Journal articles on the topic 'HCV membranous web'

To see the other types of publications on this topic, follow the link: HCV membranous web.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 33 journal articles for your research on the topic 'HCV membranous web.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Egger, Denise, Benno Wölk, Rainer Gosert, Leonardo Bianchi, Hubert E. Blum, Darius Moradpour, and Kurt Bienz. "Expression of Hepatitis C Virus Proteins Induces Distinct Membrane Alterations Including a Candidate Viral Replication Complex." Journal of Virology 76, no. 12 (June 15, 2002): 5974–84. http://dx.doi.org/10.1128/jvi.76.12.5974-5984.2002.

Full text
Abstract:
ABSTRACT Plus-strand RNA viruses characteristically replicate their genome in association with altered cellular membranes. In the present study, the capacity of hepatitis C virus (HCV) proteins to elicit intracellular membrane alterations was investigated by expressing, in tetracycline-regulated cell lines, a comprehensive panel of HCV proteins individually as well as in the context of the entire HCV polyprotein. As visualized by electron microscopy (EM), expression of the combined structural proteins core-E1-E2-p7, the NS3-4A complex, and protein NS4B induced distinct membrane alterations. By immunogold EM (IEM), the membrane-altering proteins were always found to localize to the respective altered membranes. NS4B, a protein of hitherto unknown function, induced a tight structure, designated membranous web, consisting of vesicles in a membranous matrix. Expression of the entire HCV polyprotein gave rise to membrane budding into rough endoplasmic reticulum vacuoles, to the membranous web, and to tightly associated vesicles often surrounding the membranous web. By IEM, all HCV proteins were found to be associated with the NS4B-induced membranous web, forming a membrane-associated multiprotein complex. A similar web-like structure in livers of HCV-infected chimpanzees was previously described (Pfeifer et al., Virchows Arch. B., 33:233-243, 1980). In view of this finding and the observation that all HCV proteins accumulate on the membranous web, we propose that the membranous web forms the viral replication complex in HCV-infected cells.
APA, Harvard, Vancouver, ISO, and other styles
2

Gosert, Rainer, Denise Egger, Volker Lohmann, Ralf Bartenschlager, Hubert E. Blum, Kurt Bienz, and Darius Moradpour. "Identification of the Hepatitis C Virus RNA Replication Complex in Huh-7 Cells Harboring Subgenomic Replicons." Journal of Virology 77, no. 9 (May 1, 2003): 5487–92. http://dx.doi.org/10.1128/jvi.77.9.5487-5492.2003.

Full text
Abstract:
ABSTRACT Formation of a membrane-associated replication complex, composed of viral proteins, replicating RNA, and altered cellular membranes, is a characteristic feature of plus-strand RNA viruses. Here, we demonstrate the presence of a specific membrane alteration, designated the membranous web, that contains hepatitis C virus (HCV) nonstructural proteins, as well as viral plus-strand RNA, in Huh-7 cells harboring autonomously replicating subgenomic HCV RNAs. Metabolic labeling with 5-bromouridine 5′-triphosphate in the presence of actinomycin D revealed that the membranous web is the site of viral RNA synthesis and therefore represents the replication complex of HCV.
APA, Harvard, Vancouver, ISO, and other styles
3

Sagan, Selena M., Yanouchka Rouleau, Cynthia Leggiadro, Lubica Supekova, Peter G. Schultz, Andrew I. Su, and John Paul Pezacki. "The influence of cholesterol and lipid metabolism on host cell structure and hepatitis C virus replication." Biochemistry and Cell Biology 84, no. 1 (February 1, 2006): 67–79. http://dx.doi.org/10.1139/o05-149.

Full text
Abstract:
The hepatitis C virus (HCV) replicates on a membrane protein complex composed of viral proteins, replicating RNA, and altered cellular membranes. Small-molecule inhibitors of cellular lipid–cholesterol metabolism such as 25-hydroxycholesterol, cerulenin, lovastatin, and GGTI-286 all show a negative effect on HCV replication. Perturbation of host cell lipid and cholesterol metabolism can disrupt replication complexes by altering membranous structures where replication occurs. Changes in cholesterol and (or) lipid composition can have a general effect on membrane structure. Alternatively, metabolic changes can exert a more subtle influence over replication complexes by altering localization of host proteins through alterations in lipid anchoring. Here, we use Huh-7 cells harboring subgenomic HCV replicons to demonstrate that 25-hydroxycholesterol, cerulenin, lovastatin, and GGTI-286 do not disrupt the membranous web where replication occurs, whereas cholesterol-depleting agents such as β-cyclodextrin do. Cellular imaging suggests that the HCV RNA can remain associated with subcellular compartments connected with replication complexes in the presence of metabolic inhibitors. Therefore, at least 2 different molecular mechanisms are possible for the inhibition of HCV replication through the modulation of cellular lipid and cholesterol metabolism.Key words: hepatitis C virus, lipid metabolism, fluorescence microscopy, electron microscopy, membranous web, statins.
APA, Harvard, Vancouver, ISO, and other styles
4

Wölk, Benno, Benjamin Büchele, Darius Moradpour, and Charles M. Rice. "A Dynamic View of Hepatitis C Virus Replication Complexes." Journal of Virology 82, no. 21 (August 20, 2008): 10519–31. http://dx.doi.org/10.1128/jvi.00640-08.

Full text
Abstract:
ABSTRACT Hepatitis C virus (HCV) replicates its genome in a membrane-associated replication complex (RC). Specific membrane alterations, designated membranous webs, represent predominant sites of HCV RNA replication. The principles governing HCV RC and membranous web formation are poorly understood. Here, we used replicons harboring a green fluorescent protein (GFP) insertion in nonstructural protein 5A (NS5A) to study HCV RCs in live cells. Two distinct patterns of NS5A-GFP were observed. (i) Large structures, representing membranous webs, showed restricted motility, were stable over many hours, were partitioned among daughter cells during cell division, and displayed a static internal architecture without detectable exchange of NS5A-GFP. (ii) In contrast, small structures, presumably representing small RCs, showed fast, saltatory movements over long distances. Both populations were associated with endoplasmic reticulum (ER) tubules, but only small RCs showed ER-independent, microtubule (MT)-dependent transport. We suggest that this MT-dependent transport sustains two distinct RC populations, which are both required during the HCV life cycle.
APA, Harvard, Vancouver, ISO, and other styles
5

Blanchard, Emmanuelle, and Philippe Roingeard. "The Hepatitis C Virus-Induced Membranous Web in Liver Tissue." Cells 7, no. 11 (November 1, 2018): 191. http://dx.doi.org/10.3390/cells7110191.

Full text
Abstract:
Host cell membrane rearrangements induced by the hepatitis C virus (HCV) have been exclusively studied in vitro. These studies have shown that HCV induces double-membrane vesicles (DMVs), which probably serve to separate replication sites from the cytoplasmic sensors of the innate immune response. We report for the first time the observation of HCV-induced membrane rearrangements in liver biopsy specimens from patients chronically infected with HCV. Unlike observations performed in vitro, the membranous web detected in liver tissue seems essentially made of clusters of single-membrane vesicles derived from the endoplasmic reticulum and close to lipid droplets. This suggests that the DMVs could be a hallmark of laboratory-adapted HCV strains, possibly due to their ability to achieve a high level of replication. Alternatively, the concealment of viral RNA in DMVs may be part of innate immune response mechanisms particularly developed in hepatoma cell lines cultured in vitro. In any case, this constitutes the first report showing the differences in the membranous web established by HCV in vitro and in vivo.
APA, Harvard, Vancouver, ISO, and other styles
6

Gouttenoire, Jérôme, Philippe Roingeard, François Penin, and Darius Moradpour. "Amphipathic α-Helix AH2 Is a Major Determinant for the Oligomerization of Hepatitis C Virus Nonstructural Protein 4B." Journal of Virology 84, no. 24 (October 6, 2010): 12529–37. http://dx.doi.org/10.1128/jvi.01798-10.

Full text
Abstract:
ABSTRACT Nonstructural protein 4B (NS4B) is a key organizer of hepatitis C virus (HCV) replication complex formation. It induces a specific membrane rearrangement, designated membranous web, that serves as a scaffold for the HCV replication complex. However, the mechanisms underlying membranous web formation are poorly understood. Based on fluorescence resonance energy transfer (FRET) and confirmatory coimmunoprecipitation analyses, we provide evidence for an oligomerization of NS4B in the membrane environment of intact cells. Several conserved determinants were found to be involved in NS4B oligomerization, through homotypic and heterotypic interactions. N-terminal amphipathic α-helix AH2, comprising amino acids 42 to 66, was identified as a major determinant for NS4B oligomerization. Mutations that affected the oligomerization of NS4B disrupted membranous web formation and HCV RNA replication, implying that oligomerization of NS4B is required for the creation of a functional replication complex. These findings enhance our understanding of the functional architecture of the HCV replication complex and may provide new angles for therapeutic intervention. At the same time, they expand the list of positive-strand RNA virus replicase components acting as oligomers.
APA, Harvard, Vancouver, ISO, and other styles
7

Perez-Berna, Ana J., Nuria Benseny-Cases, María José Rodríguez, Ricardo Valcarcel, José L. Carrascosa, Pablo Gastaminza, and Eva Pereiro. "Monitoring reversion of hepatitis C virus-induced cellular alterations by direct-acting antivirals using cryo soft X-ray tomography and infrared microscopy." Acta Crystallographica Section D Structural Biology 77, no. 11 (October 27, 2021): 1365–77. http://dx.doi.org/10.1107/s2059798321009955.

Full text
Abstract:
Hepatitis C virus (HCV) is an enveloped RNA virus. One of the hallmarks of HCV infection is a rearrangement of the host cell membranes, known as the `membranous web'. Full-field cryo soft X-ray tomography (cryo-SXT) in the water-window energy range (284–543 eV) was performed on the MISTRAL beamline to investigate, in whole unstained cells, the morphology of the membranous rearrangements induced in HCV replicon-harbouring cells in conditions close to the living physiological state. All morphological alterations could be reverted by a combination of sofosbuvir/daclatasvir, which are clinically approved antivirals (direct-acting antivirals; DAAs) for HCV infection. Correlatively combining cryo-SXT and 2D synchrotron-based infrared microscopy provides critical information on the chemical nature of specific infection-related structures, which allows specific patterns of the infection process or the DAA-mediated healing process to be distinguished.
APA, Harvard, Vancouver, ISO, and other styles
8

Kong, Lingbao, Akira Fujimoto, Mariko Nakamura, Haruyo Aoyagi, Mami Matsuda, Koichi Watashi, Ryosuke Suzuki, et al. "Prolactin Regulatory Element Binding Protein Is Involved in Hepatitis C Virus Replication by Interaction with NS4B." Journal of Virology 90, no. 6 (January 6, 2016): 3093–111. http://dx.doi.org/10.1128/jvi.01540-15.

Full text
Abstract:
ABSTRACTIt has been proposed that the hepatitis C virus (HCV) NS4B protein triggers the membranous HCV replication compartment, but the underlying molecular mechanism is not fully understood. Here, we screened for NS4B-associated membrane proteins by tandem affinity purification and proteome analysis and identified 202 host proteins. Subsequent screening of replicon cells with small interfering RNA identified prolactin regulatory element binding (PREB) to be a novel HCV host cofactor. The interaction between PREB and NS4B was confirmed by immunoprecipitation, immunofluorescence, and proximity ligation assays. PREB colocalized with double-stranded RNA and the newly synthesized HCV RNA labeled with bromouridine triphosphate in HCV replicon cells. Furthermore, PREB shifted to detergent-resistant membranes (DRMs), where HCV replication complexes reside, in the presence of NS4B expression in Huh7 cells. However, a PREB mutant lacking the NS4B-binding region (PREBd3) could not colocalize with double-stranded RNA and did not shift to the DRM in the presence of NS4B. These results indicate that PREB locates at the HCV replication complex by interacting with NS4B. PREB silencing inhibited the formation of the membranous HCV replication compartment and increased the protease and nuclease sensitivity of HCV replicase proteins and RNA in DRMs, respectively. Collectively, these data indicate that PREB promotes HCV RNA replication by participating in the formation of the membranous replication compartment and by maintaining its proper structure by interacting with NS4B. Furthermore, PREB was induced by HCV infectionin vitroandin vivo. Our findings provide new insights into HCV host cofactors.IMPORTANCEThe hepatitis C virus (HCV) protein NS4B can induce alteration of the endoplasmic reticulum and the formation of a membranous web structure, which provides a platform for the HCV replication complex. The molecular mechanism by which NS4B induces the membranous HCV replication compartment is not understood. We screened for NS4B-associated membrane proteins by tandem affinity purification and proteome analysis, followed by screening with small interfering RNA. We identified prolactin regulatory element binding (PREB) to be a novel HCV host cofactor. PREB is induced by HCV infection and recruited into the replication complex by interaction with NS4B. Recruited PREB promotes HCV RNA replication by participating in the formation of the membranous HCV replication compartment. To our knowledge, the effect of NS4B-binding protein on the formation of the membranous HCV replication compartment is newly described in this report. Our findings are expected to provide new insights into HCV host cofactors.
APA, Harvard, Vancouver, ISO, and other styles
9

Goueslain, Lucie, Khaled Alsaleh, Pauline Horellou, Philippe Roingeard, Véronique Descamps, Gilles Duverlie, Yann Ciczora, Czeslaw Wychowski, Jean Dubuisson, and Yves Rouillé. "Identification of GBF1 as a Cellular Factor Required for Hepatitis C Virus RNA Replication." Journal of Virology 84, no. 2 (November 11, 2009): 773–87. http://dx.doi.org/10.1128/jvi.01190-09.

Full text
Abstract:
ABSTRACT In infected cells, hepatitis C virus (HCV) induces the formation of membrane alterations referred to as membranous webs, which are sites of RNA replication. In addition, HCV RNA replication also occurs in smaller membrane structures that are associated with the endoplasmic reticulum. However, cellular mechanisms involved in the formation of HCV replication complexes remain largely unknown. Here, we used brefeldin A (BFA) to investigate cellular mechanisms involved in HCV infection. BFA acts on cell membranes by interfering with the activation of several members of the family of ADP-ribosylation factors (ARF), which can lead to a wide range of inhibitory actions on membrane-associated mechanisms of the secretory and endocytic pathways. Our data show that HCV RNA replication is highly sensitive to BFA. Individual knockdown of the cellular targets of BFA using RNA interference and the use of a specific pharmacological inhibitor identified GBF1, a guanine nucleotide exchange factor for small GTPases of the ARF family, as a host factor critically involved in HCV replication. Furthermore, overexpression of a BFA-resistant GBF1 mutant rescued HCV replication in BFA-treated cells, indicating that GBF1 is the BFA-sensitive factor required for HCV replication. Finally, immunofluorescence and electron microscopy analyses indicated that BFA does not block the formation of membranous web-like structures induced by expression of HCV proteins in a nonreplicative context, suggesting that GBF1 is probably involved not in the formation of HCV replication complexes but, rather, in their activity. Altogether, our results highlight a functional connection between the early secretory pathway and HCV RNA replication.
APA, Harvard, Vancouver, ISO, and other styles
10

Kim, Kiyoon, Young-seok Lee, Suyun Jeong, Daehong Kim, Suk Chon, Youngmi Kim Pak, Sungsoo Kim, Joohun Ha, Insug Kang, and Wonchae Choe. "A Small Molecule, 4-Phenylbutyric Acid, Suppresses HCV Replication via Epigenetically Induced Hepatic Hepcidin." International Journal of Molecular Sciences 21, no. 15 (August 1, 2020): 5516. http://dx.doi.org/10.3390/ijms21155516.

Full text
Abstract:
Hepatic hepcidin is a well-known major iron regulator and has been reported to be closely related to hepatitis C virus (HCV) replication. However, pharmacological targeting of the hepcidin in HCV replication has not been reported. A short-chain fatty acid, 4-Phenyl butyrate (4-PBA), is an acid chemical chaperone that acts as a histone deacetylase inhibitor (HDACi) to promote chromosomal histone acetylation. Here, we investigated the therapeutic effect of 4-PBA on hepcidin expression and HCV replication. We used HCV genotype 1b Huh 7.5-Con1 replicon cells and engraftment of NOD/SCID mice as in vitro and in vivo models to test the effect of 4-PBA. It was found that 4-PBA inhibited HCV replication in Huh7.5-Con1 replicon cells in a concentration- and time-dependent manner through the induction of hepcidin expression by epigenetic modification and subsequent upregulation of interferon-α signaling. HCV formed a membranous web composed of double-membrane vesicles and was utilized for RNA replication. Moreover, 4-PBA also disrupted the integrity of the membranous web and interfered with the molecular interactions critical for the assembly of the HCV replication complex. These findings suggest that 4-PBA is a key epigenetic inducer of anti-HCV hepatic hepcidin and might at least in part play a role in targeting host factors related to HCV infection as an attractive complement to current HCV therapies.
APA, Harvard, Vancouver, ISO, and other styles
11

Wong, Mun-Teng, and Steve S. Chen. "Human Choline Kinase-α Promotes Hepatitis C Virus RNA Replication through Modulation of Membranous Viral Replication Complex Formation." Journal of Virology 90, no. 20 (August 3, 2016): 9075–95. http://dx.doi.org/10.1128/jvi.00960-16.

