Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Intervalo de año de publicación
1.
mBio ; 9(3)2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29789363

RESUMEN

Viroporins are viral proteins with ion channel (IC) activity that play an important role in several processes, including virus replication and pathogenesis. While many coronaviruses (CoVs) encode two viroporins, severe acute respiratory syndrome CoV (SARS-CoV) encodes three: proteins 3a, E, and 8a. Additionally, proteins 3a and E have a PDZ-binding motif (PBM), which can potentially bind over 400 cellular proteins which contain a PDZ domain, making them potentially important for the control of cell function. In the present work, a comparative study of the functional motifs included within the SARS-CoV viroporins was performed, mostly focusing on the roles of the IC and PBM of E and 3a proteins. Our results showed that the full-length E and 3a proteins were required for maximal SARS-CoV replication and virulence, whereas viroporin 8a had only a minor impact on these activities. A virus missing both the E and 3a proteins was not viable, whereas the presence of either protein with a functional PBM restored virus viability. E protein IC activity and the presence of its PBM were necessary for virulence in mice. In contrast, the presence or absence of the homologous motifs in protein 3a did not influence virus pathogenicity. Therefore, dominance of the IC and PBM of protein E over those of protein 3a was demonstrated in the induction of pathogenesis in mice.IMPORTANCE Collectively, these results demonstrate key roles for the ion channel and PBM domains in optimal virus replication and pathogenesis and suggest that the viral viroporins and PBMs are suitable targets for antiviral therapy and for mutation in attenuated SARS-CoV vaccines.


Asunto(s)
Síndrome Respiratorio Agudo Grave/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Animales , Chlorocebus aethiops , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Células Vero , Proteínas del Envoltorio Viral/genética , Proteínas Virales/genética , Proteínas Viroporinas , Virulencia
2.
J Virol ; 91(5)2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27974570

RESUMEN

It has been shown previously in the severe acute respiratory syndrome coronavirus (SARS-CoV) that two point mutations, N15A and V25F, in the transmembrane domain (TMD) of the envelope (E) protein abolished channel activity and led to in vivo attenuation. Pathogenicity was recovered in mutants that also regained E protein channel activity. In particular, V25F was rapidly compensated by changes at multiple V25F-facing TMD residues located on a neighboring monomer, consistent with a recovery of oligomerization. Here, we show using infected cells that the same mutations, T16A and A26F, in the gamma-CoV infectious bronchitis virus (IBV) lead to, in principle, similar results. However, IBV E A26F did not abolish oligomer formation and was compensated by mutations at N- and C-terminal extramembrane domains (EMDs). The C-terminal EMD mutations clustered along an insertion sequence specific to gamma-CoVs. Nuclear magnetic resonance data are consistent with the presence of only one TMD in IBV E, suggesting that recovery of channel activity and fitness in these IBV E revertant mutants is through an allosteric interaction between EMDs and TMD. The present results are important for the development of IBV live attenuated vaccines when channel-inactivating mutations are introduced in the E protein.IMPORTANCE The ion channel activity of SARS-CoV E protein is a determinant of virulence, and abolishment of channel activity leads to viral attenuation. E deletion may be a strategy for generating live attenuated vaccines but can trigger undesirable compensatory mechanisms through modifications of other viral proteins to regain virulence. Therefore, a more suitable approach may be to introduce small but critical attenuating mutations. For this, the stability of attenuating mutations should be examined to understand the mechanisms of reversion. Here, we show that channel-inactivating mutations of the avian infectious bronchitis virus E protein introduced in a recombinant virus system are deficient in viral release and fitness and that revertant mutations also restored channel activity. Unexpectedly, most of the revertant mutations appeared at extramembrane domains, particularly along an insertion specific for gammacoronaviruses. Our structural data propose a single transmembrane domain in IBV E, suggesting an allosteric interaction between extramembrane and transmembrane domains.


Asunto(s)
Virus de la Bronquitis Infecciosa/fisiología , Canales Iónicos/genética , Proteínas del Envoltorio Viral/genética , Secuencia de Aminoácidos , Animales , Pollos , Chlorocebus aethiops , Secuencia Conservada , Canales Iónicos/química , Canales Iónicos/metabolismo , Potenciales de la Membrana , Mutación , Multimerización de Proteína , Células Vero , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Liberación del Virus
3.
Virology ; 485: 330-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26331680

RESUMEN

Severe acute respiratory syndrome coronavirus (SARS-CoV) envelope (E) protein is a viroporin involved in virulence. E protein ion channel (IC) activity is specifically correlated with enhanced pulmonary damage, edema accumulation and death. IL-1ß driven proinflammation is associated with those pathological signatures, however its link to IC activity remains unknown. In this report, we demonstrate that SARS-CoV E protein forms protein-lipid channels in ERGIC/Golgi membranes that are permeable to calcium ions, a highly relevant feature never reported before. Calcium ions together with pH modulated E protein pore charge and selectivity. Interestingly, E protein IC activity boosted the activation of the NLRP3 inflammasome, leading to IL-1ß overproduction. Calcium transport through the E protein IC was the main trigger of this process. These findings strikingly link SARS-CoV E protein IC induced ionic disturbances at the cell level to immunopathological consequences and disease worsening in the infected organism.


Asunto(s)
Calcio/metabolismo , Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Iones/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Transporte Biológico , Canales de Calcio/metabolismo , Chlorocebus aethiops , Mutación , Proteína con Dominio Pirina 3 de la Familia NLR , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Síndrome Respiratorio Agudo Grave/metabolismo , Síndrome Respiratorio Agudo Grave/virología , Células Vero , Proteínas del Envoltorio Viral/genética , Proteínas Viroporinas
4.
Virus Res ; 201: 61-6, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25733052

RESUMEN

The Middle East respiratory syndrome coronavirus (MERS-CoV) is a newly identified pathogen able of human transmission that causes a mortality of almost 40%. As in the case of SARS-CoV, MERS virus lacking E protein represents a potential vaccine. In both cases, abolishment of channel activity may be a contributor to the attenuation observed in E-deleted viruses. Herein, we report that purified MERS-CoV E protein, like SARS-CoV E protein, is almost fully α-helical, has a single α-helical transmembrane domain, and forms pentameric ion channels in lipid bilayers. Based on these similarities, and the proposed involvement of channel activity as virulence factor in SARS-CoV E protein, MERS-CoV E protein may constitute a potential drug target.


Asunto(s)
Canales Iónicos/química , Canales Iónicos/metabolismo , Coronavirus del Síndrome Respiratorio de Oriente Medio/química , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Multimerización de Proteína , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Humanos , Conformación Proteica
5.
J Virol ; 88(20): 11899-914, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25100835

RESUMEN

The small hydrophobic (SH) protein is a 64-amino-acid polypeptide encoded by the human respiratory syncytial virus (hRSV). SH protein has a single α-helical transmembrane (TM) domain that forms pentameric ion channels. Herein, we report the first inhibitor of the SH protein channel, pyronin B, and we have mapped its binding site to a conserved surface of the RSV SH pentamer, at the C-terminal end of the transmembrane domain. The validity of the SH protein structural model used has been confirmed by using a bicellar membrane-mimicking environment. However, in bicelles the α-helical stretch of the TM domain extends up to His-51, and by comparison with previous models both His-22 and His-51 adopt an interhelical/lumenal orientation relative to the channel pore. Neither His residue was found to be essential for channel activity although His-51 protonation reduced channel activity at low pH, with His-22 adopting a more structural role. The latter results are in contrast with previous patch clamp data showing channel activation at low pH, which could not be reproduced in the present work. Overall, these results establish a solid ground for future drug development targeting this important viroporin. Importance: The human respiratory syncytial virus (hRSV) is responsible for 64 million reported cases of infection and 160,000 deaths each year. Lack of adequate antivirals fuels the search for new targets for treatment. The small hydrophobic (SH) protein is a 64-amino-acid polypeptide encoded by hRSV and other paramyxoviruses, and its absence leads to viral attenuation in vivo and early apoptosis in infected cells. SH protein forms pentameric ion channels that may constitute novel drug targets, but no inhibitor for this channel activity has been reported so far. A small-molecule inhibitor, pyronin B, can reduce SH channel activity, and its likely binding site on the SH protein channel has been identified. Black lipid membrane (BLM) experiments confirm that protonation of both histidine residues reduces stability and channel activity. These results contrast with previous patch clamp data that showed low-pH activation, which we have not been able to reproduce.


Asunto(s)
Membrana Dobles de Lípidos , Virus Sincitiales Respiratorios/metabolismo , Proteínas Virales/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Sitios de Unión , Chlorocebus aethiops , Clonación Molecular , Genes Virales , Interacciones Hidrofóbicas e Hidrofílicas , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Técnicas de Placa-Clamp , Pironina/análogos & derivados , Pironina/farmacología , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/crecimiento & desarrollo , Células Vero , Ensayo de Placa Viral , Proteínas Virales/química , Proteínas Virales/genética
6.
PLoS Pathog ; 10(5): e1004077, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24788150

RESUMEN

Deletion of Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) envelope (E) gene attenuates the virus. E gene encodes a small multifunctional protein that possesses ion channel (IC) activity, an important function in virus-host interaction. To test the contribution of E protein IC activity in virus pathogenesis, two recombinant mouse-adapted SARS-CoVs, each containing one single amino acid mutation that suppressed ion conductivity, were engineered. After serial infections, mutant viruses, in general, incorporated compensatory mutations within E gene that rendered active ion channels. Furthermore, IC activity conferred better fitness in competition assays, suggesting that ion conductivity represents an advantage for the virus. Interestingly, mice infected with viruses displaying E protein IC activity, either with the wild-type E protein sequence or with the revertants that restored ion transport, rapidly lost weight and died. In contrast, mice infected with mutants lacking IC activity, which did not incorporate mutations within E gene during the experiment, recovered from disease and most survived. Knocking down E protein IC activity did not significantly affect virus growth in infected mice but decreased edema accumulation, the major determinant of acute respiratory distress syndrome (ARDS) leading to death. Reduced edema correlated with lung epithelia integrity and proper localization of Na+/K+ ATPase, which participates in edema resolution. Levels of inflammasome-activated IL-1ß were reduced in the lung airways of the animals infected with viruses lacking E protein IC activity, indicating that E protein IC function is required for inflammasome activation. Reduction of IL-1ß was accompanied by diminished amounts of TNF and IL-6 in the absence of E protein ion conductivity. All these key cytokines promote the progression of lung damage and ARDS pathology. In conclusion, E protein IC activity represents a new determinant for SARS-CoV virulence.


Asunto(s)
Canales Iónicos/fisiología , Síndrome Respiratorio Agudo Grave/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/crecimiento & desarrollo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Proteínas del Envoltorio Viral/fisiología , Secuencia de Aminoácidos , Animales , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Femenino , Interacciones Huésped-Patógeno/genética , Canales Iónicos/química , Canales Iónicos/genética , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Organismos Modificados Genéticamente , Estructura Terciaria de Proteína , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Células Vero , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
7.
Phys Chem Chem Phys ; 16(9): 3881-93, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24452437

RESUMEN

Ion channels are specialized proteins that enable the movement of charges through otherwise impermeable lipidic membranes. Their action is essential in living organisms facilitating electric signaling, muscle contraction or osmotic stress response among other effects. The protein and the lipid charges configure a polarized interface that yields local ionic concentrations and electric potentials that are very different from those of the bulk electrolyte. The combined effect of gradients of ionic concentration and electric potential causes the transport of ions through channels. Here we analyze charge regulation effects in different protein-lipid conformations, stressing how important is the role of electrostatic interactions in the ion channel function that traditionally has been rationalized paying attention mainly to changes in pore size. Tuning lipid charge combined with conductance and selectivity measurements is shown to be a complementary method to evidence lipid involvement in the structure of a biological ion channel.


Asunto(s)
Canales Iónicos/química , Lípidos/química , Alameticina/química , Alameticina/metabolismo , Antibacterianos/química , Antibacterianos/farmacología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Conductividad Eléctrica , Proteínas Hemolisinas/química , Proteínas Hemolisinas/metabolismo , Canales Iónicos/metabolismo , Transporte Iónico/efectos de los fármacos , Iones/química , Iones/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Electricidad Estática , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Proteínas Viroporinas
8.
Biochim Biophys Acta ; 1828(9): 2026-31, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23688394

RESUMEN

A partial characterization of the ion channels formed by the SARS coronavirus (CoV) envelope (E) protein was previously reported (C. Verdiá-Báguena et al., 2012 [12]). Here, we provide new significant insights on the involvement of lipids in the structure and function of the CoV E protein channel on the basis of three series of experiments. First, reversal potential measurements over a wide range of pH allow the dissection of the contributions to channel selectivity coming from ionizable residues of the protein transmembrane domain and also from the negatively charged groups of diphytanoyl phosphatidylserine (DPhPS) lipid. The corresponding effective pKas are consistent with the model pKas of the acidic residue candidates for titration. Second, the change of channel conductance with salt concentration reveals two distinct regimes (Donnan-controlled electrodiffusion and bulk-like electrodiffusion) fully compatible with the outcomes of selectivity experiments. Third, by measuring channel conductance in mixtures of neutral diphytanoyl phosphatidylcholine (DPhPC) lipids and negatively charged DPhPS lipids in low and high salt concentrations we conclude that the protein-lipid conformation in the channel is likely the same in charged and neutral lipids. Overall, the whole set of experiments supports the proteolipidic structure of SARS-CoV E channels and explains the large difference in channel conductance observed between neutral and charged membranes.


Asunto(s)
Canales Iónicos/química , Membrana Dobles de Lípidos/química , Potasio/química , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/química , Proteínas del Envoltorio Viral/química , Secuencia de Aminoácidos , Concentración de Iones de Hidrógeno , Transporte Iónico , Potenciales de la Membrana , Modelos Moleculares , Datos de Secuencia Molecular , Fosfatidilcolinas/química , Fosfatidilserinas/química , Estructura Terciaria de Proteína , Electricidad Estática , Proteínas Viroporinas
9.
Virology ; 432(2): 485-94, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-22832120

RESUMEN

Coronavirus (CoV) envelope (E) protein ion channel activity was determined in channels formed in planar lipid bilayers by peptides representing either the transmembrane domain of severe acute respiratory syndrome CoV (SARS-CoV) E protein, or the full-length E protein. Both of them formed a voltage independent ion conductive pore with symmetric ion transport properties. Mutations N15A and V25F located in the transmembrane domain prevented the ion conductivity. E protein derived channels showed no cation preference in non-charged lipid membranes, whereas they behaved as pores with mild cation selectivity in negatively-charged lipid membranes. The ion conductance was also controlled by the lipid composition of the membrane. Lipid charge also regulated the selectivity of a HCoV-229E E protein derived peptide. These results suggested that the lipids are functionally involved in E protein ion channel activity, forming a protein-lipid pore, a novel concept for CoV E protein ion channel entity.


Asunto(s)
Coronavirus Humano 229E/metabolismo , Canales Iónicos/metabolismo , Lípidos de la Membrana/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Proteínas del Envoltorio Viral/química , Secuencia de Aminoácidos , Humanos , Datos de Secuencia Molecular , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Proteínas Viroporinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA