Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 57
Filtrer
1.
J Virol ; 97(12): e0173723, 2023 Dec 21.
Article de Anglais | MEDLINE | ID: mdl-38051260

RÉSUMÉ

IMPORTANCE: Viruses are constantly evolving to promote propagation in the host. Here, we show that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) utilizes host RAD51 for replication. Silencing of RAD51 impaired SARS-CoV-2 propagation. Viral RNA colocalized with RAD51 in the cytoplasm of SARS-CoV-2-infected cells, suggesting that both viral RNA and RAD51 may form a replication complex. We, therefore, evaluated RAD51 inhibitors as possible therapeutic agents against SARS-CoV-2. Indeed, RAD51 inhibitors exerted antiviral activities against not only Wuhan but also variants of SARS-CoV-2. Molecular docking model shows that RAD51 inhibitors impede SARS-CoV-2 propagation by interfering with dimerization of RAD51. These data suggest that RAD51 may represent a novel host-based drug target for coronavirus disease 2019 treatment.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , COVID-19/métabolisme , COVID-19/virologie , Simulation de docking moléculaire , Rad51 Recombinase/antagonistes et inhibiteurs , Rad51 Recombinase/métabolisme , ARN viral , SARS-CoV-2/physiologie , Interactions hôte-pathogène
2.
Sci Rep ; 13(1): 21489, 2023 12 06.
Article de Anglais | MEDLINE | ID: mdl-38057373

RÉSUMÉ

Natural products and herbal medicine have been widely used in drug discovery for treating infectious diseases. Recent outbreak of COVID-19 requires various therapeutic strategies. Here, we used YSK-A, a mixture of three herbal components Boswellia serrata, Commiphora myrrha, and propolis, to evaluate potential antiviral activity against SARS-CoV-2. We showed that YSK-A inhibited SARS-CoV-2 propagation with an IC50 values of 12.5 µg/ml and 15.42 µg/ml in Vero E6 and Calu-3 cells, respectively. Using transcriptome analysis, we further demonstrated that YSK-A modulated various host gene expressions in Calu-3 cells. Among these, we selected 9 antiviral- or immune-related host genes for further study. By siRNA-mediated knockdown experiment, we verified that MUC5AC, LIF, CEACAM1, and GDF15 host genes were involved in antiviral activity of YSK-A. Therefore, silencing of these genes nullified YSK-A-mediated inhibition of SARS-CoV-2 propagation. These data indicate that YSK-A displays an anti-SARS-CoV-2 activity by targeting multiple antiviral genes. Although the exact antiviral mechanism of each constituent has not been verified yet, our data indicate that YSK-A has an immunomodulatory effect on SARS-CoV-2 and thus it may represent a novel natural product-derived therapeutic agent for treating COVID-19.


Sujet(s)
Produits biologiques , COVID-19 , Plantes médicinales , Chlorocebus aethiops , Animaux , SARS-CoV-2 , Produits biologiques/pharmacologie , Antiviraux/pharmacologie , Cellules Vero
3.
J Comput Aided Mol Des ; 37(9): 453-461, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37452977

RÉSUMÉ

African swine fever virus (ASFV), an extremely contagious virus with high mortality rates, causes severe hemorrhagic viral disease in both domestic and wild pigs. Fortunately, ASFV cannot be transmitted from pigs to humans. However, ongoing ASFV outbreaks could have severe economic consequences for global food security. Although ASFV was discovered several years ago, no vaccines or treatments are commercially available yet; therefore, the identification of new anti-ASFV drugs is urgently warranted. Using molecular docking and machine learning, we have previously identified pentagastrin, cangrelor, and fostamatinib as potential antiviral drugs against ASFV. Here, using machine learning combined with docking simulations, we identified natural products with a high affinity for AsfvPolX proteins. We selected five natural products (NPs) that are located close in chemical space to the six known natural flavonoids that possess anti-ASFV activity. Polygalic acid markedly reduced AsfvPolX polymerase activity in a dose-dependent manner. We propose an efficient protocol for identifying NPs as potential antiviral drugs by identifying chemical spaces containing high-affinity binders against ASFV in NP databases.


Sujet(s)
Virus de la peste porcine africaine , Peste porcine africaine , Humains , Suidae , Animaux , Peste porcine africaine/prévention et contrôle , Simulation de docking moléculaire , Protéines virales , Antiviraux/pharmacologie , Techniques in vitro , Apprentissage machine
4.
Microbiol Spectr ; 11(3): e0510522, 2023 06 15.
Article de Anglais | MEDLINE | ID: mdl-36995225

