Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 63(9): 4562-4578, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32045235

RESUMEN

The main protease of coronaviruses and the 3C protease of enteroviruses share a similar active-site architecture and a unique requirement for glutamine in the P1 position of the substrate. Because of their unique specificity and essential role in viral polyprotein processing, these proteases are suitable targets for the development of antiviral drugs. In order to obtain near-equipotent, broad-spectrum antivirals against alphacoronaviruses, betacoronaviruses, and enteroviruses, we pursued a structure-based design of peptidomimetic α-ketoamides as inhibitors of main and 3C proteases. Six crystal structures of protease-inhibitor complexes were determined as part of this study. Compounds synthesized were tested against the recombinant proteases as well as in viral replicons and virus-infected cell cultures; most of them were not cell-toxic. Optimization of the P2 substituent of the α-ketoamides proved crucial for achieving near-equipotency against the three virus genera. The best near-equipotent inhibitors, 11u (P2 = cyclopentylmethyl) and 11r (P2 = cyclohexylmethyl), display low-micromolar EC50 values against enteroviruses, alphacoronaviruses, and betacoronaviruses in cell cultures. In Huh7 cells, 11r exhibits three-digit picomolar activity against the Middle East Respiratory Syndrome coronavirus.


Asunto(s)
Antivirales/farmacología , Coronavirus/efectos de los fármacos , Enterovirus/efectos de los fármacos , Lactamas/farmacología , Peptidomiméticos/farmacología , Replicación Viral/efectos de los fármacos , Proteasas Virales 3C , Animales , Antivirales/síntesis química , Antivirales/metabolismo , Sitios de Unión , Línea Celular Tumoral , Chlorocebus aethiops , Coronavirus/enzimología , Proteasas 3C de Coronavirus , Cristalografía por Rayos X , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Diseño de Fármacos , Enterovirus/enzimología , Humanos , Lactamas/síntesis química , Lactamas/metabolismo , Peptidomiméticos/síntesis química , Peptidomiméticos/metabolismo , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacología , Unión Proteica , Células Vero , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/química , Proteínas Virales/metabolismo
2.
Antiviral Res ; 149: 58-74, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29128390

RESUMEN

The multi-domain non-structural protein 3 (Nsp3) is the largest protein encoded by the coronavirus (CoV) genome, with an average molecular mass of about 200 kD. Nsp3 is an essential component of the replication/transcription complex. It comprises various domains, the organization of which differs between CoV genera, due to duplication or absence of some domains. However, eight domains of Nsp3 exist in all known CoVs: the ubiquitin-like domain 1 (Ubl1), the Glu-rich acidic domain (also called "hypervariable region"), a macrodomain (also named "X domain"), the ubiquitin-like domain 2 (Ubl2), the papain-like protease 2 (PL2pro), the Nsp3 ectodomain (3Ecto, also called "zinc-finger domain"), as well as the domains Y1 and CoV-Y of unknown functions. In addition, the two transmembrane regions, TM1 and TM2, exist in all CoVs. The three-dimensional structures of domains in the N-terminal two thirds of Nsp3 have been investigated by X-ray crystallography and/or nuclear magnetic resonance (NMR) spectroscopy since the outbreaks of Severe Acute Respiratory Syndrome coronavirus (SARS-CoV) in 2003 as well as Middle-East Respiratory Syndrome coronavirus (MERS-CoV) in 2012. In this review, the structures and functions of these domains of Nsp3 are discussed in depth.


Asunto(s)
Coronaviridae/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Secuencia de Aminoácidos , Coronaviridae/genética , Glutamina/química , Humanos , Modelos Moleculares , Papaína/química , Papaína/metabolismo , Unión Proteica , Conformación Proteica , Dominios Proteicos , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad , Ubiquitina/química , Ubiquitina/metabolismo
3.
Viruses ; 9(9)2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28914805

RESUMEN

Hepatitis A virus (HAV) and hepatitis E virus (HEV) are causative agents of acute viral hepatitis transmitted via the fecal-oral route. Both viruses place a heavy burden on the public health and economy of developing countries. To test the possibility that HAV could be used as an expression vector for the development of a combination vaccine against hepatitis A and E infections, recombinant HAV-HEp148 was created as a vector to express an HEV neutralization epitope (HEp148) located at aa 459-606 of the HEV capsid protein. The recombinant virus expressed the HEp148 protein in a partially dimerized state in HAV-susceptible cells. Immunization with the HAV-HEp148 virus induced a strong HAV- and HEV-specific immune response in mice. Thus, the present study demonstrates a novel approach to the development of a combined hepatitis A and E vaccine.


Asunto(s)
Epítopos/inmunología , Virus de la Hepatitis A/genética , Virus de la Hepatitis A/inmunología , Anticuerpos Antihepatitis/biosíntesis , Virus de la Hepatitis E/inmunología , Vacunas contra Hepatitis Viral/inmunología , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Vectores Genéticos , Hepatitis A/inmunología , Hepatitis A/virología , Anticuerpos Antihepatitis/inmunología , Hepatitis E/inmunología , Hepatitis E/virología , Virus de la Hepatitis E/genética , Ratones , Pruebas de Neutralización , Vacunación , Vacunas Combinadas/administración & dosificación , Vacunas Combinadas/genética , Vacunas Combinadas/inmunología , Vacunas contra Hepatitis Viral/administración & dosificación , Vacunas contra Hepatitis Viral/genética
4.
Proc Natl Acad Sci U S A ; 113(35): E5192-201, 2016 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-27519799

RESUMEN

Highly pathogenic severe acute respiratory syndrome coronavirus (SARS-CoV) has developed strategies to inhibit host immune recognition. We identify cellular E3 ubiquitin ligase ring-finger and CHY zinc-finger domain-containing 1 (RCHY1) as an interacting partner of the viral SARS-unique domain (SUD) and papain-like protease (PL(pro)), and, as a consequence, the involvement of cellular p53 as antagonist of coronaviral replication. Residues 95-144 of RCHY1 and 389-652 of SUD (SUD-NM) subdomains are crucial for interaction. Association with SUD increases the stability of RCHY1 and augments RCHY1-mediated ubiquitination as well as degradation of p53. The calcium/calmodulin-dependent protein kinase II delta (CAMK2D), which normally influences RCHY1 stability by phosphorylation, also binds to SUD. In vivo phosphorylation shows that SUD does not regulate phosphorylation of RCHY1 via CAMK2D. Similarly to SUD, the PL(pro)s from SARS-CoV, MERS-CoV, and HCoV-NL63 physically interact with and stabilize RCHY1, and thus trigger degradation of endogenous p53. The SARS-CoV papain-like protease is encoded next to SUD within nonstructural protein 3. A SUD-PL(pro) fusion interacts with RCHY1 more intensively and causes stronger p53 degradation than SARS-CoV PL(pro) alone. We show that p53 inhibits replication of infectious SARS-CoV as well as of replicons and human coronavirus NL63. Hence, human coronaviruses antagonize the viral inhibitor p53 via stabilizing RCHY1 and promoting RCHY1-mediated p53 degradation. SUD functions as an enhancer to strengthen interaction between RCHY1 and nonstructural protein 3, leading to a further increase in in p53 degradation. The significance of these findings is that down-regulation of p53 as a major player in antiviral innate immunity provides a long-sought explanation for delayed activities of respective genes.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Síndrome Respiratorio Agudo Grave/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo , Sitios de Unión/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteasas 3C de Coronavirus , Cisteína Endopeptidasas/genética , Regulación hacia Abajo , Interacciones Huésped-Patógeno , Humanos , Unión Proteica , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Síndrome Respiratorio Agudo Grave/genética , Síndrome Respiratorio Agudo Grave/virología , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas Virales/genética , Replicación Viral/genética
5.
Virology ; 484: 313-322, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26149721

RESUMEN

The multi-domain non-structural protein 3 of SARS-coronavirus is a component of the viral replication/transcription complex (RTC). Among other domains, it contains three sequentially arranged macrodomains: the X domain and subdomains SUD-N as well as SUD-M within the "SARS-unique domain". The X domain was proposed to be an ADP-ribose-1"-phosphatase or a poly(ADP-ribose)-binding protein, whereas SUD-NM binds oligo(G)-nucleotides capable of forming G-quadruplexes. Here, we describe the application of a reverse genetic approach to assess the importance of these macrodomains for the activity of the SARS-CoV RTC. To this end, Renilla luciferase-encoding SARS-CoV replicons with selectively deleted macrodomains were constructed and their ability to modulate the RTC activity was examined. While the SUD-N and the X domains were found to be dispensable, the SUD-M domain was crucial for viral genome replication/transcription. Moreover, alanine replacement of charged amino-acid residues of the SUD-M domain, which are likely involved in G-quadruplex-binding, caused abrogation of RTC activity.


Asunto(s)
Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Transcripción Genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Sustitución de Aminoácidos , Genes Reporteros , Luciferasas de Renilla/análisis , Luciferasas de Renilla/genética , Estructura Terciaria de Proteína , Genética Inversa , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Eliminación de Secuencia , Coloración y Etiquetado , Proteínas no Estructurales Virales/genética
6.
Hepatology ; 62(2): 397-408, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25866017

RESUMEN

UNLABELLED: Hepatitis A virus (HAV) and hepatitis C virus (HCV) are two positive-strand RNA viruses sharing a similar biology, but causing opposing infection outcomes, with HAV always being cleared and HCV establishing persistence in the majority of infections. To gain deeper insight into determinants of replication, persistence, and treatment, we established a homogenous cell-culture model allowing a thorough comparison of RNA replication of both viruses. By screening different human liver-derived cell lines with subgenomic reporter replicons of HAV as well as of different HCV genotypes, we found that Huh7-Lunet cells supported HAV- and HCV-RNA replication with similar efficiency and limited interference between both replicases. HAV and HCV replicons were similarly sensitive to interferon (IFN), but differed in their ability to establish persistent replication in cell culture. In contrast to HCV, HAV replicated independently from microRNA-122 and phosphatidylinositol 4-kinase IIIα and ß (PI4KIII). Both viruses were efficiently inhibited by cyclosporin A and NIM811, a nonimmunosuppressive analog thereof, suggesting an overlapping dependency on cyclophilins for replication. However, analysis of a broader set of inhibitors revealed that, in contrast to HCV, HAV does not depend on cyclophilin A, but rather on adenosine-triphosphate-binding cassette transporters and FK506-binding proteins. Finally, silibinin, but not its modified intravenous formulation, efficiently inhibited HAV genome replication in vitro, suggesting oral silibinin as a potential therapeutic option for HAV infections. CONCLUSION: We established a cell-culture model enabling comparative studies on RNA replication of HAV and HCV in a homogenous cellular background with comparable replication efficiency. We thereby identified new host cell targets and potential treatment options for HAV and set the ground for future studies to unravel determinants of clearance and persistence.


Asunto(s)
Hepacivirus/genética , Virus de la Hepatitis A/genética , Interferones/farmacología , ARN Viral/genética , Replicación Viral/genética , Células Cultivadas , Femenino , Hepacivirus/efectos de los fármacos , Hepacivirus/fisiología , Hepatitis A/tratamiento farmacológico , Virus de la Hepatitis A/efectos de los fármacos , Virus de la Hepatitis A/fisiología , Hepatitis B/tratamiento farmacológico , Humanos , Interferones/uso terapéutico , Masculino , ARN Viral/efectos de los fármacos , Sensibilidad y Especificidad , Replicación Viral/efectos de los fármacos
7.
Antiviral Res ; 103: 17-24, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24393668

RESUMEN

Proteolytical cleavage of the picornaviral polyprotein is essential for viral replication. Therefore, viral proteases are attractive targets for anti-viral therapy. Most assays available for testing proteolytical activity of proteases are performed in vitro, using heterologously expressed proteases and peptide substrates. To deal with the disadvantages associated with in vitro assays, we modified a cell-based protease assay for picornavirus proteases. The assay is based on the induction of expression of a firefly luciferase reporter by a chimeric transcription factor in which the viral protease and cleavage sites are inserted between the GAL4 binding domain and the VP16 activation domain. Firefly luciferase expression is dependent on cleavage of the transcription factor by the viral protease. This biosafe assay enables testing the effect of compounds on protease activity in cells while circumventing the need for infection. We designed the assay for 3C proteases (3C(pro)) of various enteroviruses as well as of viruses of several other picornavirus genera, and show that the assay is amenable for use in a high-throughput setting. Furthermore, we show that the spectrum of activity of 3C(pro) inhibitor AG7088 (rupintrivir) not only encompasses enterovirus 3C(pro) but also 3C(pro) of foot-and-mouth disease virus (FMDV), an aphthovirus. In contrary, AG7404 (compound 1), an analogue of AG7088, had no effect on FMDV 3C(pro) activity, for which we provide a structural explanation.


Asunto(s)
Antivirales/aislamiento & purificación , Antivirales/farmacología , Evaluación Preclínica de Medicamentos/métodos , Picornaviridae/efectos de los fármacos , Picornaviridae/enzimología , Proteínas Virales/antagonistas & inhibidores , Proteasas Virales 3C , Animales , Línea Celular , Cisteína Endopeptidasas , Genes Reporteros , Humanos , Luciferasas de Luciérnaga/análisis , Luciferasas de Luciérnaga/genética , Inhibidores de Proteasas/aislamiento & purificación , Inhibidores de Proteasas/farmacología
8.
J Virol ; 87(8): 4339-51, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23388726

RESUMEN

We have determined the cleavage specificity and the crystal structure of the 3C protease of enterovirus 68 (EV68 3C(pro)). The protease exhibits a typical chymotrypsin fold with a Cys...His...Glu catalytic triad; its three-dimensional structure is closely related to that of the 3C(pro) of rhinovirus 2, as well as to that of poliovirus. The phylogenetic position of the EV68 3C(pro) between the corresponding enzymes of rhinoviruses on the one hand and classical enteroviruses on the other prompted us to use the crystal structure for the design of irreversible inhibitors, with the goal of discovering broad-spectrum antiviral compounds. We synthesized a series of peptidic α,ß-unsaturated ethyl esters of increasing length and for each inhibitor candidate, we determined a crystal structure of its complex with the EV68 3C(pro), which served as the basis for the next design round. To exhibit inhibitory activity, compounds must span at least P3 to P1'; the most potent inhibitors comprise P4 to P1'. Inhibitory activities were found against the purified 3C protease of EV68, as well as with replicons for poliovirus and EV71 (50% effective concentration [EC(50)] = 0.5 µM for the best compound). Antiviral activities were determined using cell cultures infected with EV71, poliovirus, echovirus 11, and various rhinovirus serotypes. The most potent inhibitor, SG85, exhibited activity with EC(50)s of ≈180 nM against EV71 and ≈60 nM against human rhinovirus 14 in a live virus-cell-based assay. Even the shorter SG75, spanning only P3 to P1', displayed significant activity (EC(50) = 2 to 5 µM) against various rhinoviruses.


Asunto(s)
Antivirales/farmacología , Drogas de Diseño/farmacología , Picornaviridae/efectos de los fármacos , Picornaviridae/enzimología , Inhibidores de Proteasas/farmacología , Proteínas Virales/antagonistas & inhibidores , Proteasas Virales 3C , Antivirales/química , Línea Celular , Cristalografía por Rayos X , Cisteína Endopeptidasas/química , Drogas de Diseño/química , Diseño de Fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Inhibidores de Proteasas/química , Conformación Proteica , Proteínas Virales/química
9.
PLoS One ; 7(12): e52065, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23272212

RESUMEN

Translation directed by several picornavirus IRES elements can usually take place after cleavage of eIF4G by picornavirus proteases 2A(pro) or L(pro). The hepatitis A virus (HAV) IRES is thought to be an exception to this rule because it requires intact eIF4F complex for translation. In line with previous results we report that poliovirus (PV) 2A(pro) strongly blocks protein synthesis directed by HAV IRES. However, in contrast to previous findings we now demonstrate that eIF4G cleavage by foot-and-mouth disease virus (FMDV) L(pro) strongly stimulates HAV IRES-driven translation. Thus, this is the first observation that 2A(pro) and L(pro) exhibit opposite effects to what was previously thought to be the case in HAV IRES. This effect has been observed both in hamster BHK and human hepatoma Huh7 cells. In addition, this stimulation of translation is also observed in cell free systems after addition of purified L(pro). Notably, in presence of this FMDV protease, translation directed by HAV IRES takes place when eIF2α has been inactivated by phosphorylation. Our present findings clearly demonstrate that protein synthesis directed by HAV IRES can occur when eIF4G has been cleaved and after inactivation of eIF2. Therefore, translation directed by HAV IRES without intact eIF4G and active eIF2 is similar to that observed with other picornavirus IRESs.


Asunto(s)
Factor 2 Eucariótico de Iniciación/metabolismo , Factor 4F Eucariótico de Iniciación/metabolismo , Virus de la Hepatitis A/genética , Biosíntesis de Proteínas , ARN Viral , Regiones no Traducidas 5' , Animales , Línea Celular , Sistema Libre de Células , Cricetinae , Virus de la Fiebre Aftosa/genética , Humanos , Poliovirus/genética , Proteolisis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transfección
10.
Nucleic Acids Res ; 40(7): 3042-55, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22156376

RESUMEN

Oligonucleotides as short as 6 nt in length have been shown to bind specifically and tightly to proteins and affect their biological function. Yet, sparse structural data are available for corresponding complexes. Employing a recently developed hexanucleotide array, we identified hexadeoxyribonucleotides that bind specifically to the 3C protease of hepatitis A virus (HAV 3C(pro)). Inhibition assays in vitro identified the hexanucleotide 5'-GGGGGT-3' (G(5)T) as a 3C(pro) protease inhibitor. Using (1)H NMR spectroscopy, G(5)T was found to form a G-quadruplex, which might be considered as a minimal aptamer. With the help of (1)H, (15)N-HSQC experiments the binding site for G(5)T was located to the C-terminal ß-barrel of HAV 3C(pro). Importantly, the highly conserved KFRDI motif, which has previously been identified as putative viral RNA binding site, is not part of the G(5)T-binding site, nor does G(5)T interfere with the binding of viral RNA. Our findings demonstrate that sequence-specific nucleic acid-protein interactions occur with oligonucleotides as small as hexanucleotides and suggest that these compounds may be of pharmaceutical relevance.


Asunto(s)
Cisteína Endopeptidasas/química , Desoxirribonucleótidos/química , Virus de la Hepatitis A/enzimología , Proteínas Virales/química , Proteasas Virales 3C , Antivirales/química , Sitios de Unión , Cisteína Endopeptidasas/metabolismo , Desoxirribonucleótidos/metabolismo , Dimerización , G-Cuádruplex , Resonancia Magnética Nuclear Biomolecular , ARN Viral/metabolismo , Proteínas Virales/metabolismo
11.
PLoS Pathog ; 5(5): e1000428, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19436709

RESUMEN

Since the outbreak of severe acute respiratory syndrome (SARS) in 2003, the three-dimensional structures of several of the replicase/transcriptase components of SARS coronavirus (SARS-CoV), the non-structural proteins (Nsps), have been determined. However, within the large Nsp3 (1922 amino-acid residues), the structure and function of the so-called SARS-unique domain (SUD) have remained elusive. SUD occurs only in SARS-CoV and the highly related viruses found in certain bats, but is absent from all other coronaviruses. Therefore, it has been speculated that it may be involved in the extreme pathogenicity of SARS-CoV, compared to other coronaviruses, most of which cause only mild infections in humans. In order to help elucidate the function of the SUD, we have determined crystal structures of fragment 389-652 ("SUD(core)") of Nsp3, which comprises 264 of the 338 residues of the domain. Both the monoclinic and triclinic crystal forms (2.2 and 2.8 A resolution, respectively) revealed that SUD(core) forms a homodimer. Each monomer consists of two subdomains, SUD-N and SUD-M, with a macrodomain fold similar to the SARS-CoV X-domain. However, in contrast to the latter, SUD fails to bind ADP-ribose, as determined by zone-interference gel electrophoresis. Instead, the entire SUD(core) as well as its individual subdomains interact with oligonucleotides known to form G-quadruplexes. This includes oligodeoxy- as well as oligoribonucleotides. Mutations of selected lysine residues on the surface of the SUD-N subdomain lead to reduction of G-quadruplex binding, whereas mutations in the SUD-M subdomain abolish it. As there is no evidence for Nsp3 entering the nucleus of the host cell, the SARS-CoV genomic RNA or host-cell mRNA containing long G-stretches may be targets of SUD. The SARS-CoV genome is devoid of G-stretches longer than 5-6 nucleotides, but more extended G-stretches are found in the 3'-nontranslated regions of mRNAs coding for certain host-cell proteins involved in apoptosis or signal transduction, and have been shown to bind to SUD in vitro. Therefore, SUD may be involved in controlling the host cell's response to the viral infection. Possible interference with poly(ADP-ribose) polymerase-like domains is also discussed.


Asunto(s)
G-Cuádruplex , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/química , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Electroforesis , Genoma Viral , Lisina/metabolismo , Datos de Secuencia Molecular , Mutación , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Multimerización de Proteína , Estructura Terciaria de Proteína , ARN Polimerasa Dependiente del ARN/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Proteínas no Estructurales Virales/genética , Replicación Viral
12.
BMB Rep ; 41(9): 678-83, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18823593

RESUMEN

The initial step during assembly of the hepatitis A virus particle is driven by domain 2A of P1-2A, which is the precursor of the structural proteins. The proteolytic removal of 2A from particulate VP1-2A by an as yet unknown host enzyme presumably terminates viral morphogenesis. Using a genetic approach, we show that a basic amino acid residue at the C-terminus of VP1 is required for efficient particle assembly and that host proteases trypsin and cathepsin L remove 2A from hepatitis A virus particles in vitro. Analyses of insertion mutants in the C-terminus of 2A reveal that this part of 2A is important for liberation of P1-2A from the polyprotein. The data provide the first evidence that the VP1/2A junction is involved in both viral particle assembly and maturation and, therefore, seems to coordinate the first and last steps in viral morphogenesis.


Asunto(s)
Virus de la Hepatitis A/fisiología , Precursores de Proteínas/metabolismo , Proteínas Estructurales Virales/metabolismo , Ensamble de Virus/fisiología , Secuencia de Aminoácidos , Línea Celular , Virus de la Hepatitis A/genética , Virus de la Hepatitis A/ultraestructura , Humanos , Datos de Secuencia Molecular , Mutación , Péptido Hidrolasas/metabolismo , Precursores de Proteínas/genética , Estructura Terciaria de Proteína , Proteínas Estructurales Virales/genética , Virión/metabolismo , Replicación Viral
13.
Biochem Biophys Res Commun ; 368(4): 1014-9, 2008 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-18282467

RESUMEN

The human RNA-binding protein La, is an essential trans-acting factor in IRES-dependent translation initiation of poliovirus, the prototypic picornavirus. For hepatitis A virus (HAV), an unusual member of this virus family, the role of host proteins in its inefficient translation and slow replication is unclear. Using small interfering RNA in vivo and purified La in vitro, we demonstrate for the first time that La suppresses HAV IRES-mediated translation and replication. We show that La binds specifically to distinct parts of the HAV IRES and that-unlike poliovirus-HAV proteinase 3C does not cleave La. The La-mediated suppression of HAV translation and stimulation of poliovirus translation implies unexpected mechanistic differences between viral IRES elements.


Asunto(s)
Autoantígenos/fisiología , Virus de la Hepatitis A/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Ribonucleoproteínas/fisiología , Ribosomas/fisiología , Proteasas Virales 3C , Autoantígenos/metabolismo , Línea Celular Tumoral , Cisteína Endopeptidasas/metabolismo , Humanos , Interferencia de ARN , Ribonucleoproteínas/metabolismo , Proteínas Virales/metabolismo , Antígeno SS-B
14.
Biochem Biophys Res Commun ; 364(4): 877-82, 2007 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-17976532

RESUMEN

Caused by a new coronavirus, severe acute respiratory syndrome (SARS) is a highly contagious disease associated with significant fatality that emerged in 2003. The molecular cause of the unusually high human pathogenicity of the SARS coronavirus (SARS-CoV) is still unknown. In an effort to characterize molecular components of the virus that are absent in other coronaviruses, all of which are considerably less pathogenic for humans, we recombinantly produced the SARS-unique domain (SUD) within non-structural protein 3 (Nsp3) of SARS-CoV and characterized its nucleic-acid binding properties. Zone-interference gel electrophoresis and electrophoretic mobility shift assays revealed a specific affinity of SUD for oligo(G)-strings. A few such segments are present in the SARS-CoV genome, but also in mRNAs of host proteins involved in the regulation of signaling pathways. A putative role of SUD in virus-induced apoptosis or survival of host cells is discussed.


Asunto(s)
Nucleótidos de Guanina/química , Nucleótidos de Guanina/metabolismo , Oligorribonucleótidos/química , Oligorribonucleótidos/metabolismo , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Estructura Terciaria de Proteína
15.
Biochem Biophys Res Commun ; 364(4): 725-30, 2007 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-17967440

RESUMEN

The poly(rC)-binding protein PCBP2 has multiple functions in post-transcriptional control of host and viral gene expression. Since it interacts with picornaviral RNA structures, it was proposed that PCBP2 regulates viral genome translation and replication. The hepatitis A virus (HAV), an atypical picornavirus, contains an unusual pyrimidine-rich tract (pY1) with unknown functions. Using in vivo and in vitro assays, we provide direct evidence that PCBP2 interacts with pY1 and that binding is mediated by KH domains 1 and 3. Proteolytic cleavage by the viral protease 3C generates a C-terminally truncated polypeptide with highly reduced RNA affinity. The results suggest that during HAV infection PCBP2 cleavage might specifically down-regulate viral protein synthesis, thereby giving way to viral RNA synthesis.


Asunto(s)
Virus de la Hepatitis A/química , Virus de la Hepatitis A/enzimología , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , ARN/química , ARN/metabolismo , Sitios de Unión , Unión Proteica
16.
Nucleic Acids Res ; 35(17): 5975-84, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17726047

RESUMEN

Proteolytic cleavage of translation initiation factors is a means to interfere with mRNA circularization and to induce translation arrest during picornaviral replication or apoptosis. It was shown that the regulated cleavages of eukaryotic initiation factor (eIF) 4G and poly(A)-binding protein (PABP) by viral proteinases correlated with early and late arrest of host cap-dependent and viral internal ribosome entry site (IRES)-dependent translation, respectively. Here we show that in contrast to coxsackievirus, eIF4G is not a substrate of proteinase 3C of hepatitis A virus (HAV 3C(pro)). However, PABP is cleaved by HAV 3C(pro) in vitro and in vivo, separating the N-terminal RNA-binding domain (NTD) of PABP from the C-terminal protein-interaction domain. In vitro, NTD has a dominant negative effect on HAV IRES-dependent translation and an enhanced binding affinity to the RNA structural element pY1 in the 5' nontranslated region of the HAV RNA that is essential for viral genome replication. The results point to a regulatory role of PABP cleavage in RNA template switching of viral translation to RNA synthesis.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Regulación Viral de la Expresión Génica , Virus de la Hepatitis A/genética , Iniciación de la Cadena Peptídica Traduccional , Proteínas de Unión a Poli(A)/metabolismo , Proteínas Virales/metabolismo , Proteasas Virales 3C , Regiones no Traducidas 5'/química , Regiones no Traducidas 5'/metabolismo , Línea Celular Tumoral , Factor 4G Eucariótico de Iniciación/metabolismo , Virus de la Hepatitis A/enzimología , Humanos , Modelos Genéticos , Proteínas de Unión a Poli(A)/química , Proteínas de Unión a Poli(A)/fisiología , ARN Viral/química , ARN Viral/metabolismo , Eliminación de Secuencia
17.
Virus Res ; 130(1-2): 296-302, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17640757

RESUMEN

Hepatitis A virus (HAV) protein 2A has the capacity to harbor and expose a short foreign epitope. The chimeric virus, HAV-gp41, bearing seven amino acids of the 2F5 epitope of the HIV glycoprotein gp41, was shown to replicate in cell culture and laboratory animals and to induce a humoral immune response. As an extension of this work, we now investigated the possibility to insert longer epitopes, their impact on genetic stability, and the production of chimeric HAV. Twenty-seven amino acid residues of either HIV gp41, comprising the 2F5 epitope, or of a mimotope (F78) of the hypervariable region 1 of the hepatitis C virus (HCV) envelope protein E2 were inserted near the C-terminus of HAV 2A and viral capsid formation and replication were studied. The genome of the chimeric virus (HAV-F78) had reduced replication ability, yet the sedimentation profile of the chimeric particles was unchanged and the HCV sequence was maintained over serial viral passages. In contrast, no capsids were formed when an extended HIV epitope of 27 residues was inserted, precluding the rescue of infectious chimeric virus. Based on structural analyses, the data suggest that the isoelectric point (pI) and/or the secondary structure of the chimeric proteins are essential determinants that affect HAV particle formation for which protein 2A serves as an assembly signal.


Asunto(s)
Epítopos/inmunología , Virus de la Hepatitis A/fisiología , Ensamble de Virus , Replicación Viral , Animales , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/inmunología , Proteína gp41 de Envoltorio del VIH/genética , Proteína gp41 de Envoltorio del VIH/inmunología , Virus de la Hepatitis A/inmunología , Punto Isoeléctrico , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología
18.
Antiviral Res ; 73(2): 101-11, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17014915

RESUMEN

Its stable particle structure combined with its high immunogenicity makes the hepatitis A virus (HAV) a perfect carrier to expose foreign epitopes to the host immune system. In an earlier report [Beneduce, F., Kusov, Y., Klinger, M., Gauss-Müller, V., Morace, G., 2002. Chimeric hepatitis A virus particles presenting a foreign epitope (HIV gp41) at their surface. Antiviral Res. 55, 369-377] chimeric virus-like particles (HAV-gp41) were described that carried at their surface the dominant gp41 epitope 2F5 (2F5e) of the human immunodeficiency virus HIV-1. Extending this work, we now report that chimeric virus HAV-gp41 replicates in HAV-susceptible cells as well as in non-human primates. Infected marmosets developed both an anti-HAV and anti-2F5 epitope immune response. Furthermore, an HIV-neutralizing antibody response was elicited in guinea pigs immunized with HAV-gp41 chimeric particles. The results demonstrate that the replication-competent chimeric HAV-gp41 can serve as either a live or a subunit vaccine for eliciting of antibodies directed against a foreign antigenic epitope.


Asunto(s)
Proteína gp41 de Envoltorio del VIH/inmunología , VIH-1/inmunología , Virus de la Hepatitis A/inmunología , Vacunas contra el SIDA/inmunología , Vacunas contra el SIDA/farmacología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Callithrix , Línea Celular Tumoral , Epítopos/genética , Epítopos/inmunología , Cobayas , Proteína gp41 de Envoltorio del VIH/genética , VIH-1/genética , Vacunas contra la Hepatitis A/inmunología , Vacunas contra la Hepatitis A/farmacología , Virus de la Hepatitis A/genética , Virus de la Hepatitis A/fisiología , Humanos , Datos de Secuencia Molecular , Replicación Viral
19.
J Virol ; 80(11): 5599-610, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16699041

RESUMEN

Infection by hepatitis A virus (HAV) can cause acute hepatitis and, rarely, fulminant liver failure, in particular in patients chronically infected with hepatitis C virus. Based on our previous observation that small interfering RNAs (siRNAs) can silence translation and replication of the firefly luciferase-encoding HAV replicon, we now exploited this technology to demonstrate the effect of siRNAs on viral infection in Huh-7 cells. Freshly and persistently infected cells were transfected with siRNAs targeting various sites in the HAV nonstructural genes. Compared to a single application, consecutive siRNA transfections targeting multiple sequences in the viral genome resulted in a more efficient and sustained silencing effect than a single transfection. In most instances, multiple applications of a single siRNA led to the emergence of viral escape mutants with mutated target sites that rendered these genomes resistant to RNA interference (RNAi). Efficient and sustained suppression of the viral infectivity was achieved after consecutive applications of an siRNA targeting a computer-predicted hairpin structure. This siRNA holds promise as a therapeutic tool for severe courses of HAV infection. In addition, the results provide new insight into the structural bases for sequence-specific RNAi.


Asunto(s)
Silenciador del Gen/efectos de los fármacos , Hepacivirus/efectos de los fármacos , Hepatitis/prevención & control , Interferencia de ARN , ARN Interferente Pequeño/farmacología , ARN Viral/biosíntesis , Línea Celular Tumoral , Hepacivirus/genética , Hepatitis/genética , Hepatitis/metabolismo , Humanos , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , ARN Viral/genética , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
20.
Biochem Biophys Res Commun ; 330(4): 1217-23, 2005 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-15823573

RESUMEN

Small interfering RNAs (siRNAs) targeting the coding region of hepatitis A virus (HAV) were shown to specifically inhibit viral genome replication. Compared to the coding region, the HAV internal ribosomal entry site (IRES) in the 5' non-coding region is highly sequence-conserved and folds into stable secondary structures. Here, we report efficient and sustained RNA interference mediated by both RNase III-prepared siRNA (esiRNA) and vector-derived short hairpin RNAs (shRNAs) that are targeted to various domains of the HAV IRES. Using reporter constructs, and the DNA-based HAV replicon system, we found that shRNAs targeting the HAV IRES domains IIIc and V sustainably suppressed genome translation and replication whereas the IRES domains IIIa and IV were resistant to RNA interference. Our study suggests that some HAV IRES domains might be used as a universal and effective target for specific inhibition of HAV infection.


Asunto(s)
Virus de la Hepatitis A/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Secuencia de Bases , Vectores Genéticos , Genoma Viral , Virus de la Hepatitis A/genética , Virus de la Hepatitis A/fisiología , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Biosíntesis de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/síntesis química , ARN Interferente Pequeño/genética , ARN Viral/genética , Ribonucleasa III/química , Ribosomas/genética , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...