Full text
Abstract:
ABSTRACTHepatitis C virus (HCV) infection reorganizes cellular membranes to create an active viral replication site named the membranous web (MW). The role that human choline kinase-α (hCKα) plays in HCV replication remains elusive. Here, we first showed that hCKα activity, not the CDP-choline pathway, promoted viral RNA replication. Confocal microscopy and subcellular fractionation of HCV-infected cells revealed that a small fraction of hCKα colocalized with the viral replication complex (RC) on the endoplasmic reticulum (ER) and that HCV infection increased hCKα localization to the ER. In the pTM-NS3-NS5B model, NS3-NS5B expression increased the localization of the wild-type, not the inactive D288A mutant, hCKα on the ER, and hCKα activity was required for effective trafficking of hCKα and NS5A to the ER. Coimmunoprecipitation showed that hCKα was recruited onto the viral RC presumably through its binding to NS5A domain 1 (D1). hCKα silencing or treatment with CK37, an hCKα activity inhibitor, abolished HCV-induced MW formation. In addition, hCKα depletion hindered NS5A localization on the ER, interfered with NS5A and NS5B colocalization, and mitigated NS5A-NS5B interactions but had no apparent effect on NS5A-NS4B and NS4B-NS5B interactions. Nevertheless, hCKα activity was not essential for the binding of NS5A to hCKα or NS5B. These findings demonstrate that hCKα forms a complex with NS5A and that hCKα activity enhances the targeting of the complex to the ER, where hCKα protein, not activity, mediates NS5A binding to NS5B, thereby promoting functional membranous viral RC assembly and viral RNA replication.IMPORTANCEHCV infection reorganizes the cellular membrane to create an active viral replication site named the membranous web (MW). Here, we report that human choline kinase-α (hCKα) acts as an essential host factor for HCV RNA replication. A fraction of hCKα colocalizes with the viral replication complex (RC) on the endoplasmic reticulum (ER) in HCV-infected cells. NS3-NS5B expression increases ER localization of wild-type, but not D288A mutant, hCKα, and hCKα activity facilitates the transport of itself and NS5A to the ER. Silencing or inactivation of hCKα abrogates MW formation. Moreover, hCKα is recruited by NS5A independent of hCKα activity, presumably through binding to NS5A D1. hCKα activity then mediates the ER targeting of the hCKα-NS5A complex. On the ER membrane, hCKα protein,per se, induces NS5A binding to NS5B, thereby promoting membranous RC formation and viral RNA replication. Our study may benefit the development of hCKα-targeted anti-HCV therapeutics.
APA, Harvard, Vancouver, ISO, and other styles
12

Hansen, Marianne D., Ingvild B. Johnsen, Kim A. Stiberg, Tatyana Sherstova, Takaji Wakita, Gabriel Mary Richard, Richard K. Kandasamy, Eliane F. Meurs, and Marit W. Anthonsen. "Hepatitis C virus triggers Golgi fragmentation and autophagy through the immunity-related GTPase M." Proceedings of the National Academy of Sciences 114, no. 17 (April 7, 2017): E3462—E3471. http://dx.doi.org/10.1073/pnas.1616683114.

Full text
Abstract:
Positive-stranded RNA viruses, such as hepatitis C virus (HCV), assemble their viral replication complexes by remodeling host intracellular membranes to a membranous web. The precise composition of these replication complexes and the detailed mechanisms by which they are formed are incompletely understood. Here we show that the human immunity-related GTPase M (IRGM), known to contribute to autophagy, plays a previously unrecognized role in this process. We show that IRGM is localized at the Golgi apparatus and regulates the fragmentation of Golgi membranes in response to HCV infection, leading to colocalization of Golgi vesicles with replicating HCV. Our results show that IRGM controls phosphorylation of GBF1, a guanine nucleotide exchange factor for Arf-GTPases, which normally operates in Golgi membrane dynamics and vesicle coating in resting cells. We also find that HCV triggers IRGM-mediated phosphorylation of the early autophagy initiator ULK1, thereby providing mechanistic insight into the role of IRGM in HCV-mediated autophagy. Collectively, our results identify IRGM as a key Golgi-situated regulator that links intracellular membrane remodeling by autophagy and Golgi fragmentation with viral replication.
APA, Harvard, Vancouver, ISO, and other styles
13

Castro, Victoria, Gema Calvo, Ginés Ávila-Pérez, Marlène Dreux, and Pablo Gastaminza. "Differential Roles of Lipin1 and Lipin2 in the Hepatitis C Virus Replication Cycle." Cells 8, no. 11 (November 18, 2019): 1456. http://dx.doi.org/10.3390/cells8111456.

Full text
Abstract:
Although their origin, nature and structure are not identical, a common feature of positive-strand RNA viruses is their ability to subvert host lipids and intracellular membranes to generate replication and assembly complexes. Recently, lipin1, a cellular enzyme that converts phosphatidic acid into diacylglycerol, has been implicated in the formation of the membranous web that hosts hepatitis C virus (HCV) replicase. In the liver, lipin1 cooperates with lipin2 to maintain glycerolipid homeostasis. We extended our previous study of the lipin family on HCV infection, by determining the impact of the lipin2 silencing on viral replication. Our data reveal that lipin2 silencing interferes with HCV virion secretion at late stages of the infection, without significantly affecting viral replication or assembly. Moreover, uninfected lipin2-, but not lipin1-deficient cells display alterations in mitochondrial and Golgi apparatus morphology, suggesting that lipin2 contributes to the maintenance of the overall organelle architecture. Finally, our data suggest a broader function of lipin2 for replication of HCV and other RNA viruses, in contrast with the specific impact of lipin1 silencing on HCV replication. Overall, this study reveals distinctive functions of lipin1 and lipin2 in cells of hepatic origin, a context in which they are often considered functionally redundant.
APA, Harvard, Vancouver, ISO, and other styles
14

Castro, Victoria, Gema Calvo, Ginés Ávila-Pérez, Marlène Dreux, and Pablo Gastaminza. "Differential Roles of Lipin1 and Lipin2 in the Hepatitis C Virus Replication Cycle." Proceedings 50, no. 1 (June 9, 2020): 30. http://dx.doi.org/10.3390/proceedings2020050030.

Full text
Abstract:
Although their origin, nature and structure are not identical, a common feature of positive-strand RNA viruses is their ability to subvert host lipids and intracellular membranes to generate replication and assembly complexes. Recently, lipin1, a cellular enzyme that converts phosphatidate into diacylglycerol, has been involved in the formation of the membranous web that hosts hepatitis C virus (HCV) replicase. In the liver, lipin1 cooperates with lipin2 to maintain glycerolipid homeostasis. We extended our previous study of the lipin family in HCV infection by determining the impact of the lipin2 silencing on viral replication. In contrast to the specific impact of lipin1 silencing on HCV replication, our data suggest a broader function of lipin2 not only in HCV infection, but also for the replication of other RNA viruses. Moreover, uninfected lipin2- but not lipin1-deficient cells display alterations in mitochondrial and Golgi morphology, suggesting that lipin2 contributes to the maintenance of the overall organelle architecture. Coinciding with Golgi fragmentation, our data reveal that lipin2 silencing mainly interferes with HCV virion secretion at late stages of the infection without significantly affecting viral replication or assembly. Overall, this study reveals distinctive functions of lipin1 and lipin2 in cells of hepatic origin, a context in which they are often considered functionally redundant.
APA, Harvard, Vancouver, ISO, and other styles
15

Stone, Michelle, Shuaizheng Jia, Won Do Heo, Tobias Meyer, and Kouacou V. Konan. "Participation of Rab5, an Early Endosome Protein, in Hepatitis C Virus RNA Replication Machinery." Journal of Virology 81, no. 9 (February 14, 2007): 4551–63. http://dx.doi.org/10.1128/jvi.01366-06.

Full text
Abstract:
ABSTRACT Like most positive-strand RNA viruses, hepatitis C virus (HCV) is believed to replicate its genome on the surface of rearranged membranes. We have shown previously that HCV NS4AB, but not the product NS4B, inhibits endoplasmic reticulum (ER)-to-Golgi protein traffic (K. V. Konan, T. H. Giddings, Jr., M. Ikeda, K. Li, S. M. Lemon, and K. Kirkegaard, J. Virol. 77:7843-7855). However, both NS4AB and NS4B can induce “membranous web” formation, first reported by Egger et al. (D. B Egger, R. Gosert, L. Bianchi, H. E. Blum, D. Moradpour, and K. Bienz, J. Virol. 76:5974-5984), which is also observed in HCV-infected cells (Y. Rouille, F. Helle, D. Delgrange, P. Roingeard, C. Voisset, E. Blanchard, S. Belouzard, J. McKeating, A. H. Patel, G. Maertens, T. Wakita, C. Wychowski, and J. Dubuisson, J. Virol. 80:2832-2841) and cells that bear a subgenomic NS5A-green fluorescent protein (GFP) replicon (D. Moradpour, M. J. Evans, R. Gosert, Z. Yuan, H. E. Blum, S. P. Goff, B. D. Lindenbach, and C. M. Rice, J. Virol. 78:7400-7409). To determine the intracellular origin of the web, we examined NS4B colocalization with endogenous cellular markers in the context of the full-length or subgenomic replicon. We found that, in addition to ER markers, early endosome (EE) proteins, including Rab5, were associated with web-inducing protein NS4B. Furthermore, an immunoisolated fraction containing NS4B was found to contain both ER and EE proteins. Using fluorescence microscopy, we showed that wild-type and constitutively active Rab5 proteins were associated with NS4B. Interestingly, expression of dominant-negative Rab5 resulted in significant loss of GFP fluorescence in NS5A-GFP replicon cells. We also found that a small reduction in Rab5 protein expression decreased HCV RNA synthesis significantly. Furthermore, transfection of labeled Rab5 small interfering RNAs into NS5A-GFP replicon cells resulted in a significant decrease in GFP fluorescence. Finally, Rab5 protein was found to coimmunoprecipitate with HCV NS4B. These studies suggest that EE proteins, including Rab5, may play a role in HCV genome replication or web formation.
APA, Harvard, Vancouver, ISO, and other styles
16

Chatterji, Udayan, Michael Bobardt, Andrew Tai, Malcolm Wood, and Philippe A. Gallay. "Cyclophilin and NS5A Inhibitors, but Not Other Anti-Hepatitis C Virus (HCV) Agents, Preclude HCV-Mediated Formation of Double-Membrane-Vesicle Viral Factories." Antimicrobial Agents and Chemotherapy 59, no. 5 (February 9, 2015): 2496–507. http://dx.doi.org/10.1128/aac.04958-14.