RÉSUMÉ

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19). SARS-CoV-2 propagation is mediated by the protein interaction between viral proteins and host cells. Tyrosine kinase has been implicated in viral replication, and hence, it has become a target for developing antiviral drugs. We have previously reported that receptor tyrosine kinase inhibitor blocks the replication of hepatitis C virus (HCV). In the present study, we investigated two receptor tyrosine kinase-specific inhibitors, amuvatinib and imatinib, for their potential antiviral efficacies against SARS-CoV-2. Treatment with either amuvatinib or imatinib displays an effective inhibitory activity against SARS-CoV-2 propagation without an obvious cytopathic effect in Vero E6 cells. Notably, amuvatinib exerts a stronger antiviral activity than imatinib against SARS-CoV-2 infection. Amuvatinib blocks SARS-CoV-2 infection with a 50% effective concentration (EC50) value ranging from ~0.36 to 0.45 µM in Vero E6 cells. We further demonstrate that amuvatinib inhibits SARS-CoV-2 propagation in human lung Calu-3 cells. Using pseudoparticle infection assay, we verify that amuvatinib blocks SARS-CoV-2 at the entry step of the viral life cycle. More specifically, amuvatinib inhibits SARS-CoV-2 infection at the binding-attachment step. Moreover, amuvatinib exhibits highly efficient antiviral activity against emerging SARS-CoV-2 variants. Importantly, we demonstrate that amuvatinib inhibits SARS-CoV-2 infection by blocking ACE2 cleavage. Taken together, our data suggest that amuvatinib may provide a potential therapeutic agent for the treatment of COVID-19. IMPORTANCE Tyrosine kinase has been implicated in viral replication and has become an antiviral drug target. Here, we chose two well-known receptor tyrosine kinase inhibitors, amuvatinib and imatinib, and evaluated their drug potencies against SARS-CoV-2. Surprisingly, amuvatinib displays a stronger antiviral activity than imatinib against SARS-CoV-2. Amuvatinib blocks SARS-CoV-2 infection by inhibiting ACE2 cleavage and the subsequent soluble ACE2 receptor. All these data suggest that amuvatinib may be a potential therapeutic agent in SARS-CoV-2 prevention for those experiencing vaccine breakthroughs.


Sujet(s)
COVID-19 , Animaux , Humains , SARS-CoV-2 , Mésilate d'imatinib/pharmacologie , Mésilate d'imatinib/usage thérapeutique , Angiotensin-converting enzyme 2 , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , Protein-tyrosine kinases/pharmacologie , Étapes du cycle de vie
5.
Mol Cells ; 45(10): 702-717, 2022 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-35993162

RÉSUMÉ

Hepatitis C virus (HCV) infection can lead to chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. HCV employs diverse strategies to evade host antiviral innate immune responses to mediate a persistent infection. In the present study, we show that nonstructural protein 5A (NS5A) interacts with an NF-κB inhibitor immunomodulatory kinase, IKKε, and subsequently downregulats beta interferon (IFN-ß) promoter activity. We further demonstrate that NS5A inhibits DDX3-mediated IKKε and interferon regulatory factor 3 (IRF3) phosphorylation. We also note that hyperphosphorylation of NS5A mediats protein interplay between NS5A and IKKε, thereby contributing to NS5A-mediated modulation of IFN-ß signaling. Lastly, NS5A inhibits IKKε-dependent p65 phosphorylation and NF-κB activation. Based on these findings, we propose NS5A as a novel regulator of IFN signaling events, specifically by inhibiting IKKε downstream signaling cascades through its interaction with IKKε. Taken together, these data suggest an additional mechanistic means by which HCV modulates host antiviral innate immune responses to promote persistent viral infection.


Sujet(s)
Hépatite C , Protéines virales non structurales , Antiviraux , Hepacivirus/métabolisme , Humains , I-kappa B Kinase/métabolisme , Immunité innée , Facteur-3 de régulation d'interféron/métabolisme , Interféron bêta/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Protéines virales non structurales/métabolisme
6.
Mol Ther ; 30(5): 1994-2004, 2022 05 04.
Article de Anglais | MEDLINE | ID: mdl-35007757

RÉSUMÉ

Adeno-associated virus (AAV)-mediated gene delivery holds great promise for gene therapy. However, the non-invasive delivery of AAV for lung tissues has not been adequately established. Here, we revealed that the intratracheal administration of an appropriate amount of AAV2/8 predominantly targets lung tissue. AAV-mediated gene delivery that we used in this study induced the expression of the desired protein in lung parenchymal cells, including alveolar type II cells. We harnessed the technique to develop severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-susceptible mice. Three kinds of immune function-relevant gene knockout (KO) mice were transduced with AAV encoding human angiotensin-converting enzyme 2 (hACE2) and then injected with SARS-CoV-2. Among these mice, type I interferon receptor (IFNAR) KO mice showed increased viral titer in the lungs compared to that in the other KO mice. Moreover, nucleocapsid protein of SARS-CoV-2 and multiple lesions in the trachea and lung were observed in AAV-hACE2-transduced, SARS-CoV-2-infected IFNAR KO mice, indicating the involvement of type I interferon signaling in the protection of SARS-CoV-2. In this study, we demonstrate the ease and rapidness of the intratracheal administration of AAV for targeting lung tissue in mice, and this can be used to study diverse pulmonary diseases.


Sujet(s)
COVID-19 , SARS-CoV-2 , Animaux , COVID-19/thérapie , Dependovirus/génétique , Modèles animaux de maladie humaine , Prédisposition aux maladies , Poumon/anatomopathologie , Souris , Souris transgéniques , SARS-CoV-2/génétique
7.
Mol Cells ; 45(3): 148-157, 2022 Mar 31.
Article de Anglais | MEDLINE | ID: mdl-34949741

RÉSUMÉ

Hepatitis C virus (HCV) is a major cause of chronic liver disease and is highly dependent on cellular proteins for viral propagation. Using protein microarray analysis, we identified 90 cellular proteins as HCV nonstructural 5A (NS5A) interacting partners, and selected telomere length regulation protein (TEN1) for further study. TEN1 forms a heterotrimeric complex with CTC and STN1, which is essential for telomere protection and maintenance. Telomere length decreases in patients with active HCV, chronic liver disease, and hepatocellular carcinoma. However, the molecular mechanism of telomere length shortening in HCV-associated disease is largely unknown. In the present study, protein interactions between NS5A and TEN1 were confirmed by immunoprecipitation assays. Silencing of TEN1 reduced both viral RNA and protein expression levels of HCV, while ectopic expression of the siRNA-resistant TEN1 recovered the viral protein level, suggesting that TEN1 was specifically required for HCV propagation. Importantly, we found that TEN1 is re-localized from the nucleus to the cytoplasm in HCV-infected cells. These data suggest that HCV exploits TEN1 to promote viral propagation and that telomere protection is compromised in HCV-infected cells. Overall, our findings provide mechanistic insight into the telomere shortening in HCV-infected cells.


Sujet(s)
Hepacivirus , Hépatite C , Hepacivirus/physiologie , Hépatite C/génétique , Humains , ARN viral , Télomère/génétique , Raccourcissement des télomères
8.
Int J Mol Sci ; 22(24)2021 Dec 14.
Article de Anglais | MEDLINE | ID: mdl-34948216

RÉSUMÉ

African swine fever virus (ASFV) is a highly contagious virus that causes severe hemorrhagic viral disease resulting in high mortality in domestic and wild pigs, until few antiviral agents can inhibit ASFV infections. Thus, new anti-ASFV drugs need to be urgently identified. Recently, we identified pentagastrin as a potential antiviral drug against ASFVs using molecular docking and machine learning models. However, the scoring functions are easily influenced by properties of protein pockets, resulting in a scoring bias. Here, we employed the 5'-P binding pocket of AsfvPolX as a potential binding site to identify antiviral drugs and classified 13 AsfvPolX structures into three classes based on pocket parameters calculated by the SiteMap module. We then applied principal component analysis to eliminate this scoring bias, which was effective in making the SP Glide score more balanced between 13 AsfvPolX structures in the dataset. As a result, we identified cangrelor and fostamatinib as potential antiviral drugs against ASFVs. Furthermore, the classification of the pocket properties of AsfvPolX protein can provide an alternative approach to identify novel antiviral drugs by optimizing the scoring function of the docking programs. Here, we report a machine learning-based novel approach to generate high binding affinity compounds that are individually matched to the available classification of the pocket properties of AsfvPolX protein.


Sujet(s)
Virus de la peste porcine africaine/effets des médicaments et des substances chimiques , Peste porcine africaine/traitement médicamenteux , Antiviraux/pharmacologie , Virus de la peste porcine africaine/métabolisme , Animaux , Sites de fixation/effets des médicaments et des substances chimiques , Apprentissage machine , Suidae , Protéines virales/métabolisme , Réplication virale/effets des médicaments et des substances chimiques
9.
Mol Cells ; 44(9): 688-695, 2021 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-34518443

RÉSUMÉ

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has become a global health concern. Various SARS-CoV-2 vaccines have been developed and are being used for vaccination worldwide. However, no therapeutic agents against coronavirus disease 2019 (COVID-19) have been developed so far; therefore, new therapeutic agents are urgently needed. In the present study, we evaluated several hepatitis C virus direct-acting antivirals as potential candidates for drug repurposing against COVID-19. Theses include asunaprevir (a protease inhibitor), daclatasvir (an NS5A inhibitor), and sofosbuvir (an RNA polymerase inhibitor). We found that asunaprevir, but not sofosbuvir and daclatasvir, markedly inhibited SARS-CoV-2-induced cytopathic effects in Vero E6 cells. Both RNA and protein levels of SARS-CoV-2 were significantly decreased by treatment with asunaprevir. Moreover, asunaprevir profoundly decreased virion release from SARS-CoV-2-infected cells. A pseudoparticle entry assay revealed that asunaprevir blocked SARS-CoV-2 infection at the binding step of the viral life cycle. Furthermore, asunaprevir inhibited SARS-CoV-2 propagation in human lung Calu-3 cells. Collectively, we found that asunaprevir displays broad-spectrum antiviral activity and therefore might be worth developing as a new drug repurposing candidate for COVID-19.


Sujet(s)
Antiviraux/pharmacologie , Traitements médicamenteux de la COVID-19 , Isoquinoléines/pharmacologie , SARS-CoV-2/croissance et développement , Sulfonamides/pharmacologie , Inhibiteurs de protéases virales/pharmacologie , Réplication virale/effets des médicaments et des substances chimiques , Animaux , Carbamates/pharmacologie , Lignée cellulaire , Chlorocebus aethiops , DNA-directed RNA polymerases/antagonistes et inhibiteurs , Cellules HEK293 , Hepacivirus/effets des médicaments et des substances chimiques , Humains , Imidazoles/pharmacologie , Pyrrolidines/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Sofosbuvir/pharmacologie , Valine/analogues et dérivés , Valine/pharmacologie , Cellules Vero , Protéines virales non structurales/antagonistes et inhibiteurs
10.
Molecules ; 26(12)2021 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-34208385

RÉSUMÉ

African swine fever virus (ASFV) causes a highly contagious and severe hemorrhagic viral disease with high mortality in domestic pigs of all ages. Although the virus is harmless to humans, the ongoing ASFV epidemic could have severe economic consequences for global food security. Recent studies have found a few antiviral agents that can inhibit ASFV infections. However, currently, there are no vaccines or antiviral drugs. Hence, there is an urgent need to identify new drugs to treat ASFV. Based on the structural information data on the targets of ASFV, we used molecular docking and machine learning models to identify novel antiviral agents. We confirmed that compounds with high affinity present in the region of interest belonged to subsets in the chemical space using principal component analysis and k-means clustering in molecular docking studies of FDA-approved drugs. These methods predicted pentagastrin as a potential antiviral drug against ASFVs. Finally, it was also observed that the compound had an inhibitory effect on AsfvPolX activity. Results from the present study suggest that molecular docking and machine learning models can play an important role in identifying potential antiviral drugs against ASFVs.


Sujet(s)
Virus de la peste porcine africaine/effets des médicaments et des substances chimiques , Peste porcine africaine/traitement médicamenteux , Antiviraux/composition chimique , Antiviraux/pharmacologie , Apprentissage machine/normes , Peste porcine africaine/immunologie , Peste porcine africaine/virologie , Virus de la peste porcine africaine/immunologie , Virus de la peste porcine africaine/isolement et purification , Séquence d'acides aminés , Animaux , DNA-directed DNA polymerase/composition chimique , DNA-directed DNA polymerase/métabolisme , Conception de médicament , Simulation de docking moléculaire , Pentagastrine/composition chimique , Pentagastrine/pharmacologie , Suidae , Protéines virales/composition chimique , Protéines virales/métabolisme
11.
J Microbiol ; 59(1): 101-109, 2021 Jan.
Article de Anglais | MEDLINE | ID: mdl-33355889

RÉSUMÉ

Hepatitis C virus (HCV) life cycle is highly dependent on cellular proteins for viral propagation. In order to identify the cellular factors involved in HCV propagation, we previously performed a protein microarray assay using the HCV nonstructural 5A (NS5A) protein as a probe. Of ∼9,000 human cellular proteins immobilized in a microarray, adenosylhomocysteinase like 1 (AHCYL1) was among 90 proteins identified as NS5A interactors. Of these candidates, AHCYL1 was selected for further study. In the present study, we verified the physical interaction between NS5A and AHCYL1 by both in vitro pulldown and coimmunoprecipitation assays. Furthermore, HCV NS5A interacted with endogenous AHCYL1 in Jc1-infected cells. Both NS5A and AHCYL1 were colocalized in the cytoplasmic region in HCV-replicating cells. siRNAmediated knockdown of AHCYL1 abrogated HCV propagation. Exogenous expression of the siRNA-resistant AHCYL1 mutant, but not of the wild-type AHCYL1, restored HCV protein expression levels, indicating that AHCYL1 was required specifically for HCV propagation. Importantly, AHCYL1 was involved in the HCV internal ribosome entry site-mediated translation step of the HCV life cycle. Finally, we demonstrated that the proteasomal degradation pathway of AHCYL1 was modulated by persistent HCV infection. Collectively, these data suggest that HCV may modulate the AHCYL1 protein to promote viral propagation.


Sujet(s)
Hepacivirus/métabolisme , Hépatite C/enzymologie , Protéines virales non structurales/métabolisme , Hepacivirus/génétique , Hepacivirus/croissance et développement , Hépatite C/génétique , Hépatite C/virologie , Interactions hôte-pathogène , Humains , Liaison aux protéines , Protéines virales non structurales/génétique
12.
J Virol ; 94(19)2020 09 15.
Article de Anglais | MEDLINE | ID: mdl-32727880

RÉSUMÉ

Hepatitis C virus (HCV) exploits cellular proteins to facilitate viral propagation. To identify the cellular factors involved in the HCV life cycle, we previously performed protein microarray assays using either HCV nonstructural 5A (NS5A) protein or core protein as a probe. Interestingly, cellular cortactin strongly interacted with both NS5A and core. Cortactin is an actin-binding protein critically involved in tumor progression by regulating the migration and invasion of cancerous cells. Protein interaction between cortactin and NS5A or core was confirmed by coimmunoprecipitation and immunofluorescence assays. We showed that cortactin interacted with NS5A and core via the N-terminal acidic domain of cortactin. Cortactin expression levels were not altered by HCV infection. Small interfering RNA (siRNA)-mediated knockdown of cortactin dramatically decreased HCV protein expression and infectivity levels, whereas overexpression of cortactin increased viral propagation. Ectopic expression of the siRNA-resistant cortactin recovered the viral infectivity, suggesting that cortactin was specifically required for HCV propagation. We further showed that cortactin was involved in the assembly step without affecting viral entry, HCV internal ribosome entry site (IRES)-mediated translation, and the replication steps of the HCV life cycle. Of note, silencing of cortactin markedly reduced both NS5A and core protein levels on the lipid droplets (LDs), and this effect was reversed by the overexpression of cortactin. Importantly, NS5A and core promoted cell migration by activating the phosphorylation of cortactin at tyrosine residues 421 and 466. Taken together, these data suggest that cortactin is not only involved in HCV assembly but also plays an important role in the cell migration.IMPORTANCE Cortactin is a cytoskeletal protein that regulates cell migration in response to a number of extracellular stimuli. The functional involvement of cortactin in the virus life cycle is not yet fully understood. The most significant finding is that cortactin strongly interacted with both hepatitis C virus (HCV) core and NS5A. Cortactin is involved in HCV assembly by tethering core and NS5A on the lipid droplets (LDs) with no effect on LD biogenesis. It was noteworthy that HCV NS5A and core activated cortactin by phosphorylation at tyrosines 421 and 466 to regulate cell migration. Collectively, our study shows that cortactin is a novel host factor involved in viral production and HCV-associated pathogenesis.


Sujet(s)
Cortactine/métabolisme , Hepacivirus/physiologie , Protéines virales non structurales/métabolisme , Virion/physiologie , Assemblage viral/physiologie , Lignée cellulaire , Protéines du cytosquelette/métabolisme , Cellules HEK293 , Hépatite C/virologie , Antigènes de l'hépatite C/métabolisme , Humains , Immunoprécipitation , Phosphorylation , Petit ARN interférent/génétique , Pénétration virale , Réplication virale
13.
Mol Cells ; 43(5): 469-478, 2020 May 31.
Article de Anglais | MEDLINE | ID: mdl-32344996

RÉSUMÉ

Hepatitis C virus (HCV) propagation is highly dependent on cellular proteins. To identify the host factors involved in HCV propagation, we previously performed protein microarray assays and identified the LIM and SH3 domain protein 1 (LASP-1) as an HCV NS5A-interacting partner. LASP-1 plays an important role in the regulation of cell proliferation, migration, and protein-protein interactions. Alteration of LASP-1 expression has been implicated in hepatocellular carcinoma. However, the functional involvement of LASP1 in HCV propagation and HCV-induced pathogenesis has not been elucidated. Here, we first verified the protein interaction of NS5A and LASP-1 by both in vitro pulldown and coimmunoprecipitation assays. We further showed that NS5A and LASP-1 were colocalized in the cytoplasm of HCV infected cells. NS5A interacted with LASP-1 through the proline motif in domain I of NS5A and the tryptophan residue in the SH3 domain of LASP-1. Knockdown of LASP-1 increased HCV replication in both HCV-infected cells and HCV subgenomic replicon cells. LASP-1 negatively regulated viral propagation and thereby overexpression of LASP-1 decreased HCV replication. Moreover, HCV propagation was decreased by wild-type LASP-1 but not by an NS5A binding-defective mutant of LASP-1. We further demonstrated that LASP-1 was involved in the replication stage of the HCV life cycle. Importantly, LASP-1 expression levels were increased in persistently infected cells with HCV. These data suggest that HCV modulates LASP-1 via NS5A in order to regulate virion levels and maintain a persistent infection.


Sujet(s)
Protéines du cytosquelette/métabolisme , Hepacivirus/physiologie , Hépatite C/virologie , Protéines à homéodomaine/métabolisme , Protéines à domaine LIM/métabolisme , Protéines virales non structurales/métabolisme , Protéines du cytosquelette/génétique , Régulation de l'expression des gènes , Techniques de knock-down de gènes , Cellules HEK293 , Hépatite C/transmission , Protéines à homéodomaine/génétique , Humains , Protéines à domaine LIM/génétique , Analyse par réseau de protéines , Liaison aux protéines , Domaines protéiques/génétique , Protéines virales non structurales/génétique , Réplication virale
14.
J Virol ; 94(5)2020 02 14.
Article de Anglais | MEDLINE | ID: mdl-31801866

RÉSUMÉ

Hepatitis C virus (HCV) is a major etiologic agent of chronic liver diseases. HCV is highly dependent on cellular machinery for viral propagation. Using protein microarray analysis, we previously identified 90 cellular proteins as nonstructural 5A (NS5A) interacting partners. Of these, protein kinase C and casein kinase substrate in neurons protein 2 (PACSIN2) was selected for further study. PACSIN2 belongs to the PACSIN family, which is involved in the formation of caveolae. Protein interaction between NS5A and PACSIN2 was confirmed by pulldown assay and further verified by both coimmunoprecipitation and immunofluorescence assays. We showed that PACSIN2 interacted with domain I of NS5A and the Fer-CIP4 homology (FCH)-Bin/amphiphysin/Rvs (F-BAR) region of PACSIN2. Interestingly, NS5A specifically attenuated protein kinase C alpha (PKCα)-mediated phosphorylation of PACSIN2 at serine 313 by interrupting PACSIN2 and PKCα interaction. In fact, mutation of the serine 313 to alanine (S313A) of PACSIN2 increased protein interaction with NS5A. Silencing of PACSIN2 decreased both viral RNA and protein expression levels of HCV. Ectopic expression of the small interfering RNA (siRNA)-resistant PACSIN2 recovered the viral infectivity, suggesting that PACSIN2 was specifically required for HCV propagation. PACSIN2 was involved in viral assembly without affecting other steps of the HCV life cycle. Indeed, overexpression of PACSIN2 promoted NS5A and core protein (core) interaction. We further showed that inhibition of PKCα increased NS5A and core interaction, suggesting that phosphorylation of PACSIN2 might influence HCV assembly. Moreover, PACSIN2 was required for lipid droplet formation via modulating extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation. Taken together, these data indicate that HCV modulates PACSIN2 via NS5A to promote virion assembly.IMPORTANCE PACSIN2 is a lipid-binding protein that triggers the tubulation of the phosphatidic acid-containing membranes. The functional involvement of PACSIN2 in the virus life cycle has not yet been demonstrated. We showed that phosphorylation of PACSIN2 displayed a negative effect on NS5A and core interaction. The most significant finding is that NS5A prevents PKCα from binding to PACSIN2. Therefore, the phosphorylation level of PACSIN2 is decreased in HCV-infected cells. We showed that HCV NS5A interrupted PKCα-mediated PACSIN2 phosphorylation at serine 313, thereby promoting NS5A-PACSIN2 interaction. We further demonstrated that PACSIN2 modulated lipid droplet formation through ERK1/2 phosphorylation. These data provide evidence that PACSIN2 is a proviral cellular factor required for viral propagation.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Hepacivirus/physiologie , Motifs et domaines d'intéraction protéique , Protéines virales non structurales/métabolisme , Assemblage viral/physiologie , Protéines adaptatrices de la transduction du signal/génétique , Lignée cellulaire , Régulation de l'expression des gènes viraux , Hépatite C/virologie , Humains , Immunoprécipitation , Phosphorylation , Maturation post-traductionnelle des protéines , Petit ARN interférent , ARN viral/métabolisme , Réplication virale/physiologie
15.
Sci Rep ; 9(1): 7288, 2019 05 13.
Article de Anglais | MEDLINE | ID: mdl-31086268

RÉSUMÉ

Hepatitis C virus (HCV) is the major causative agent of chronic liver diseases, including liver cirrhosis and hepatocellular carcinoma. The recent development of highly effective direct-acting antivirals (DAAs) has revolutionized the treatment of HCV patients. However, these DAAs are exorbitantly expensive for the majority of HCV patients worldwide. Moreover, these drugs still show genotypic difference in cure rate and have some resistant-associated variants. Tylophorine, a natural compound derived from Tylophora indica plants, is known to have anti-inflammatory and anti-cancerous growth activities. In the present study, we showed that two tylophorine intermediates, 5-Oxo-1-[(2,3,6,7-tetramethoxy-9-phenanthrenyl) methyl]-L-proline (O859585) and 2,3,6,7-tetramethoxy-9-phenanthrenecarboxylic acid (T298875), displayed anti-HCV activity with an EC50 of 38.25 µM for T298875 and 29.11~35.3 µM for O859585 in various HCV genotypes. We demonstrated that O859585 efficiently blocked HCV attachment by neutralizing free viral particles without affecting other stages of the HCV life cycle and interferon stimulation. O859585 interrupted binding between HCV E2 and CD81. Of note, co-treatment of O859585 with either interferon alpha (IFNα) or sofosbuvir exerted either an additive or synergistic antiviral activity in HCV-infected cells with no measurable effect on cell viability. Most importantly, O859585 in combination with IFNα and sofosbuvir exhibited synergistic effects on anti-HCV activity in primary human hepatocytes. Collectively, these data suggest that O859585 may be a novel antiviral agent for HCV therapy.


Sujet(s)
Alcaloïdes/pharmacologie , Antiviraux/pharmacologie , Hepacivirus/effets des médicaments et des substances chimiques , Hépatite C chronique/traitement médicamenteux , Indolizine/pharmacologie , Phénanthrènes/pharmacologie , Proline/pharmacologie , Pénétration virale/effets des médicaments et des substances chimiques , Alcaloïdes/composition chimique , Alcaloïdes/usage thérapeutique , Antiviraux/composition chimique , Antiviraux/usage thérapeutique , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Association de médicaments , Cellules HEK293 , Hepacivirus/métabolisme , Hépatite C chronique/virologie , Hépatocytes/métabolisme , Hépatocytes/virologie , Humains , Indolizine/composition chimique , Indolizine/usage thérapeutique , Interféron alpha/pharmacologie , Interféron alpha/usage thérapeutique , Phénanthrènes/composition chimique , Phénanthrènes/usage thérapeutique , Culture de cellules primaires , Proline/usage thérapeutique , Sofosbuvir/pharmacologie , Sofosbuvir/usage thérapeutique , Antigène CD81/métabolisme , Tylophora/composition chimique , Protéines de l'enveloppe virale/métabolisme
16.
J Virol ; 93(2)2019 01 15.
Article de Anglais | MEDLINE | ID: mdl-30381483

RÉSUMÉ

Hepatitis C virus (HCV) infection may cause chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. HCV exploits cellular machineries to establish persistent infection. We demonstrate here that ubiquitin-conjugating enzyme E2S (UBE2S), a member of the ubiquitin-conjugating enzyme family (E2s), was downregulated by endoplasmic reticulum stress caused by HCV in Huh7 cells. UBE2S interacted with domain I of HCV NS5A and degraded NS5A protein through the Lys11-linked proteasome-dependent pathway. Overexpression of UBE2S suppressed viral propagation, while depletion of UBE2S expression increased viral infectivity. Enzymatically inactive UBE2S C95A mutant exerted no antiviral activity, suggesting that ubiquitin-conjugating enzymatic activity was required for the suppressive role of UBE2S. Chromatin ubiquitination plays a crucial role in the DNA damage response. We showed that the levels of UBE2S and Lys11 chains bound to the chromatin were markedly decreased in the context of HCV replication, rendering HCV-infected cells more sensitive to DNA damage. These data suggest that HCV counteracts antiviral activity of UBE2S to optimize viral propagation and may contribute to HCV-induced liver pathogenesis.IMPORTANCE Protein homeostasis is essential to normal cell function. HCV infection disturbs the protein homeostasis in the host cells. Therefore, host cells exert an anti-HCV activity in order to maintain normal cellular metabolism. We showed that UBE2S interacted with HCV NS5A and degraded NS5A protein through the Lys11-linked proteasome-dependent pathway. However, HCV has evolved to overcome host antiviral activity. We demonstrated that the UBE2S expression level was suppressed in HCV-infected cells. Since UBE2S is an ubiquitin-conjugating enzyme and this enzyme activity is involved in DNA damage repair, HCV-infected cells are more sensitive to DNA damage, and thus UBE2S may contribute to viral oncogenesis.


Sujet(s)
Régulation négative , Hepacivirus/pathogénicité , Hépatite C/métabolisme , Ubiquitin-conjugating enzymes/métabolisme , Protéines virales non structurales/métabolisme , Lignée cellulaire , Chromatine/métabolisme , Altération de l'ADN , Réticulum endoplasmique/métabolisme , Stress du réticulum endoplasmique , Cellules HEK293 , Hepacivirus/métabolisme , Hépatite C/virologie , Humains , Proteasome endopeptidase complex/métabolisme , Protéolyse , Ubiquitin-conjugating enzymes/composition chimique , Ubiquitination , Protéines virales non structurales/composition chimique , Réplication virale
17.
Sci Rep ; 8(1): 15486, 2018 10 19.
Article de Anglais | MEDLINE | ID: mdl-30341327

RÉSUMÉ

Hepatitis C virus (HCV) exploits an extensive network of host proteins to maintain chronic infection. Using RNA-Seq technology, we identified 30 host genes that were differentially expressed in cell culture grown HCV (HCVcc)-infected cells. Of these candidate genes, we selected solute carrier family 3 member 2 (SLC3A2) for further investigation. SLC3A2, also known as CD98hc, is a member of the solute carrier family and encodes a subunit of heterodimeric amino acid transporter. SLC3A2 and LAT1 constitute a heterodimeric transmembrane protein complex that catalyzes amino acid transport. In this study, we showed that HCV upregulated both mRNA and protein expression levels of SLC3A2 and this upregulation occurred through NS3/4A-mediated oxidative stress. HCV also elevated SLC3A2/LAT1 complex level and thus mammalian target of rapamycin complex 1 (mTORC1) signaling was activated. We further showed that L-leucine transport level was significantly increased in Jc1-infected cells as compared with mock-infected cells. Using RNA interference technology, we demonstrated that SLC3A2 was specifically required for the entry step but not for other stages of the HCV life cycle. These data suggest that SLC3A2 plays an important role in regulating HCV entry. Collectively, HCV exploits SLC3A2 for viral propagation and upregulation of SLC3A2 may contribute to HCV-mediated pathogenesis.


Sujet(s)
Chaine lourde de l'antigène CD98/métabolisme , Hepacivirus/physiologie , Hépatite C/virologie , Complexes multiprotéiques/métabolisme , Chaine lourde de l'antigène CD98/génétique , Régulation de l'expression des gènes , Cellules HEK293 , Humains , Transporteur-1 d'acides aminés neutres à longue chaîne/métabolisme , Leucine/métabolisme , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Stress oxydatif , Transport des protéines , Petit ARN interférent/génétique , Transduction du signal , Protéines virales non structurales/métabolisme , Pénétration virale , Réplication virale
18.
J Virol ; 92(11)2018 06 01.
Article de Anglais | MEDLINE | ID: mdl-29563287

RÉSUMÉ

RAD51-associated protein 1 (RAD51AP1) is a member of the multiprotein complexes postulated to carry out RAD51-mediated homologous recombination and DNA repair in mammalian cells. In the present study, we showed that hepatitis C virus (HCV) NS5A directly bound RAD51AP1 and increased the protein level of RAD51AP1 through modulation of the ubiquitin-proteasome pathway. We also demonstrated that RAD51AP1 protein levels were increased in the liver tissues of HCV-infected patients and NS5A-transgenic mice. Importantly, NS5A impaired DNA repair by disrupting the RAD51/RAD51AP1/UAF1 complex and rendered HCV-infected cells more sensitive to DNA damage. Silencing of RAD51AP1 expression resulted in a decrease of viral propagation. We further demonstrated that RAD51AP1 was involved in the assembly step of the HCV life cycle by protecting viral RNA. These data suggest that HCV exploits RAD51AP1 to promote viral propagation and thus that host DNA repair is compromised in HCV-infected cells. Overall, our findings provide mechanistic insight into the pathogenesis of HCV infection.IMPORTANCE Chronic infection with HCV is the leading cause of hepatocellular carcinoma (HCC). However, the molecular mechanisms underlying HCV-induced HCC are not fully understood. Here we demonstrate that the HCV NS5A protein physically interacts with RAD51AP1 and increases the RAD51AP1 protein level through modulation of the ubiquitin-proteasome pathway. HCV coopts host RAD51AP1 to protect viral RNA at an assembly step of the HCV life cycle. Note that the RAD51 protein accumulates in the cytoplasm of HCV-infected cells, and thus the RAD51/RAD51AP1/UAF1-mediated DNA damage repair system in the nucleus is compromised in HCV-infected cells. Our data may provide new insight into the molecular mechanisms of HCV-induced pathogenesis.


Sujet(s)
Protéines de transport/génétique , Réparation de l'ADN/génétique , Hepacivirus/pathogénicité , Protéines nucléaires/génétique , ARN viral/génétique , Rad51 Recombinase/métabolisme , Assemblage viral/génétique , Animaux , Apolipoprotéines E/génétique , Lignée cellulaire , Altération de l'ADN/génétique , Protéines de liaison à l'ADN , Hepacivirus/génétique , Humains , Souris , Souris transgéniques , Interférence par ARN , Petit ARN interférent/génétique , Protéines de liaison à l'ARN , Ubiquitination , Protéines virales non structurales/métabolisme
19.
Front Microbiol ; 8: 1249, 2017.
Article de Anglais | MEDLINE | ID: mdl-28729862

RÉSUMÉ

Hepatitis C virus (HCV) is a leading cause of chronic liver disease affecting over 170 million people worldwide. Chronic infection with HCV progresses to liver fibrosis, cirrhosis, and hepatocellular carcinoma. HCV exploits host cellular factors for viral propagation. To investigate the cellular factors required for HCV propagation, we screened a siRNA library targeting human cell cycle genes using cell culture grown HCV-infected cells. In the present study, we selected and characterized a gene encoding Rad51. Rad51, a member of a conserved recombinase family, is an essential factor for homologous recombination and repair of double-strand DNA breaks. We demonstrated that siRNA-mediated knockdown of Rad51 significantly inhibited HCV propagation without affecting HCV RNA replication. Silencing of Rad51 impaired secretion of infectious HCV particles and thus intracellular viruses were accumulated. We showed that HCV NS3 specifically interacted with Rad51 and accumulated Rad51 in the cytosol. Furthermore, Rad51 was coprecipitated with NS3 and HCV RNA. By employing membrane flotation and protease protection assays, we also demonstrated that Rad51 was co-fractionated with HCV NS3 on the lipid raft. These data indicate that Rad51 may be a component of the HCV RNA replication complex. Collectively, these data suggest that HCV may exploit cellular Rad51 to promote viral propagation and thus Rad51 may be a potential therapeutic target for HCV.

20.
Front Microbiol ; 8: 1129, 2017.
Article de Anglais | MEDLINE | ID: mdl-28674529

RÉSUMÉ

Abl is a central regulator of multiple cellular processes controlling actin dynamics, proliferation, and differentiation. Here, we showed that knockdown of Abl impaired hepatitis C virus (HCV) propagation. Treatment of Abl tyrosine kinase-specific inhibitor, imatinib and dasatinib, also significantly decreased HCV RNA and protein levels in HCV-infected cells. We showed that both imatinib and dasatinib selectively inhibited HCV infection at the entry step of HCV life cycle, suggesting that Abl kinase activity may be necessary for HCV entry. Using HCV pseudoparticle infection assays, we verified that Abl is required for viral entry. By employing transferrin uptake and immunofluorescence assays, we further demonstrated that Abl was involved in HCV entry at a clathrin-mediated endocytosis step. These data suggest that Abl may represent a novel host factor for HCV entry.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE