Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cells ; 44(9): 688-695, 2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34518443

ABSTRACT

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.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Isoquinolines/pharmacology , SARS-CoV-2/growth & development , Sulfonamides/pharmacology , Viral Protease Inhibitors/pharmacology , Virus Replication/drug effects , Animals , Carbamates/pharmacology , Cell Line , Chlorocebus aethiops , DNA-Directed RNA Polymerases/antagonists & inhibitors , HEK293 Cells , Hepacivirus/drug effects , Humans , Imidazoles/pharmacology , Pyrrolidines/pharmacology , SARS-CoV-2/drug effects , Sofosbuvir/pharmacology , Valine/analogs & derivatives , Valine/pharmacology , Vero Cells , Viral Nonstructural Proteins/antagonists & inhibitors
2.
J Microbiol ; 59(1): 101-109, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33355889

ABSTRACT

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.


Subject(s)
Hepacivirus/metabolism , Hepatitis C/enzymology , Viral Nonstructural Proteins/metabolism , Hepacivirus/genetics , Hepacivirus/growth & development , Hepatitis C/genetics , Hepatitis C/virology , Host-Pathogen Interactions , Humans , Protein Binding , Viral Nonstructural Proteins/genetics
3.
J Virol ; 94(19)2020 09 15.
Article in English | MEDLINE | ID: mdl-32727880

ABSTRACT

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.


Subject(s)
Cortactin/metabolism , Hepacivirus/physiology , Viral Nonstructural Proteins/metabolism , Virion/physiology , Virus Assembly/physiology , Cell Line , Cytoskeletal Proteins/metabolism , HEK293 Cells , Hepatitis C/virology , Hepatitis C Antigens/metabolism , Humans , Immunoprecipitation , Phosphorylation , RNA, Small Interfering/genetics , Virus Internalization , Virus Replication
4.
Mol Cells ; 43(5): 469-478, 2020 May 31.
Article in English | MEDLINE | ID: mdl-32344996

ABSTRACT

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.


Subject(s)
Cytoskeletal Proteins/metabolism , Hepacivirus/physiology , Hepatitis C/virology , Homeodomain Proteins/metabolism , LIM Domain Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Cytoskeletal Proteins/genetics , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Hepatitis C/transmission , Homeodomain Proteins/genetics , Humans , LIM Domain Proteins/genetics , Protein Array Analysis , Protein Binding , Protein Domains/genetics , Viral Nonstructural Proteins/genetics , Virus Replication
5.
J Virol ; 94(5)2020 02 14.
Article in English | MEDLINE | ID: mdl-31801866

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Hepacivirus/physiology , Protein Interaction Domains and Motifs , Viral Nonstructural Proteins/metabolism , Virus Assembly/physiology , Adaptor Proteins, Signal Transducing/genetics , Cell Line , Gene Expression Regulation, Viral , Hepatitis C/virology , Humans , Immunoprecipitation , Phosphorylation , Protein Processing, Post-Translational , RNA, Small Interfering , RNA, Viral/metabolism , Virus Replication/physiology
6.
Sci Rep ; 9(1): 7288, 2019 05 13.
Article in English | MEDLINE | ID: mdl-31086268

ABSTRACT

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.


Subject(s)
Alkaloids/pharmacology , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Indolizines/pharmacology , Phenanthrenes/pharmacology , Proline/pharmacology , Virus Internalization/drug effects , Alkaloids/chemistry , Alkaloids/therapeutic use , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Drug Therapy, Combination , HEK293 Cells , Hepacivirus/metabolism , Hepatitis C, Chronic/virology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Indolizines/chemistry , Indolizines/therapeutic use , Interferon-alpha/pharmacology , Interferon-alpha/therapeutic use , Phenanthrenes/chemistry , Phenanthrenes/therapeutic use , Primary Cell Culture , Proline/therapeutic use , Sofosbuvir/pharmacology , Sofosbuvir/therapeutic use , Tetraspanin 28/metabolism , Tylophora/chemistry , Viral Envelope Proteins/metabolism
7.
J Virol ; 93(2)2019 01 15.
Article in English | MEDLINE | ID: mdl-30381483

ABSTRACT

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.


Subject(s)
Down-Regulation , Hepacivirus/pathogenicity , Hepatitis C/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Viral Nonstructural Proteins/metabolism , Cell Line , Chromatin/metabolism , DNA Damage , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , HEK293 Cells , Hepacivirus/metabolism , Hepatitis C/virology , Humans , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitination , Viral Nonstructural Proteins/chemistry , Virus Replication
8.
Sci Rep ; 8(1): 15486, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341327

ABSTRACT

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.


Subject(s)
Fusion Regulatory Protein 1, Heavy Chain/metabolism , Hepacivirus/physiology , Hepatitis C/virology , Multiprotein Complexes/metabolism , Fusion Regulatory Protein 1, Heavy Chain/genetics , Gene Expression Regulation , HEK293 Cells , Humans , Large Neutral Amino Acid-Transporter 1/metabolism , Leucine/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Oxidative Stress , Protein Transport , RNA, Small Interfering/genetics , Signal Transduction , Viral Nonstructural Proteins/metabolism , Virus Internalization , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...