Full text
Abstract:
ABSTRACTAlthough the mechanisms of action (MoA) of nonstructural protein 3 inhibitors (NS3i) and NS5B inhibitors (NS5Bi) are well understood, the MoA of cyclophilin inhibitors (CypI) and NS5A inhibitors (NS5Ai) are not fully defined. In this study, we examined whether CypI and NS5Ai interfere with hepatitis C virus (HCV) RNA synthesis of replication complexes (RCs) or with an earlier step of HCV RNA replication, the creation of double-membrane vesicles (DMVs) essential for HCV RNA replication. In contrast to NS5Bi, both CypI and NS5Ai do not block HCV RNA synthesis by way of RCs, suggesting that they exert their antiviral activity prior to the establishment of enzymatically active RCs. We found that viral replication is not a precondition for DMV formation, since the NS3-NS5B polyprotein or NS5A suffices to create DMVs. Importantly, only CypI and NS5Ai, but not NS5Bi, mir-122, or phosphatidylinositol-4 kinase IIIα (PI4KIIIα) inhibitors, prevent NS3-NS5B-mediated DMV formation. NS3-NS5B was unable to create DMVs in cyclophilin A (CypA) knockdown (KD) cells. We also found that the isomerase activity of CypA is absolutely required for DMV formation. This not only suggests that NS5A and CypA act in concert to build membranous viral factories but that CypI and NS5Ai mediate their early anti-HCV effects by preventing the formation of organelles, where HCV replication is normally initiated. This is the first investigation to examine the effect of a large panel of anti-HCV agents on DMV formation, and the results reveal that CypI and NS5Ai act at the same membranous web biogenesis step of HCV RNA replication, thus indicating a new therapeutic target of chronic hepatitis C.
APA, Harvard, Vancouver, ISO, and other styles
17

Rouillé, Yves, François Helle, David Delgrange, Philippe Roingeard, Cécile Voisset, Emmanuelle Blanchard, Sandrine Belouzard, et al. "Subcellular Localization of Hepatitis C Virus Structural Proteins in a Cell Culture System That Efficiently Replicates the Virus." Journal of Virology 80, no. 6 (March 15, 2006): 2832–41. http://dx.doi.org/10.1128/jvi.80.6.2832-2841.2006.

Full text
Abstract:
ABSTRACT Due to the recent development of a cell culture model, hepatitis C virus (HCV) can be efficiently propagated in cell culture. This allowed us to reinvestigate the subcellular localization of HCV structural proteins in the context of an infectious cycle. In agreement with previous reports, confocal immunofluorescence analysis of the subcellular localization of HCV structural proteins indicated that, in infected cells, the glycoprotein heterodimer is retained in the endoplasmic reticulum. However, in contrast to other studies, the glycoprotein heterodimer did not accumulate in other intracellular compartments or at the plasma membrane. As previously reported, an association between the capsid protein and lipid droplets was also observed. In addition, a fraction of labeling was consistent with the capsid protein being localized in a membranous compartment that is associated with the lipid droplets. However, in contrast to previous reports, the capsid protein was not found in the nucleus or in association with mitochondria or other well-defined intracellular compartments. Surprisingly, no colocalization was observed between the glycoprotein heterodimer and the capsid protein in infected cells. Electron microscopy analyses allowed us to identify a membrane alteration similar to the previously reported “membranous web.” However, no virus-like particles were found in this type of structure. In addition, dense elements compatible with the size and shape of a viral particle were seldom observed in infected cells. In conclusion, the cell culture system for HCV allowed us for the first time to characterize the subcellular localization of HCV structural proteins in the context an infectious cycle.
APA, Harvard, Vancouver, ISO, and other styles
18

Okamoto, Toru, Hiroko Omori, Yuuki Kaname, Takayuki Abe, Yorihiro Nishimura, Tetsuro Suzuki, Tatsuo Miyamura, Tamotsu Yoshimori, Kohji Moriishi, and Yoshiharu Matsuura. "A Single-Amino-Acid Mutation in Hepatitis C Virus NS5A Disrupting FKBP8 Interaction Impairs Viral Replication." Journal of Virology 82, no. 7 (January 23, 2008): 3480–89. http://dx.doi.org/10.1128/jvi.02253-07.

Full text
Abstract:
ABSTRACT Hepatitis C virus (HCV) nonstructural protein 5A (NS5A) regulates viral replication through its interaction with host and other viral proteins. We have previously shown that FK506-binding protein 8 (FKBP8) binds to NS5A and recruits Hsp90 to form a complex that participates in the replication of HCV. In this study, we examined the biochemical characteristics of the interaction and the intracellular localization of NS5A and FKBP8. Surface plasmon resonance analysis revealed that the dissociation constant of the interaction between the purified FKBP8 and NS5A expressed in bacteria was 82 nM. Mutational analyses of NS5A revealed that a single amino acid residue of Val or Ile at position 121, which is well conserved among all genotypes of HCV, is critical for the specific interaction with FKBP8. Substitution of the Val121 to Ala drastically impaired the replication of HCV replicon cells, and the drug-resistant replicon cells emerging after drug selection were shown to have reverted to the original arrangement by replacing Ala121 with Val. Examination of individual fields of the replicon cells by both fluorescence microscopy and electron microscopy (the correlative fluorescence microscopy-electron microscopy technique) revealed that FKBP8 is partially colocalized with NS5A in the cytoplasmic structure known as the membranous web. These results suggest that specific interaction of NS5A with FKBP8 in the cytoplasmic compartment plays a crucial role in the replication of HCV.
APA, Harvard, Vancouver, ISO, and other styles
19

Guo, Min, Rongjuan Pei, Qi Yang, Huang Cao, Yun Wang, Chunchen Wu, Jizheng Chen, et al. "Phosphatidylserine-Specific Phospholipase A1 Involved in Hepatitis C Virus Assembly through NS2 Complex Formation." Journal of Virology 89, no. 4 (December 10, 2014): 2367–77. http://dx.doi.org/10.1128/jvi.02982-14.

Full text
Abstract:
ABSTRACTSeveral members of the phospholipase family have been reported to be involved in hepatitis C virus (HCV) replication. Here, we identified another phospholipase, phosphatidylserine-specific phospholipase A1 (PLA1A), as a host factor involved in HCV assembly. PLA1A was upregulated by HCV infection, and PLA1A knockdown significantly reduced J399EM (genotype 2a) HCV propagation at the assembly step but not the entry, RNA replication, and protein translation steps of the life cycle. Protein localization and interaction analysis further revealed a role of PLA1A in the interaction of NS2-E2 and NS2-NS5A, as the formation of the NS2-E2 and NS2-NS5A complexes was weakened in the absence of PLA1A. In addition, PLA1A stabilized the NS2/NS5A dotted structure during infection. These data suggest that PLA1A plays an important role in bridging the membrane-associated NS2-E2 complex and the NS5A-associated replication complex via its interaction with E2, NS2, and NS5A, which leads to a coordinating interaction between the structural and nonstructural proteins and facilitates viral assembly.IMPORTANCEHepatitis C virus (HCV) genomic replication is driven by the replication complex and occurs at the membranous web, while the lipid droplet is the organelle in which virion assembly is initiated. In this study, we identified phosphatidylserine-specific phospholipase A1 (PLA1A), a member of phospholipase A 1 family, as a novel host factor involved in the assembly process of HCV. PLA1A, which is induced by HCV infection at a late infection stage, interacts with HCV E2, NS2, and NS5A proteins and enhances and stabilizes the NS2-E2 and NS2-NS5A complex formation, which is essential for viral assembly. Thus, PLA1A is an important host factor which is involved in the initiation of the viral assembly in close proximity to Core-decorated lipid droplets through bringing together the HCV replication complex and envelope complex.
APA, Harvard, Vancouver, ISO, and other styles
20

Taguwa, Shuhei, Hiroto Kambara, Hiroko Omori, Hideki Tani, Takayuki Abe, Yoshio Mori, Tetsuro Suzuki, Tamotsu Yoshimori, Kohji Moriishi, and Yoshiharu Matsuura. "Cochaperone Activity of Human Butyrate-Induced Transcript 1 Facilitates Hepatitis C Virus Replication through an Hsp90-Dependent Pathway." Journal of Virology 83, no. 20 (August 5, 2009): 10427–36. http://dx.doi.org/10.1128/jvi.01035-09.

Full text
Abstract:
ABSTRACT Hepatitis C virus (HCV) nonstructural protein 5A (NS5A) is a component of the replication complex consisting of several host and viral proteins. We have previously reported that human butyrate-induced transcript 1 (hB-ind1) recruits heat shock protein 90 (Hsp90) and FK506-binding protein 8 (FKBP8) to the replication complex through interaction with NS5A. To gain more insights into the biological functions of hB-ind1 in HCV replication, we assessed the potential cochaperone-like activity of hB-ind1, because it has significant homology with cochaperone p23, which regulates Hsp90 chaperone activity. The chimeric p23 in which the cochaperone domain was replaced with the p23-like domain of hB-ind1 exhibited cochaperone activity comparable to that of the authentic p23, inhibiting the glucocorticoid receptor signaling in an Hsp90-dependent manner. Conversely, the chimeric hB-ind1 in which the p23-like domain was replaced with the cochaperone domain of p23 resulted in the same level of recovery of HCV propagation as seen in the authentic hB-ind1 in cells with knockdown of the endogenous hB-ind1. Immunofluorescence analyses revealed that hB-ind1 was colocalized with NS5A, FKBP8, and double-stranded RNA in the HCV replicon cells. HCV replicon cells exhibited a more potent unfolded-protein response (UPR) than the parental and the cured cells upon treatment with an inhibitor for Hsp90. These results suggest that an Hsp90-dependent chaperone pathway incorporating hB-ind1 is involved in protein folding in the membranous web for the circumvention of the UPR and that it facilitates HCV replication.
APA, Harvard, Vancouver, ISO, and other styles
21

Konan, Kouacou V., Thomas H. Giddings, Masanori Ikeda, Kui Li, Stanley M. Lemon, and Karla Kirkegaard. "Nonstructural Protein Precursor NS4A/B from Hepatitis C Virus Alters Function and Ultrastructure of Host Secretory Apparatus." Journal of Virology 77, no. 14 (July 15, 2003): 7843–55. http://dx.doi.org/10.1128/jvi.77.14.7843-7855.2003.

Full text
Abstract:
ABSTRACT The nonstructural proteins of hepatitis C virus (HCV) have been shown previously to localize to the endoplasmic reticulum (ER) when expressed singly or in the context of other HCV proteins. To determine whether the expression of HCV nonstructural proteins alters ER function, we tested the effect of expression of NS2/3/4A, NS4A, NS4B, NS4A/B, NS4B/5A, NS5A, and NS5B from genotype 1b HCV on anterograde traffic from the ER to the Golgi apparatus. Only the nominal precursor protein NS4A/B affected the rate of ER-to-Golgi traffic, slowing the rate of Golgi-specific modification of the vesicular stomatitis virus G protein expressed by transfection by approximately threefold. This inhibition of ER-to-Golgi traffic was not observed upon expression of the processed proteins NS4A and NS4B, singly or in combination. To determine whether secretion of other cargo proteins was inhibited by NS4A/B expression, we monitored the appearance of newly synthesized proteins on the cell surface in the presence and absence of NS4A/B expression; levels of all were reduced in the presence of NS4A/B. This reduction is also seen in cells that contain genome length HCV replicons: the rate of appearance of major histocompatibility complex class I (MHC-I) on the cell surface was reduced by three- to fivefold compared to that for a cured cell line. The inhibition of protein secretion caused by NS4A/B does not correlate with the ultrastructural changes leading to the formation a “membranous web” (D. Egger et al., J. Virol. 76:5974-5984, 2002), which can be caused by expression of NS4B alone. Inhibition of global ER-to-Golgi traffic could, by reducing cytokine secretion, MHC-I presentation, and transport of labile membrane proteins to the cell surface, have significant effects on the host immune response to HCV infection.
APA, Harvard, Vancouver, ISO, and other styles
22

Madan, Vanesa, David Paul, Volker Lohmann, and Ralf Bartenschlager. "Inhibition of HCV Replication by Cyclophilin Antagonists Is Linked to Replication Fitness and Occurs by Inhibition of Membranous Web Formation." Gastroenterology 146, no. 5 (May 2014): 1361–72. http://dx.doi.org/10.1053/j.gastro.2014.01.055.

Full text
APA, Harvard, Vancouver, ISO, and other styles
23

Reghellin, V., L. Donnici, S. Fenu, V. Berno, V. Calabrese, M. Pagani, S. Abrignani, F. Peri, R. De Francesco, and P. Neddermann. "NS5A Inhibitors Impair NS5A–Phosphatidylinositol 4-Kinase IIIα Complex Formation and Cause a Decrease of Phosphatidylinositol 4-Phosphate and Cholesterol Levels in Hepatitis C Virus-Associated Membranes." Antimicrobial Agents and Chemotherapy 58, no. 12 (September 15, 2014): 7128–40. http://dx.doi.org/10.1128/aac.03293-14.

Full text
Abstract:
ABSTRACTThe hepatitis C virus (HCV) nonstructural (NS) protein 5A is a multifunctional protein that plays a central role in viral replication and assembly. Antiviral agents directly targeting NS5A are currently in clinical development. Although the elucidation of the mechanism of action (MOA) of NS5A inhibitors has been the focus of intensive research, a detailed understanding of how these agents exert their antiviral effect is still lacking. In this study, we observed that the downregulation of NS5A hyperphosphorylation is associated with the actions of NS5A inhibitors belonging to different chemotypes. NS5A is known to recruit the lipid kinase phosphatidylinositol 4-kinase IIIα (PI4KIIIα) to the HCV-induced membranous web in order to generate phosphatidylinositol 4-phosphate (PI4P) at the sites of replication. We demonstrate that treatment with NS5A inhibitors leads to an impairment in the NS5A-PI4KIIIα complex formation that is paralleled by a significant reduction in PI4P and cholesterol levels within the endomembrane structures of HCV-replicating cells. A similar decrease in PI4P and cholesterol levels was also obtained upon treatment with a PI4KIIIα-targeting inhibitor. In addition, both the NS5A and PI4KIIIα classes of inhibitors induced similar subcellular relocalization of the NS5A protein, causing the formation of large cytoplasmic NS5A-containing clusters previously reported to be one of the hallmarks of inhibition of the action of PI4KIIIα. Because of the similarities between the effects induced by treatment with PI4KIIIα or NS5A inhibitors and the observation that agents targeting NS5A impair NS5A-PI4KIIIα complex formation, we speculate that NS5A inhibitors act by interfering with the function of the NS5A-PI4KIIIα complex.
APA, Harvard, Vancouver, ISO, and other styles
24

Lundin, Marika, Hannah Lindström, Caroline Grönwall, and Mats A. A. Persson. "Dual topology of the processed hepatitis C virus protein NS4B is influenced by the NS5A protein." Journal of General Virology 87, no. 11 (November 1, 2006): 3263–72. http://dx.doi.org/10.1099/vir.0.82211-0.

Full text
Abstract:
Among the least-known hepatitis C virus proteins is the non-structural protein 4B (NS4B). It localizes to the endoplasmic reticulum (ER) membrane and induces membrane changes, resulting in a membranous web that is reported to be the locale for virus replication. A model was presented previously for the topology of recombinant HCV NS4B of the 1a genotype based on in vitro data. In this model, the N-terminal tail of a considerable fraction of the NS4B molecules was translocated into the ER lumen via a post-translational process, giving the protein a dual transmembrane topology. It is now reported that translocation of the N terminus also occurs for processed NS4B expressed in cells in the context of the polyprotein. In the presence of NS5A, however, a lower degree of translocation was observed, which may indicate that NS5A influences the topology of NS4B. In vitro expression studies of NS4B from all major genotypes demonstrated that translocation of the N terminus to the ER lumen is conserved across genotypes. This clearly suggests an important function for this feature. Furthermore, when disrupting a previously reported amphipathic helix (AH) in the N terminus of NS4B, translocation was inhibited. As a disrupted AH also abolished the ability of NS4B to rearrange membranes, these data indicate for the first time an association between translocation of the N terminus and membrane rearrangement. Finally, the present experiments also confirm the predicted location of the first luminal loop to be around aa 112.
APA, Harvard, Vancouver, ISO, and other styles
25

Moradpour, Darius, Matthew J. Evans, Rainer Gosert, Zhenghong Yuan, Hubert E. Blum, Stephen P. Goff, Brett D. Lindenbach, and Charles M. Rice. "Insertion of Green Fluorescent Protein into Nonstructural Protein 5A Allows Direct Visualization of Functional Hepatitis C Virus Replication Complexes." Journal of Virology 78, no. 14 (July 15, 2004): 7400–7409. http://dx.doi.org/10.1128/jvi.78.14.7400-7409.2004.

Full text
Abstract:
ABSTRACT Hepatitis C virus (HCV) replicates its genome in a membrane-associated replication complex, composed of viral proteins, replicating RNA and altered cellular membranes. We describe here HCV replicons that allow the direct visualization of functional HCV replication complexes. Viable replicons selected from a library of Tn7-mediated random insertions in the coding sequence of nonstructural protein 5A (NS5A) allowed the identification of two sites near the NS5A C terminus that tolerated insertion of heterologous sequences. Replicons encoding green fluorescent protein (GFP) at these locations were only moderately impaired for HCV RNA replication. Expression of the NS5A-GFP fusion protein could be demonstrated by immunoblot, indicating that the GFP was retained during RNA replication and did not interfere with HCV polyprotein processing. More importantly, expression levels were robust enough to allow direct visualization of the fusion protein by fluorescence microscopy. NS5A-GFP appeared as brightly fluorescing dot-like structures in the cytoplasm. By confocal laser scanning microscopy, NS5A-GFP colocalized with other HCV nonstructural proteins and nascent viral RNA, indicating that the dot-like structures, identified as membranous webs by electron microscopy, represent functional HCV replication complexes. These findings reveal an unexpected flexibility of the C-terminal domain of NS5A and provide tools for studying the formation and turnover of HCV replication complexes in living cells.
APA, Harvard, Vancouver, ISO, and other styles
26

Knodel, Markus, Paul Targett-Adams, Alfio Grillo, Eva Herrmann, and Gabriel Wittum. "Advanced Hepatitis C Virus Replication PDE Models within a Realistic Intracellular Geometric Environment." International Journal of Environmental Research and Public Health 16, no. 3 (February 12, 2019): 513. http://dx.doi.org/10.3390/ijerph16030513.

Full text
Abstract:
The hepatitis C virus (HCV) RNA replication cycle is a dynamic intracellular process occurring in three-dimensional space (3D), which is difficult both to capture experimentally and to visualize conceptually. HCV-generated replication factories are housed within virus-induced intracellular structures termed membranous webs (MW), which are derived from the Endoplasmatic Reticulum (ER). Recently, we published 3D spatiotemporal resolved diffusion–reaction models of the HCV RNA replication cycle by means of surface partial differential equation (sPDE) descriptions. We distinguished between the basic components of the HCV RNA replication cycle, namely HCV RNA, non-structural viral proteins (NSPs), and a host factor. In particular, we evaluated the sPDE models upon realistic reconstructed intracellular compartments (ER/MW). In this paper, we propose a significant extension of the model based upon two additional parameters: different aggregate states of HCV RNA and NSPs, and population dynamics inspired diffusion and reaction coefficients instead of multilinear ones. The combination of both aspects enables realistic modeling of viral replication at all scales. Specifically, we describe a replication complex state consisting of HCV RNA together with a defined amount of NSPs. As a result of the combination of spatial resolution and different aggregate states, the new model mimics a cis requirement for HCV RNA replication. We used heuristic parameters for our simulations, which were run only on a subsection of the ER. Nevertheless, this was sufficient to allow the fitting of core aspects of virus reproduction, at least qualitatively. Our findings should help stimulate new model approaches and experimental directions for virology.
APA, Harvard, Vancouver, ISO, and other styles
27

Tang, Hengli, and Henry Grisé. "Cellular and molecular biology of HCV infection and hepatitis." Clinical Science 117, no. 2 (June 15, 2009): 49–65. http://dx.doi.org/10.1042/cs20080631.

Full text
Abstract:
HCV (hepatitis C virus) infects nearly 3% of the population worldwide and has emerged as a major causative agent of liver disease, resulting in acute and chronic infections that can lead to fibrosis, cirrhosis and hepatocellular carcinoma. Hepatitis C represents the leading cause of liver transplantation in the United States and Europe. A positive-strand RNA virus of the Flaviviridae family, HCV contains a single-stranded RNA genome of approx. 9600 nucleotides. The genome RNA serves as both mRNA for translation of viral proteins and the template for RNA replication. Cis-acting RNA elements within the genome regulate RNA replication by forming secondary structures that interact with each other and trans-acting factors. Although structural proteins are clearly dispensable for RNA replication, recent evidence points to an important role of several non-structural proteins in particle assembly and release, turning their designation on its head. HCV enters host cells through receptor-mediated endocytosis, and the process requires the co-ordination of multiple cellular receptors and co-receptors. RNA replication takes place at specialized intracellular membrane structures called ‘membranous webs’ or ‘membrane-associated foci’, whereas viral assembly probably occurs on lipid droplets and endoplasmic reticulum. Liver inflammation plays a central role in the liver damage seen in hepatitis C, but many HCV proteins also directly contribute to HCV pathogenesis. In the present review, the molecular and cellular aspects of the HCV life cycle and the role of viral proteins in pathological liver conditions caused by HCV infection are described.
APA, Harvard, Vancouver, ISO, and other styles
28

Merugu, Ramchander, and Kalpana V. Singh. "MOLECULAR DOCKING OF AMITRIPTYLINE TO CERULOPLASMIN, RETINOL-BINDING PROTEIN, AND SERUM ALBUMIN." Asian Journal of Pharmaceutical and Clinical Research 11, no. 2 (February 1, 2018): 169. http://dx.doi.org/10.22159/ajpcr.2018.v11i2.22721.

Full text
Abstract:
Objective: A drug’s efficiency depends on the binding capacity of the drug with the particular plasma protein. The less bound drug can be easily diffused through cell membranes. The present study deals with in silico studies of amitriptyline binding to three plasma proteins human ceruloplasmin (HCP), cellular retinol-binding protein (CRBP), and human serum albumin (HSA) and tries to establish the binding capacity behavior with the frontier molecular orbital approach.Methods: Amitriptyline is selected as legend and docked with three plasma proteins HCP, HCP PDB ID 1KCW, CRBP PDB ID 5LJC, and HSA. Docking calculations were carried out using docking server. frontier molecular orbital calculations are performed through web-based computational chemistry interface WEBMO version 17.0.012e using server Buchhner.chem.hope.edu. on computational engine MOPAC.Results: HCP and HSA predominantly show polar and hydrophobic interactions, whereas CRBP forms hydrogen bond apart from polar and hydrophobic interactions. Favorable values of inhibition constant, Ki, is obtained which is equal to 1.13 μM for CRBP, 6.00 μM for HCP, and 2.00 μM for has.Conclusion: A studies prove that amitriptyline can bind to all three plasma proteins, namely, HCP, CRBP, and HSA. Amitriptyline binds to an HSA and HCP through polar and hydrophobic interactions while weak electrostatic interactions felicitate diffusion of amitriptyline through the plasma membrane. Comparatively, strong hydrogen bond in CRBP may make the bound drug to be released at a slow rate. Strong binding of amitriptyline to CRBP is also evident from the least value of inhibition constant, Ki, which is equal to 1.13 μM for CRBP, 6.00 μM for HCP, and 2.00 μM for has.
APA, Harvard, Vancouver, ISO, and other styles
29

Stoeck, Ina Karen, Ji-Young Lee, Keisuke Tabata, Inés Romero-Brey, David Paul, Philipp Schult, Volker Lohmann, Lars Kaderali, and Ralf Bartenschlager. "Hepatitis C Virus Replication Depends on Endosomal Cholesterol Homeostasis." Journal of Virology 92, no. 1 (October 18, 2017). http://dx.doi.org/10.1128/jvi.01196-17.

Full text
Abstract:
ABSTRACT Similar to other positive-strand RNA viruses, hepatitis C virus (HCV) causes massive rearrangements of intracellular membranes, resulting in a membranous web (MW) composed of predominantly double-membrane vesicles (DMVs), the presumed sites of RNA replication. DMVs are enriched for cholesterol, but mechanistic details on the source and recruitment of cholesterol to the viral replication organelle are only partially known. Here we focused on selected lipid transfer proteins implicated in direct lipid transfer at various endoplasmic reticulum (ER)-membrane contact sites. RNA interference (RNAi)-mediated knockdown identified several hitherto unknown HCV dependency factors, such as steroidogenic acute regulatory protein-related lipid transfer domain protein 3 (STARD3), oxysterol-binding protein-related protein 1A and -B (OSBPL1A and -B), and Niemann-Pick-type C1 (NPC1), all residing at late endosome and lysosome membranes and required for efficient HCV RNA replication but not for replication of the closely related dengue virus. Focusing on NPC1, we found that knockdown or pharmacological inhibition caused cholesterol entrapment in lysosomal vesicles concomitant with decreased cholesterol abundance at sites containing the viral replicase factor NS5A. In untreated HCV-infected cells, unesterified cholesterol accumulated at the perinuclear region, partially colocalizing with NS5A at DMVs, arguing for NPC1-mediated endosomal cholesterol transport to the viral replication organelle. Consistent with cholesterol being an important structural component of DMVs, reducing NPC1-dependent endosomal cholesterol transport impaired MW integrity. This suggests that HCV usurps lipid transfer proteins, such as NPC1, at ER-late endosome/lysosome membrane contact sites to recruit cholesterol to the viral replication organelle, where it contributes to MW functionality. IMPORTANCE A key feature of the replication of positive-strand RNA viruses is the rearrangement of the host cell endomembrane system to produce a membranous replication organelle (RO). The underlying mechanisms are far from being elucidated fully. In this report, we provide evidence that HCV RNA replication depends on functional lipid transport along the endosomal-lysosomal pathway that is mediated by several lipid transfer proteins, such as the Niemann-Pick type C1 (NPC1) protein. Pharmacological inhibition of NPC1 function reduced viral replication, impaired the transport of cholesterol to the viral replication organelle, and altered organelle morphology. Besides NPC1, our study reports the importance of additional endosomal and lysosomal lipid transfer proteins required for viral replication, thus contributing to our understanding of how HCV manipulates their function in order to generate a membranous replication organelle. These results might have implications for the biogenesis of replication organelles of other positive-strand RNA viruses.
APA, Harvard, Vancouver, ISO, and other styles
30

Kong, Lingbao, Haruyo Aoyagi, Zibing Yang, Tao Ouyang, Mami Matsuda, Akira Fujimoto, Koichi Watashi, et al. "Surfeit 4 Contributes to the Replication of Hepatitis C Virus Using Double-Membrane Vesicles." Journal of Virology 94, no. 2 (October 23, 2019). http://dx.doi.org/10.1128/jvi.00858-19.

Full text
Abstract:
ABSTRACT A number of positive-strand RNA viruses, such as hepatitis C virus (HCV) and poliovirus, use double-membrane vesicles (DMVs) as replication sites. However, the role of cellular proteins in DMV formation during virus replication is poorly understood. HCV NS4B protein induces the formation of a “membranous web” structure that provides a platform for the assembly of viral replication complexes. Our previous screen of NS4B-associated host membrane proteins by dual-affinity purification, liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS), and small interfering RNA (siRNA) methods revealed that the Surfeit 4 (Surf4) gene, which encodes an integral membrane protein, is involved in the replication of the JFH1 subgenomic replicon. Here, we investigated in detail the effect of Surf4 on HCV replication. Surf4 affects HCV replication in a genotype-independent manner, whereas HCV replication does not alter Surf4 expression. The influence of Surf4 on HCV replication indicates that while Surf4 regulates replication, it has no effect on entry, translation, assembly, or release. Analysis of the underlying mechanism showed that Surf4 is recruited into HCV RNA replication complexes by NS4B and is involved in the formation of DMVs and the structural integrity of RNA replication complexes. Surf4 also participates in the replication of poliovirus, which uses DMVs as replication sites, but it has no effect on the replication of dengue virus, which uses invaginated/sphere-type vesicles as replication sites. These findings clearly show that Surf4 is a novel cofactor that is involved in the replication of positive-strand RNA viruses using DMVs as RNA replication sites, which provides valuable clues for DMV formation during positive-strand RNA virus replication. IMPORTANCE Hepatitis C virus (HCV) NS4B protein induces the formation of a membranous web (MW) structure that provides a platform for the assembly of viral replication complexes. The main constituents of the MW are double-membrane vesicles (DMVs). Here, we found that the cellular protein Surf4, which maintains endoplasmic reticulum (ER)-Golgi intermediate compartments and the Golgi compartment, is recruited into HCV RNA replication complexes by NS4B and is involved in the formation of DMVs. Moreover, Surf4 participates in the replication of poliovirus, which uses DMVs as replication sites, but has no effect on the replication of dengue virus, which uses invaginated vesicles as replication sites. These results indicate that the cellular protein Surf4 is involved in the replication of positive-strand RNA viruses that use DMVs as RNA replication sites, providing new insights into DMV formation during virus replication and potential targets for the diagnosis and treatment of positive-strand RNA viruses.
APA, Harvard, Vancouver, ISO, and other styles
31

Fahmy, Ahmed M., and Patrick Labonté. "The autophagy elongation complex (ATG5-12/16L1) positively regulates HCV replication and is required for wild-type membranous web formation." Scientific Reports 7, no. 1 (January 9, 2017). http://dx.doi.org/10.1038/srep40351.

Full text
APA, Harvard, Vancouver, ISO, and other styles
32

Wong, Mun-Teng, and Steve S. Chen. "Hepatitis C Virus Subverts Human Choline Kinase-α To Bridge Phosphatidylinositol-4-Kinase IIIα (PI4KIIIα) and NS5A and Upregulates PI4KIIIα Activation, Thereby Promoting the Translocation of the Ternary Complex to the Endoplasmic Reticulum for Viral Replication." Journal of Virology 91, no. 16 (May 31, 2017). http://dx.doi.org/10.1128/jvi.00355-17.

Full text
Abstract:
ABSTRACT In this study, we elucidated the mechanism by which human choline kinase-α (hCKα) interacts with nonstructural protein 5A (NS5A) and phosphatidylinositol-4-kinase IIIα (PI4KIIIα), the lipid kinase crucial for maintaining the integrity of virus-induced membranous webs, and modulates hepatitis C virus (HCV) replication. hCKα activity positively modulated phosphatidylinositol-4-phosphate (PI4P) levels in HCV-expressing cells, and hCKα-mediated PI4P accumulation was abolished by AL-9, a PI4KIIIα-specific inhibitor. hCKα colocalized with NS5A and PI4KIIIα or PI4P; NS5A expression increased hCKα and PI4KIIIα colocalization; and hCKα formed a ternary complex with PI4KIIIα and NS5A, supporting the functional interplay of hCKα with PI4KIIIα and NS5A. PI4KIIIα inactivation by AL-9 or hCKα inactivation by CK37, a specific hCKα inhibitor, impaired the endoplasmic reticulum (ER) localization and colocalization of these three molecules. Interestingly, hCKα knockdown or inactivation inhibited PI4KIIIα-NS5A binding. In an in vitro PI4KIIIα activity assay, hCKα activity slightly increased PI4KIIIα basal activity but greatly augmented NS5A-induced PI4KIIIα activity, supporting the essential role of ternary complex formation in robust PI4KIIIα activation. Concurring with the upregulation of PI4P production and viral replication, overexpression of active hCKα-R (but not the D288A mutant) restored PI4KIIIα and NS5A translocation to the ER in hCKα stable knockdown cells. Furthermore, active PI4KIIIα overexpression restored PI4P production, PI4KIIIα and NS5A translocation to the ER, and viral replication in CK37-treated cells. Based on our results, hCKα functions as an indispensable regulator that bridges PI4KIIIα and NS5A and potentiates NS5A-stimulated PI4KIIIα activity, which then facilitates the targeting of the ternary complex to the ER for viral replication. IMPORTANCE The mechanisms by which hCKα activity modulates the transport of the hCKα-NS5A complex to the ER are not understood. In the present study, we investigated how hCKα interacts with PI4KIIIα (a key element that maintains the integrity of the “membranous web” structure) and NS5A to regulate viral replication. We demonstrated that HCV hijacks hCKα to bridge PI4KIIIα and NS5A, forming a ternary complex, which then stimulates PI4KIIIα activity to produce PI4P. Pronounced PI4P synthesis then redirects the translocation of the ternary complex to the ER-derived, PI4P-enriched membrane for assembly of the viral replication complex and viral replication. Our study provides novel insights into the indispensable modulatory role of hCKα in the recruitment of PI4KIIIα to NS5A and in NS5A-stimulated PI4P production and reveals a new perspective for understanding the impact of profound PI4KIIIα activation on the targeting of PI4KIIIα and NS5A to the PI4P-enriched membrane for viral replication complex formation.
APA, Harvard, Vancouver, ISO, and other styles
33

Rabêlo, Hannah Taynnan de Lima Bezerra, José Henrique de Araújo Cruz, Gymenna Maria Tenório Guênes, Abrahão Alves de Oliveira Filho, and Maria Angélica Satyro Gomes Alves. "Anestésicos locais utilizados na Odontologia: uma revisão de literatura." ARCHIVES OF HEALTH INVESTIGATION 8, no. 9 (February 20, 2020). http://dx.doi.org/10.21270/archi.v8i9.4655.

Full text
Abstract:
Introdução: Os anestésicos locais são substâncias químicas capazes de bloquear de forma reversível a transmissão de impulsos nervosos no local onde forem aplicados, sendo fundamentais no âmbito da Odontologia para controle da dor. Objetivo: Realizar revisão de literatura sobre os principais anestésicos locais utilizados na Odontologia. Metodologia: O estudo trata-se de uma revisão narrativa realizada entre os meses de janeiro e março de 2018. As bases de dados para a busca da literatura foram Scielo, Pubmed, Web of Science, Bireme. As palavras-chaves usadas foram “Anestésicos Locais”, “Mecanismos de Ação”, “Farmacologia”, “Odontologia”, “Relação Estrutura-atividade”, “Canais de Sódio”, presentes no DeCS. Discussão: A cocaína foi o primeiro composto a ser utilizado como anestésico local e foi a partir desta que os novos anestésicos foram desenvolvidos. Os anestésicos locais apresentam na sua estrutura molecular um anel aromático, uma cadeia intermediária e um grupo amina. Eles atuam sobre os neurônios e seu sítio de ação são os canais de sódio dependente de voltagem, aos quais se ligam reversivelmente, abolindo a excitabilidade neuronal. A classificação dos anestésicos locais é definida quanto à estrutura química e quanto à duração de ação, sendo os principais utilizados na Odontologia lidocaína, mepivacaína, articaína, prilocaína, cloridrato de bupivacaína. Conclusão: Conforme a literatura revisada, é necessário o conhecimento do cirurgião-dentista sobre as características farmacológicas individuais dos anestésicos locais e as sistêmicas do paciente para uma escolha correta, já que sua utilização é variável para cada usuário, e a manipulação inadequada desses fármacos pode levar a sérios riscos para a saúde do paciente.Descritores: Odontologia; Anestésicos locais; Farmacologia; Relação Estrutura-Atividade; Canais de Sódio; Mecanismos Moleculares de Ação Farmacológica.Almeida FM. Controle medicamentoso da dor. In: Estrela C. Dor odontogênica. São Paulo: Artes Médicas; 2001. p.243-61.Silva AP, Diniz AS, Araújo FA, Souza CC. Presença da queixa de dor em pacientes classificados segundo o protocolo de Manchester. Rev Enferm Centro Oeste Mineiro. 2013;3(1):507-17.Rang HP, Dale MM, Ritter JM, Flower RJ, Henderson G. Farmacologia. 8. ed. Rio de Janeiro: Elsevier; 2016.Paiva LCA, Cavalcanti AL. Anestésicos locais em odontologia: Uma revisão de literatura. UEPG Ci Biol Saúde. 2005; 11(2):35-42.Soares RG, Salles AA, Irala LED, Limongi O. Como escolher um adequado anestésico local para as diferentes situações na clínica odontológica diária? RSBO. 2006;3(1):35-40.Miller RD, Hondeghem LM. Anestésicos Locais. In: Katzung BG. Farmacologia básica e clínica. 10. ed. Rio de Janeiro: AMGH Editora; 2010. p.301-7.Becker D, Reeed K. Essentials of local anesthetic pharmacology. Anesth Prog. 2006;53(3):98-108.Alves RIL. Anestésicos locais [dissertação]. Porto, Portugal: Universidade Fernando Pessoa; 2013.Malamed SF. Manual de anestesia local. 6. ed. Rio de Janeiro: Elsevier; 2013.Ferreira AAA, Silva ID, Diniz RS, Guerra GCB. Anestésicos locais: revisando o mecanismo de ação molecular. Infarma. 2006;18(5/6):15-18.Anjos ED, Carvalho RWF. Complicações sistêmicas em anestesia local. In: Lubiana NB. Pro-Odonto Cirurgia. 2. ed. Porto Alegre: Artmed; 2007. p.143-78.Carvalho RWF, Pereira CU, Anjos ED, Laureano Filho JR, Vasconcelos BCE. Anestésicos locais: como escolher e prevenir complicações sistêmicas. Rev Port Estomatol Med Dent Cir Maxilofac. 2010;51(2):113-20.Teixeira RN. Anestesia local sem vasoconstritor versus com vasoconstritor [dissertação] Porto, Portugal: Faculdade de Ciências da Saúde, Universidade Fernando Pessoa; 2014.Carvalho B, Fritzen EL, Parodes AG, Santos RB, Gedoz L. O emprego dos anestésicos locais em Odontologia: revisão de literatura. Rev bras odontol. 2013;70(2):178-81.Santaella GM. Soluções anestésicas locais: uma revisão de literatura [monografia]: Florianópolis: Universidade Federal de Santa Catarina; 2011.Reis Jr A. Sigmund Freud (1856-1939) e Karl Köller (1857-1944) e a descoberta da anestesia local. Rev Bras Anestesiol. 2009;59(2):244-57. Bobbio A. História Sinótica da Anestesia. São Paulo: Novel; 1969.Byck R. Freud e a Cocaína. Rio de Janeiro: Espaço e Tempo; 1989.Araújo DR, Paula E, Fraceto LF. Anestésicos locais: interação com membranas biológicas e com o canal de sódio voltagem-dependente. Quim Nova. 2008;31(7):1775-83.Catterall WA, Mackie K. Local anesthetics. In: Brunton LL, Lazo JS, Parker KL. Goodman and Gillman’s the pharmacologic basis of therapeutics. 11. ed. New York: McGraw Hill; 2006. p. 369-85.Carvalho JCA. Farmacologia dos anestésicos locais. Rev Bras Anestesiol. 1994;44(1):75-82.Faria FAC, Marzola C. Farmacologia dos anestésicos locais – considerações gerias. BCI. 2001;8(29):19-30.Bahl R. Local anestesia in dentistry. Anesth Prog. 2004;51(4):138-42.Udelsmann A, Dreyer E, Melo MS, Bonfim MR, Borsoi LFA, Oliveira TG. Lipídeos nas intoxicações por anestésicos locais. ABCD arq bras cir dig. 2012;25(3):169-72.Fozzard HA, Lee PJ, Lipkind JM. Mechanism of local anesthetic drug action on voltage-gated sodium channels. Curr Pharm Des. 2005;11(21):2671-86.Jackson T, Mclure HA. Pharmacology of local anesthetics. Ophthalmol Clin North Am. 2006;19(2):155-61.Schulman JM, Strichartz GR. Farmacologia dos Anestésicos Locais. In: Golan DE, Tashjian Jr AH, Armstrong EJ, Armstrong AW. Princípios de Farmacologia: a base fisiopatológica da farmacoterapia. 2. ed. Rio de Janeiro: Guanabara Koogan; 2009. p.131-145.Golan DE, Tashjian Jr AH, Armstrong EJ, Armstrong AW. Princípios de Farmacologia: a base fisiopatológica da farmacoterapia. 3. ed. Rio de Janeiro: Guanabara Koogan; 2014.Marieb EM, Hoehn K. Anatomia e Fisiologia. 3. ed. Porto Alegre: Artmed; 2009.Carvalho-De-Souza JL. Análise do efeito inibitório do eugenol sobre canais para Na+ ativados por voltagem em neurônios sensitivos [tese]. São Paulo: Instituto de Ciências Biomédicas – USP; 2009.Andrade ED. Terapêutica medicamentosa em odontologia. 3. ed. Rio de Janeiro: Artes Médicas; 2014.Parise GK, Ferranti KN, Grando CP. Sais anestésicos utilizados na odontologia: revisão de literatura. J Oral Investig. 2017;6(1):75-84.Gordh T. Lidocaine: the origino of a modern local anesthetic. Anesthesiology. 2010;113(6):1433-37.Peñarrocha M, Sanchis BJM, Martínez GJM. Anestesia local em odontologia. Rio de Janeiro: Guanabara Koogan; 2008.DEF. Dicionário de especialidades farmacêuticas 2016. 44. ed. Rio de Janeiro: Publicações Científicas; 2016.Vasconcelos RJH, Nogueira RVB, Leal AKR, Oliveira CTV, Bezerra JGB. Alterações sistêmicas decorrente do uso da lidocaína e prilocaína na prática odontológica. Rev cir traumatol buco-maxilo-fac. 2002;1(2):13-19.Montan MF, Cogo K, Bergamaschi CC, Volpato MC, Andrade ED. Mortalidade relacionada ao uso de anestésicos locais em odontologia. RGO. 2007;55(2):197-202.Gaffen AS, Haas DA. Survey of local anesthetic use by ontario dentists. J Can Dent Assoc. 2009;75(9):649.Souza LMA, Ramacciato JC, Motta RHL. Uso de Anestésicos locais em pacientes idosos. RGO. 2011;59:25-30.Yagiela JA, Dowd FJ, Johnson BS, Mariotti AJ, Neidle EA. Farmacologia e terapêutica para dentistas. 6. ed. Rio de Janeiro: Elsevier; 2011.Mojumdar EH, Lyubartsev AP. Molecular dynamics simulations of local anesthetic articaine in a lipid bilayer. Biophys Chem. 2010;153(1):27-35.Maniglia-Ferreira C, Almeida-Gomes F, Carvalho-Sousa B, Barbosa AV, Lins CC, Souza FD et al. Clinical evaluation of three anesthetics in endodontics. Acta Odontol Latinoam. 2009;22(1):21-6.Moore PA, Doll B, Delie RA, Hersh EV, Korostoff J, Johnson S et al. Hemostatic and anesthetic efficacy of 4% articaine HCL with 1:200,00 epinephrine and 4% articaine HCL with 1:100,000 epinephrine when administrated intraorally for periodontal surgery. J Periodontol. 2007;78(2):247-53.Santos CF, Modena KC, Giglio FP, Sakai VT, Calvo AM, Colombini BL et al. Epinephrine concentration (1,100,00 or 1,200,000) does not affect the clinical efficacy of 4% articaine forlower third molar removal: a double-blind, randomized, cross study. J Oral Maxillofac Surg. 2007;65(12):2445-52.Massaro F. Lipossomal bupivacaine: a long-acting local anesthetic for postsurgical analgesia. Focus On. 2012; 47:213-26.Dantas MVM, Gabrielli MAC, Hochuli VE. Effect of mepivacaine 2% with adrenaline 1:100.000 in blood pressure. Rev Odontol UNESP. 2008;37(3):223-27.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography