis revealed that ACE2-TMPRSS2 interaction requires the enzymatic activity of TMPRSS2 and the carboxypeptidase domain of ACE2. Together, our data identify ACE2 as a new co-factor or stabilizer of TMPRSS2 activity and as a novel host cell factor involved in proteolytic activation and spread of IAV in human airway cells. Furthermore, our data indicate that ACE2 is involved in the TMPRSS2-catalyzed activation of additional respiratory viruses including MERS-CoV.IMPORTANCEProteolytic cleavage of viral envelope proteins by host cell proteases is essential for the infectivity of many viruses and relevant proteases provide promising drug targets. The transmembrane serine protease 2 (TMPRSS2) has been identified as a major activating protease of several respiratory viruses, including influenza A virus. TMPRSS2 was previously shown to interact with angiotensin-converting enzyme 2 (ACE2). Here, we report the mechanistic details of this interaction. We demonstrate that ACE2 increases or stabilizes the enzymatic activity of TMPRSS2. Furthermore, we describe ACE2 involvement in TMPRSS2-catalyzed cleavage of the influenza A virus hemagglutinin and MERS-CoV spike protein in human airway cells. These findings expand our knowledge of the activation of respiratory viruses by TMPRSS2 and the host cell factors involved. In addition, our results could help to elucidate a physiological role for TMPRSS2.


Sujet(s)
Angiotensin-converting enzyme 2 , Virus de la grippe A , Poumon , Protéolyse , Serine endopeptidases , Animaux , Chiens , Humains , Angiotensin-converting enzyme 2/déficit , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/métabolisme , Biocatalyse , Lignée cellulaire , Furine/métabolisme , Glycoprotéine hémagglutinine du virus influenza/métabolisme , Virus de la grippe A/croissance et développement , Virus de la grippe A/métabolisme , Poumon/cytologie , Poumon/virologie , Coronavirus du syndrome respiratoire du Moyen-Orient/métabolisme , Liaison aux protéines , Serine endopeptidases/métabolisme , Glycoprotéine de spicule des coronavirus/métabolisme , Pénétration virale , Réplication virale
11.
J Virol ; 98(4): e0197223, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38470155

RÉSUMÉ

The coordinated packaging of the segmented genome of the influenza A virus (IAV) into virions is an essential step of the viral life cycle. This process is controlled by the interaction of packaging signals present in all eight viral RNA (vRNA) segments and the viral nucleoprotein (NP), which binds vRNA via a positively charged binding groove. However, mechanistic models of how the packaging signals and NP work together to coordinate genome packaging are missing. Here, we studied genome packaging in influenza A/SC35M virus mutants that carry mutated packaging signals as well as specific amino acid substitutions at the highly conserved lysine (K) residues 184 and 229 in the RNA-binding groove of NP. Because these lysines are acetylated and thus neutrally charged in infected host cells, we replaced them with glutamine to mimic the acetylated, neutrally charged state or arginine to mimic the non-acetylated, positively charged state. Our analysis shows that the coordinated packaging of eight vRNAs is influenced by (i) the charge state of the replacing amino acid and (ii) its location within the RNA-binding groove. Accordingly, we propose that lysine acetylation induces different charge states within the RNA-binding groove of NP, thereby supporting the activity of specific packaging signals during coordinated genome packaging. IMPORTANCE: Influenza A viruses (IAVs) have a segmented viral RNA (vRNA) genome encapsidated by multiple copies of the viral nucleoprotein (NP) and organized into eight distinct viral ribonucleoprotein complexes. Although genome segmentation contributes significantly to viral evolution and adaptation, it requires a highly sophisticated genome-packaging mechanism. How eight distinct genome complexes are incorporated into the virion is poorly understood, but previous research suggests an essential role for both vRNA packaging signals and highly conserved NP amino acids. By demonstrating that the packaging process is controlled by charge-dependent interactions of highly conserved lysine residues in NP and vRNA packaging signals, our study provides new insights into the sophisticated packaging mechanism of IAVs.


Sujet(s)
Virus de la grippe A , Protéines nucléocapside , Encapsidation du génome viral , Animaux , Chiens , Humains , Substitution d'acide aminé , Lignée cellulaire , Génome viral , Virus de la grippe A/composition chimique , Virus de la grippe A/génétique , Virus de la grippe A/métabolisme , Lysine/génétique , Protéines nucléocapside/composition chimique , Protéines nucléocapside/génétique , Protéines nucléocapside/métabolisme , ARN viral/métabolisme , Encapsidation du génome viral/génétique , Virion/composition chimique , Virion/génétique , Virion/métabolisme , Mutation , Électricité statique
12.
Viruses ; 16(3)2024 03 09.
Article de Anglais | MEDLINE | ID: mdl-38543786

RÉSUMÉ

Influenza A viruses (IAVs) possess a segmented genome consisting of eight viral RNAs (vRNAs) associated with multiple copies of viral nucleoprotein (NP) and a viral polymerase complex. Despite the crucial role of RNA structure in IAV replication, the impact of NP binding on vRNA structure is not well understood. In this study, we employed SHAPE chemical probing to compare the structure of NS and M vRNAs of WSN IAV in various states: before the addition of NP, in complex with NP, and after the removal of NP. Comparison of the RNA structures before the addition of NP and after its removal reveals that NP, while introducing limited changes, remodels local structures in both vRNAs and long-range interactions in the NS vRNA, suggesting a potentially biologically relevant RNA chaperone activity. In contrast, NP significantly alters the structure of vRNAs in vRNA/NP complexes, though incorporating experimental data into RNA secondary structure prediction proved challenging. Finally, our results suggest that NP not only binds single-stranded RNA but also helices with interruptions, such as bulges or small internal loops, with a preference for G-poor and C/U-rich regions.


Sujet(s)
Virus de la grippe A , Protéines nucléocapside , Virus de la grippe A/génétique , Virus de la grippe A/métabolisme , Nucléoprotéines/métabolisme , ARN viral/métabolisme , Génomique
13.
Comput Biol Med ; 172: 108316, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38503091

RÉSUMÉ

Influenza, a pervasive viral respiratory illness, remains a significant global health concern. The influenza A virus, capable of causing pandemics, necessitates timely identification of specific subtypes for effective prevention and control, as highlighted by the World Health Organization. The genetic diversity of influenza A virus, especially in the hemagglutinin protein, presents challenges for accurate subtype prediction. This study introduces PreIS as a novel pipeline utilizing advanced protein language models and supervised data augmentation to discern subtle differences in hemagglutinin protein sequences. PreIS demonstrates two key contributions: leveraging pre-trained protein language models for influenza subtype classification and utilizing supervised data augmentation to generate additional training data without extensive annotations. The effectiveness of the pipeline has been rigorously assessed through extensive experiments, demonstrating a superior performance with an impressive accuracy of 94.54% compared to the current state-of-the-art model, the MC-NN model, which achieves an accuracy of 89.6%. PreIS also exhibits proficiency in handling unknown subtypes, emphasizing the importance of early detection. Pioneering the classification of HxNy subtypes solely based on the hemagglutinin protein chain, this research sets a benchmark for future studies. These findings promise more precise and timely influenza subtype prediction, enhancing public health preparedness against influenza outbreaks and pandemics. The data and code underlying this article are available in https://github.com/CBRC-lab/PreIS.


Sujet(s)
Virus de la grippe A , Grippe humaine , Humains , Hémagglutinines , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/métabolisme , Virus de la grippe A/génétique , Virus de la grippe A/métabolisme , Séquence d'acides aminés
14.
Arch Virol ; 169(4): 74, 2024 Mar 13.
Article de Anglais | MEDLINE | ID: mdl-38480558

RÉSUMÉ

Triple motif protein 21 (TRIM21) has an antiviral function that inhibits various viral infections. However, its role in the progress of influenza A virus (IAV) infection is unclear. In this study, we investigated the role and molecular mechanism of TRIM21 in IAV infection. RT-qPCR was used to determine the level of TRIM21 mRNA, and ELISA was used to measure the levels of IFN-α, IFN-ß, IL-6, and TNF-α. The levels of the TRIM21, NP, TBK1, IRF3, p-TBK1, and p-IRF3 proteins were estimated by Western blot. The results showed that, after IAV infection, TRIM21 was upregulated in clinical patient serum and A549 cells, and this was correlated with the IFN response. Overexpression of TRIM21 reduced IAV replication and transcription in in vitro cell experiments. TRIM21 also increased IFN-α and IFN-ß levels and decreased IL-6 and TNF-α levels in A549 cells. In addition, overexpression of TRIM21 inhibited IAV-induced apoptosis. Further experiments demonstrated that TBK1-IRF3 signaling was activated by TRIM21 and was involved in the inhibitory effect of TRIM21 on virus replication. In summary, our study suggests that TRIM21 inhibits viral replication by activating the TBK1-IRF3 signaling pathway, further inhibiting the infection process of IAV.


Sujet(s)
Sous-type H1N1 du virus de la grippe A , Virus de la grippe A , Grippe humaine , Humains , Cellules A549 , Inflammation , Virus de la grippe A/métabolisme , Sous-type H1N1 du virus de la grippe A/métabolisme , Grippe humaine/génétique , Facteur-3 de régulation d'interféron/génétique , Facteur-3 de régulation d'interféron/métabolisme , Interféron alpha/métabolisme , Interleukine-6/génétique , Interleukine-6/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Transduction du signal/physiologie , Facteur de nécrose tumorale alpha/métabolisme
15.
J Immunol ; 212(9): 1479-1492, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38477617

RÉSUMÉ

During avian influenza virus (AIV) infection, host defensive proteins promote antiviral innate immunity or antagonize viral components to limit viral replication. UFM1-specific ligase 1 (UFL1) is involved in regulating innate immunity and DNA virus replication in mammals, but the molecular mechanism by which chicken (ch)UFL1 regulates AIV replication is unclear. In this study, we first identified chUFL1 as a negative regulator of AIV replication by enhancing innate immunity and disrupting the assembly of the viral polymerase complex. Mechanistically, chUFL1 interacted with chicken stimulator of IFN genes (chSTING) and contributed to chSTING dimerization and the formation of the STING-TBK1-IRF7 complex. We further demonstrated that chUFL1 promoted K63-linked polyubiquitination of chSTING at K308 to facilitate chSTING-mediated type I IFN production independent of UFMylation. Additionally, chUFL1 expression was upregulated in response to AIV infection. Importantly, chUFL1 also interacted with the AIV PA protein to inhibit viral polymerase activity. Furthermore, chUFL1 impeded the nuclear import of the AIV PA protein and the assembly of the viral polymerase complex to suppress AIV replication. Collectively, these findings demonstrate that chUFL1 restricts AIV replication by disrupting the viral polymerase complex and facilitating type I IFN production, which provides new insights into the regulation of AIV replication in chickens.


Sujet(s)
Virus de la grippe A , Grippe chez les oiseaux , Interféron de type I , Ubiquitin-protein ligases , Réplication virale , Animaux , Poulets/génétique , Immunité innée , Virus de la grippe A/métabolisme , Virus de la grippe A/physiologie , Grippe chez les oiseaux/métabolisme , Nucleotidyltransferases , Réplication virale/génétique , Ubiquitin-protein ligases/métabolisme
16.
Emerg Microbes Infect ; 13(1): 2332652, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38517705

RÉSUMÉ

A diverse population of avian influenza A viruses (AIVs) are maintained in wild birds and ducks yet the zoonotic potential of AIVs in these environmental reservoirs and the host-virus interactions involved in mammalian infection are not well understood. In studies of a group of subtype H1N1 AIVs isolated from migratory wild birds during surveillance in North America, we previously identified eight amino acids in the polymerase genes PB2 and PB1 that were important for the transmissibility of these AIVs in a ferret model of human influenza virus transmission. In this current study we found that PB2 containing amino acids associated with transmissibility at 67, 152, 199, 508, and 649 and PB1 at 298, 642, and 667 were associated with more rapid viral replication kinetics, greater infectivity, more active polymerase complexes and greater kinetics of viral genome replication and transcription. Pathogenicity in the mouse model was also impacted, evident as greater weight loss and lung pathology associated with greater inflammatory lung cytokine expression. Further, these AIVs all contained the avian-type amino acids of PB2-E627, D701, G590, Q591 and T271. Therefore, our study provides novel insights into the role of the AIV polymerase complex in the zoonotic transmission of AIVs in mammals.


Sujet(s)
Sous-type H1N1 du virus de la grippe A , Virus de la grippe A , Grippe chez les oiseaux , Souris , Animaux , Humains , Sous-type H1N1 du virus de la grippe A/génétique , Acides aminés/génétique , Interactions hôte-microbes , Protéines virales/génétique , Protéines virales/métabolisme , Furets , Virus de la grippe A/métabolisme , Oiseaux , Nucleotidyltransferases , Réplication virale/génétique , Phylogenèse
17.
Bioorg Med Chem Lett ; 101: 129672, 2024 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-38387691

RÉSUMÉ

Influenza and COVID-19 continue to pose global threats to public health. Classic antiviral drugs have certain limitations, coupled with frequent viral mutations leading to many drugs being ineffective, the development of new antiviral drugs is urgent. Meanwhile, the invasion of influenza virus can cause an immune response, and an excessive immune response can generate a large number of inflammatory storms, leading to tissue damage. Toll-like receptor 3 (TLR3) recognizes virus dsRNA to ignite the innate immune response, and inhibit TLR3 can block the excess immune response and protect the host tissues. Taking TLR3 as the target, SMU-CX1 was obtained as the specific TLR3 inhibitor by high-throughput screening of 15,700 compounds with IC50 value of 0.11 µM. Its anti-influenza A virus activity with IC50 ranged from 0.14 to 0.33 µM against multiple subtypes of influenza A virus and also showed promising anti-SARS-CoV-2 activity with IC50 at 0.43 µM. Primary antiviral mechanism study indicated that SMU-CX1 significantly inhibited PB2 and NP protein of viruses, it can also inhibit inflammatory factors in host cells including IFN-ß, IP-10 and CCL-5. In conclusion, this study demonstrates the potential of SMU-CX1 in inhibiting IAV and SARS-CoV-2 activity, thereby offering a novel approach for designing antiviral drugs against highly pathogenic viruses.


Sujet(s)
COVID-19 , Ellipticines , Virus de la grippe A , Humains , Virus de la grippe A/métabolisme , SARS-CoV-2/métabolisme , Récepteur de type Toll-3/génétique , Récepteur de type Toll-3/métabolisme , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique
18.
Virol J ; 21(1): 33, 2024 01 29.
Article de Anglais | MEDLINE | ID: mdl-38287375

RÉSUMÉ

BACKGROUND: Influenza A virus (IAV) can cause severe and life-threatening illness in humans and animals. Therefore, it is important to search for host antiviral proteins and elucidate their antiviral mechanisms for the development of potential treatments. As a part of human innate immunity, host restriction factors can inhibit the replication of viruses, among which SAM and HD domain containing deoxynucleoside triphosphate triphosphohydrolase 1 (SAMHD1) can restrict the replication of viruses, such as HIV and enterovirus EV71. Viruses also developed countermeasures in the arms race with their hosts. There are few reports about whether SAMHD1 has a restriction effect on IAV. METHODS: To investigate the impact of IAV infection on SAMHD1 expression in A549 cells, we infected A549 cells with a varying multiplicity of infection (MOI) of IAV and collected cell samples at different time points for WB and RT-qPCR analysis to detect viral protein and SAMHD1 levels. The virus replication level in the cell culture supernatant was determined using TCID50 assay. Luciferase assay was used to reveal that H5N1 virus polymerase acidic protein (PA) affected the activity of the SAMHD1 promoter. To assess the antiviral capacity of SAMHD1, we generated a knockdown and overexpressed cell line for detecting H5N1 replication. RESULTS: In this study, we observed that SAMHD1 can restrict the intracellular replication of H5N1 and that the H5N1 viral protein PA can downregulate the expression of SAMHD1 by affecting SAMHD1 transcriptional promoter activity. We also found that SAMHD1's ability to restrict H5N1 is related to phosphorylation at 592-tyrosine. CONCLUSIONS: In conclusion, we found that SAMHD1 may affect the replication of IAVs as a host restriction factor and be countered by PA. Furthermore, SAMHD1 may be a potential target for developing antiviral drugs.


Sujet(s)
Sous-type H5N1 du virus de la grippe A , Virus de la grippe A , Grippe humaine , Animaux , Humains , Virus de la grippe A/métabolisme , Facteurs de transcription/métabolisme , Protéine-1 contenant un domaine SAM et un domaine HD/métabolisme , Réplication virale , Protéines virales/métabolisme , Antiviraux/pharmacologie , Antiviraux/métabolisme , Facteur-3 de régulation d'interféron/métabolisme
19.
Microsc Res Tech ; 87(6): 1131-1145, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38270267

RÉSUMÉ

The nuclear export protein of the influenza A virus (NEP) is involved in many important processes of the virus life cycle. This makes it an attractive target for the treatment of a disease caused by a virus. Previously it has been shown, that recombinant variants of NEP are highly prone to aggregation in solution under various conditions with the formation of amyloid-like aggregates. In the present work, the amyloid nature of NEP aggregates was evidenced by Congo red binding assays. Atomic force microscopy has shown that NEP can form two types of spherical nanoparticles, which provide an alternative pathway for the formation of amyloid-like fibrils. Type I of these "fibrillogenic" spheres, formed under physiological conditions, represents the micelle-like particles with height 10-60 nm, which can generate worm-like flexible fibrils with the diameter 2.5-4.0 nm, length 20-500 nm and the Young's modulus ~73 MPa. Type II spherical aggregates with size of about 400-1000 nm, formed at elevated temperatures, includes fractions of drop-like and vesicle-like particles, generating more rigid amyloid-like fibrils with height of ~8 nm, and length of up to 2 µm. The hypothetical mechanism of fibril formation via nanospherical structures was suggested. RESEARCH HIGHLIGHTS: AFM has revealed two types of the influenza A virus nuclear export protein spherical aggregates. They provide an alternative pathway for the formation of amyloid-like fibrils. The mechanism of fibril formation via spherical structures is suggested.


Sujet(s)
Virus de la grippe A , Protéines nucléaires , Transport nucléaire actif , Virus de la grippe A/métabolisme , Microscopie à force atomique , Amyloïde/métabolisme
20.
ACS Infect Dis ; 10(2): 650-661, 2024 02 09.
Article de Anglais | MEDLINE | ID: mdl-38173147

RÉSUMÉ

Sulfated N-glycans play a crucial role in the interaction between influenza A virus (IAV) and its host. These glycans have been found to enhance viral replication, highlighting their significance in IAV propagation. This study investigated the expression of acidic N-glycans, specifically sulfated and phosphorylated glycans, in the egg whites of 72 avian species belonging to the Order Anseriformes (waterfowls). We used the glycoblotting-based sulphoglycomics approach to elucidate the diversity of acidic N-glycans and infer their potential role in protecting embryos from infections. Family-specific variations in sulfated and phosphorylated N-glycan profiles were identified in waterfowl egg whites. Different waterfowl species exhibited distinct expressions of sulfated trans-Gal(+) and trans-Gal(-) N-glycan structures. Additionally, species-specific expression of phosphorylated N-glycans was observed. Furthermore, it was found that waterfowl species with high avian influenza virus (AIV) prevalence displayed a higher abundance of phosphorylated hybrid and high-mannose N-glycans on their egg whites. These findings shed light on the importance of phosphorylated and sulfated N-glycans in understanding the role of acidic glycans in IAV propagation.


Sujet(s)
Virus de la grippe A , Animaux , Prévalence , Virus de la grippe A/métabolisme , Polyosides
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE