Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
J Mol Recognit ; 31(9): e2718, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29687510

RESUMEN

The outer capsid spike protein VP4 of rotaviruses is a major determinant of infectivity and serotype specificity. Proteolytic cleavage of VP4 into 2 domains, VP8* and VP5*, enhances rotaviral infectivity. Interactions between the VP4 carbohydrate-binding domain (VP8*) and cell surface glycoconjugates facilitate initial virus-cell attachment and subsequent cell entry. Our saturation transfer difference nuclear magnetic resonance (STD NMR) and isothermal titration calorimetry (ITC) studies demonstrated that VP8*64-224 of canine rotavirus strain K9 interacts with N-acetylneuraminic and N-glycolylneuraminic acid derivatives, exhibiting comparable binding epitopes to VP8* from other neuraminidase-sensitive animal rotaviruses from pigs (CRW-8), cattle (bovine Nebraska calf diarrhoea virus, NCDV), and Rhesus monkeys (Simian rhesus rotavirus, RRV). Importantly, evidence was obtained for a preference by K9 rotavirus for the N-glycolyl- over the N-acetylneuraminic acid derivative. This indicates that a VP4 serotype 5A rotavirus (such as K9) can exhibit a neuraminic acid receptor preference that differs from that of a serotype 5B rotavirus (such as RRV) and the receptor preference of rotaviruses can vary within a particular VP4 genotype.


Asunto(s)
Proteínas de la Cápside/genética , Ácidos Neuramínicos/química , Rotavirus/genética , Proteínas no Estructurales Virales/genética , Animales , Proteínas de la Cápside/química , Carbohidratos/química , Carbohidratos/genética , Bovinos , Perros , Epítopos/genética , Epítopos/inmunología , Unión Proteica/genética , Dominios Proteicos/genética , Rotavirus/química , Porcinos , Proteínas no Estructurales Virales/química
2.
J Biol Chem ; 290(19): 12403-14, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25814665

RESUMEN

Human rotavirus is the leading cause of severe gastroenteritis in infants and children under the age of 5 years in both developed and developing countries. Human lactadherin, a milk fat globule membrane glycoprotein, inhibits human rotavirus infection in vitro, whereas bovine lactadherin is not active. Moreover, it protects breastfed infants against symptomatic rotavirus infections. To explore the potential antiviral activity of lactadherin sourced by equines, we undertook a proteomic analysis of milk fat globule membrane proteins from donkey milk and elucidated its amino acid sequence. Alignment of the human, bovine, and donkey lactadherin sequences revealed the presence of an Asp-Gly-Glu (DGE) α2ß1 integrin-binding motif in the N-terminal domain of donkey sequence only. Because integrin α2ß1 plays a critical role during early steps of rotavirus host cell adhesion, we tested a minilibrary of donkey lactadherin-derived peptides containing DGE sequence for anti-rotavirus activity. A 20-amino acid peptide containing both DGE and RGD motifs (named pDGE-RGD) showed the greatest activity, and its mechanism of antiviral action was characterized; pDGE-RGD binds to integrin α2ß1 by means of the DGE motif and inhibits rotavirus attachment to the cell surface. These findings suggest the potential anti-rotavirus activity of equine lactadherin and support the feasibility of developing an anti-rotavirus peptide that acts by hindering virus-receptor binding.


Asunto(s)
Antígenos de Superficie/química , Glucolípidos/química , Glicoproteínas/química , Glicoproteínas de Membrana/química , Proteínas de la Leche/química , Péptidos/química , Infecciones por Rotavirus/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Bovinos , Membrana Celular/metabolismo , Supervivencia Celular , Equidae , Caballos , Humanos , Concentración 50 Inhibidora , Integrinas/química , Gotas Lipídicas , Leche , Datos de Secuencia Molecular , Proteómica , Rotavirus/metabolismo , Infecciones por Rotavirus/tratamiento farmacológico , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
3.
PLoS Pathog ; 10(3): e1003998, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24676425

RESUMEN

It has been proposed that rotavirus infection promotes the progression of genetically-predisposed children to type 1 diabetes, a chronic autoimmune disease marked by infiltration of activated lymphocytes into pancreatic islets. Non-obese diabetic (NOD) mice provide a model for the human disease. Infection of adult NOD mice with rhesus monkey rotavirus (RRV) accelerates diabetes onset, without evidence of pancreatic infection. Rather, RRV spreads to the pancreatic and mesenteric lymph nodes where its association with antigen-presenting cells, including dendritic cells, induces cellular maturation. RRV infection increases levels of the class I major histocompatibility complex on B cells and proinflammatory cytokine expression by T cells at these sites. In autoimmunity-resistant mice and human mononuclear cells from blood, rotavirus-exposed plasmacytoid dendritic cells contribute to bystander polyclonal B cell activation through type I interferon expression. Here we tested the hypothesis that rotavirus induces bystander activation of lymphocytes from NOD mice by provoking dendritic cell activation and proinflammatory cytokine secretion. NOD mouse splenocytes were stimulated with rotavirus and assessed for activation by flow cytometry. This stimulation activated antigen-presenting cells and B cells independently of virus strain and replicative ability. Instead, activation depended on virus dose and was prevented by blockade of virus decapsidation, inhibition of endosomal acidification and interference with signaling through Toll-like receptor 7 and the type I interferon receptor. Plasmacytoid dendritic cells were more efficiently activated than conventional dendritic cells by RRV, and contributed to the activation of B and T cells, including islet-autoreactive CD8+ T cells. Thus, a double-stranded RNA virus can induce Toll-like receptor 7 signaling, resulting in lymphocyte activation. Our findings suggest that bystander activation mediated by type I interferon contributes to the lymphocyte activation observed following RRV infection of NOD mice, and may play a role in diabetes acceleration by rotavirus.


Asunto(s)
Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/virología , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/inmunología , Infecciones por Rotavirus/inmunología , Transducción de Señal , Receptor Toll-Like 7/inmunología , Animales , Diabetes Mellitus Tipo 1/inmunología , Citometría de Flujo , Interferones/biosíntesis , Interferones/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Rotavirus/inmunología , Transducción de Señal/inmunología
4.
Diabetologia ; 58(6): 1149-59, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25794781

RESUMEN

Viruses are considered to be potential key modulators of type 1 diabetes mellitus, with several possible mechanisms proposed for their modes of action. Here we discuss the evidence for virus involvement, including pancreatic infection and the induction of T cell-mediated molecular mimicry. A particular focus of this review is the further possibility that virus infection triggers bystander activation of pre-existing autoreactive lymphocytes. In this scenario, the virus triggers dendritic cell maturation and proinflammatory cytokine secretion by engaging pattern recognition receptors. These proinflammatory cytokines provoke bystander autoreactive lymphocyte activation in the presence of cognate autoantigen, which leads to enhanced beta cell destruction. Importantly, this mechanism does not necessarily involve pancreatic virus infection, and its virally non-specific nature suggests that it might represent a means commonly employed by multiple viruses. The ability of viruses specifically associated with type 1 diabetes, including group B coxsackievirus, rotavirus and influenza A virus, to induce these responses is also examined. The elucidation of a mechanism shared amongst several viruses for accelerating progression to type 1 diabetes would facilitate the identification of important targets for disease intervention.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Activación de Linfocitos/inmunología , Animales , Autoantígenos/inmunología , Efecto Espectador , Citocinas/metabolismo , Células Dendríticas/citología , Diabetes Mellitus Tipo 1/virología , Enterovirus/inmunología , Humanos , Inflamación , Virus de la Influenza A/inmunología , Ratones , Imitación Molecular , Páncreas/virología , Péptidos/química , Polimorfismo Genético , Receptores de Reconocimiento de Patrones/metabolismo , Rotavirus/inmunología , Linfocitos T/citología , Virosis/complicaciones , Virosis/inmunología
5.
J Gen Virol ; 96(Pt 7): 1768-76, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25701827

RESUMEN

Rotavirus is a leading cause of severe gastroenteritis in infants worldwide. Rotavirus nonstructural protein 1 (NSP1) is a virulence factor that inhibits innate host immune responses. NSP1 from some rotaviruses targets host interferon response factors (IRFs), leading to inhibition of type I interferon expression. A few rotaviruses encode an NSP1 that inhibits the NF-κB pathway by targeting ß-TrCP, a protein required for IκB degradation and NF-κB activation. Available evidence suggests that these NSP1 properties involve proteosomal degradation of target proteins. We show here that NSP1 from several human rotaviruses and porcine rotavirus CRW-8 inhibits the NF-κB pathway, but cannot degrade IRF3. Furthermore, ß-TrCP levels were much reduced in cells infected with these rotaviruses. This provides strong evidence that ß-TrCP degradation is required for NF-κB pathway inhibition by NSP1 and demonstrates the relevance of ß-TrCP degradation to rotavirus infection. C-terminal regions of NSP1, including a serine-containing motif resembling the ß-TrCP recognition motif of IκB, were required for NF-κB inhibition. CRW-8 infection of HT-29 intestinal epithelial cells induced significant levels of IFN-ß and CCL5 but not IL-8. This contrasts with monkey rotavirus SA11-4F, whose NSP1 inhibits IRF3 but not NF-κB. Substantial amounts of IL-8 but not IFN-ß or CCL5 were secreted from HT-29 cells infected with SA11-4F. Our results show that human rotaviruses commonly inhibit the NF-κB pathway by degrading ß-TrCP and thus stabilizing IκB. They suggest that NSP1 plays an important role during human rotavirus infection by inhibiting the expression of NF-κB-dependent cytokines, such as IL-8.


Asunto(s)
FN-kappa B/antagonistas & inhibidores , Rotavirus/inmunología , Proteínas no Estructurales Virales/metabolismo , Proteínas con Repetición de beta-Transducina/antagonistas & inhibidores , Animales , Línea Celular , Células Epiteliales/inmunología , Células Epiteliales/virología , Humanos , Evasión Inmune , Proteolisis , Porcinos
6.
J Gen Virol ; 96(12): 3519-3524, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26404393

RESUMEN

Specific roles have been ascribed to each of the 12 known rotavirus proteins apart from the non-structural protein 6 (NSP6). However, NSP6 may be present at sites of viral replication within the cytoplasm. Here we report that NSP6 from diverse species of rotavirus A localizes to mitochondria via conserved sequences in a predicted N-terminal a-helix. This suggests that NSP6 may affect mitochondrial functions during rotavirus infection.


Asunto(s)
Mitocondrias/fisiología , Rotavirus/metabolismo , Proteínas no Estructurales Virales/fisiología , Animales , Línea Celular , Regulación Viral de la Expresión Génica/fisiología , Células HEK293 , Humanos , Transporte de Proteínas , Rotavirus/genética , Replicación Viral
7.
Chembiochem ; 16(15): 2176-81, 2015 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-26250751

RESUMEN

Rotavirus-cell binding is the essential first step in rotavirus infection. This binding is a major determinant of rotavirus tropism, as host cell invasion is necessary to initiate infection. Initial rotavirus-cell interactions are mediated by carbohydrate-recognizing domain VP8* of the rotavirus capsid spike protein VP4. Here, we report the first observation of significant structural rearrangement of VP8* from human and animal rotavirus strains upon glycan receptor binding. The structural adaptability of rotavirus VP8* delivers important insights into how human and animal rotaviruses utilize the wider range of cellular glycans identified as VP8* binding partners. Furthermore, our studies on rotaviruses with atypical genetic makeup provide information expected to be critical for understanding the mechanisms of animal rotavirus gene emergence in humans and support implementation of epidemiologic surveillance of animal reservoirs as well as future vaccination schemes.


Asunto(s)
Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Especificidad del Huésped , Receptores Virales/metabolismo , Infecciones por Rotavirus/virología , Rotavirus/química , Secuencia de Aminoácidos , Animales , Carbohidratos/farmacología , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína/efectos de los fármacos , Receptores Virales/química , Rotavirus/genética , Rotavirus/metabolismo , Alineación de Secuencia
8.
J Virol ; 88(8): 4558-71, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24501414

RESUMEN

UNLABELLED: N-acetyl- and N-glycolylneuraminic acids (Sia) and α2ß1 integrin are frequently used by rotaviruses as cellular receptors through recognition by virion spike protein VP4. The VP4 subunit VP8*, derived from Wa rotavirus, binds the internal N-acetylneuraminic acid on ganglioside GM1. Wa infection is increased by enhanced internal Sia access following terminal Sia removal from main glycan chains with sialidase. The GM1 ligand cholera toxin B (CTB) reduces Wa infectivity. Here, we found sialidase treatment increased cellular GM1 availability and the infectivity of several other human (including RV-3) and animal rotaviruses, typically rendering them susceptible to methyl α-d-N-acetylneuraminide treatment, but did not alter α2ß1 usage. CTB reduced the infectivity of these viruses. Aceramido-GM1 inhibited Wa and RV-3 infectivity in untreated and sialidase-treated cells, and GM1 supplementation increased their infectivity, demonstrating the importance of GM1 for infection. Wa recognition of α2ß1 and internal Sia were at least partially independent. Rotavirus usage of GM1 was mapped to VP4 using virus reassortants, and RV-3 VP8* bound aceramido-GM1 by saturation transfer difference nuclear magnetic resonance (STD NMR). Most rotaviruses recognizing terminal Sia did not use GM1, including RRV. RRV VP8* interacted minimally with aceramido-GM1 by STD NMR. Unusually, TFR-41 rotavirus infectivity depended upon terminal Sia and GM1. Competition of CTB, Sia, and/or aceramido-GM1 with cell binding by VP8* from representative rotaviruses showed that rotavirus Sia and GM1 preferences resulted from VP8*-cell binding. Our major finding is that infection by human rotaviruses of commonly occurring VP4 serotypes involves VP8* binding to cell surface GM1 glycan, typically including the internal N-acetylneuraminic acid. IMPORTANCE: Rotaviruses, the major cause of severe infantile gastroenteritis, recognize cell surface receptors through virus spike protein VP4. Several animal rotaviruses are known to bind sialic acids at the termini of main carbohydrate chains. Conversely, only a single human rotavirus is known to bind sialic acid. Interestingly, VP4 of this rotavirus bound to sialic acid that forms a branch on the main carbohydrate chain of the GM1 ganglioside. Here, we use several techniques to demonstrate that other human rotaviruses exhibit similar GM1 usage properties. Furthermore, binding by VP4 to cell surface GM1, involving branched sialic acid recognition, is shown to facilitate infection. In contrast, most animal rotaviruses that bind terminal sialic acids did not utilize GM1 for VP4 cell binding or infection. These studies support a significant role for GM1 in mediating host cell invasion by human rotaviruses.


Asunto(s)
Gangliósidos/metabolismo , Integrina alfa2beta1/metabolismo , Ácidos Neuramínicos/metabolismo , Receptores Virales/metabolismo , Infecciones por Rotavirus/metabolismo , Rotavirus/fisiología , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Humanos , Integrina alfa2beta1/genética , Ácido N-Acetilneuramínico/metabolismo , Unión Proteica , Receptores Virales/genética , Rotavirus/genética , Infecciones por Rotavirus/genética , Infecciones por Rotavirus/virología
9.
J Gen Virol ; 95(Pt 8): 1723-1733, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24814927

RESUMEN

The importance of innate immunity to rotaviruses is exemplified by the range of strategies evolved by rotaviruses to interfere with the IFN response. We showed previously that rotaviruses block gene expression induced by type I and II IFNs, through a mechanism allowing activation of signal transducer and activator of transcription (STAT) 1 and STAT2 but preventing their nuclear accumulation. This normally occurs through activated STAT1/2 dimerization, enabling an interaction with importin α5 that mediates transport into the nucleus. In rotavirus-infected cells, STAT1/2 inhibition may limit the antiviral actions of IFN produced early in infection. Here we further analysed the block to STAT1/2 nuclear accumulation, showing that activated STAT1 accumulates in the cytoplasm in rotavirus-infected cells. STAT1/2 nuclear accumulation was inhibited by rotavirus even in the presence of the nuclear export inhibitor Leptomycin B, demonstrating that enhanced nuclear export is not involved in STAT1/2 cytoplasmic retention. The ability to inhibit STAT nuclear translocation was completely conserved amongst the group A rotaviruses tested, including a divergent avian strain. Analysis of mutant rotaviruses indicated that residues after amino acid 47 of NSP1 are dispensable for STAT inhibition. Furthermore, expression of any of the 12 Rhesus monkey rotavirus proteins did not inhibit IFN-stimulated STAT1 nuclear translocation. Finally, co-immunoprecipitation experiments from transfected epithelial cells showed that STAT1/2 binds importin α5 normally following rotavirus infection. These findings demonstrate that rotavirus probably employs a novel strategy to inhibit IFN-induced STAT signalling, which acts after STAT activation and binding to the nuclear import machinery.


Asunto(s)
Interacciones Huésped-Patógeno , Evasión Inmune , Interferones/antagonistas & inhibidores , Rotavirus/fisiología , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT2/antagonistas & inhibidores , Línea Celular , Humanos , Interferones/inmunología , Carioferinas/metabolismo , Rotavirus/inmunología
10.
J Gen Virol ; 94(Pt 6): 1151-1160, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23486667

RESUMEN

Rotavirus is a leading cause of severe dehydrating diarrhoea in infants and young children. Following rotavirus infection in the intestine an innate immune response is rapidly triggered. This response leads to the induction of type I and type III interferons (IFNs) and other cytokines, resulting in a reduction in viral replication. Here we review the current literature describing the detection of rotavirus infection by pattern recognition receptors within host cells, the subsequent molecular mechanisms leading to IFN and cytokine production, and the processes leading to reduced rotavirus replication and the development of protective immunity. Rotavirus countermeasures against innate responses, and their roles in modulating rotavirus replication in mice, also are discussed. By linking these different aspects of innate immunity, we provide a comprehensive overview of the host's first line of defence against rotavirus infection. Understanding these processes is expected to be of benefit in improving strategies to combat rotavirus disease.


Asunto(s)
Inmunidad Innata , Infecciones por Rotavirus/inmunología , Rotavirus/inmunología , Animales , Humanos , Ratones , Rotavirus/fisiología , Infecciones por Rotavirus/genética , Infecciones por Rotavirus/virología
11.
J Virol ; 86(24): 13456-66, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23035213

RESUMEN

The rotavirus spike protein domain VP8* is essential for recognition of cell surface carbohydrate receptors, notably those incorporating N-acylneuraminic acids (members of the sialic acid family). N-Acetylneuraminic acids occur naturally in both animals and humans, whereas N-glycolylneuraminic acids are acquired only through dietary uptake in normal human tissues. The preference of animal rotaviruses for these natural N-acylneuraminic acids has not been comprehensively established, and detailed structural information regarding the interactions of different rotaviruses with N-glycolylneuraminic acids is lacking. In this study, distinct specificities of VP8* for N-acetyl- and N-glycolylneuraminic acids were revealed using biophysical techniques. VP8* protein from the porcine rotavirus CRW-8 and the bovine rotavirus Nebraska calf diarrhea virus (NCDV) showed a preference for N-glycolyl- over N-acetylneuraminic acids, in contrast to results obtained with rhesus rotavirus (RRV). Crystallographic structures of VP8* from CRW-8 and RRV with bound methyl-N-glycolylneuraminide revealed the atomic details of their interactions. We examined the influence of amino acid type at position 157, which is proximal to the ligand's N-acetyl or N-glycolyl moiety and can mutate upon cell culture adaptation. A structure-based hypothesis derived from these results could account for rotavirus discrimination between the N-acylneuraminic acid forms. Infectivity blockade experiments demonstrated that the determined carbohydrate specificities of these VP8* domains directly correlate with those of the corresponding infectious virus. This includes an association between CRW-8 adaption to cell culture, decreased competition by N-glycolylneuraminic acid for CRW-8 infectivity, and a Pro157-to-Ser157 mutation in VP8* that reduces binding affinity for N-glycolylneuraminic acid.


Asunto(s)
Ácidos Neuramínicos/metabolismo , Receptores Virales/metabolismo , Rotavirus/fisiología , Ácidos Siálicos/metabolismo , Secuencia de Bases , Calorimetría , Cristalografía por Rayos X , Cartilla de ADN , Citometría de Flujo , Modelos Moleculares , Ácidos Neuramínicos/química , Resonancia Magnética Nuclear Biomolecular , Ácidos Siálicos/química
12.
J Biol Chem ; 286(8): 6165-74, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21138834

RESUMEN

The human α2ß1 integrin binds collagen and acts as a cellular receptor for rotaviruses and human echovirus 1. These ligands require the inserted (I) domain within the α2 subunit of α2ß1 for binding. Previous studies have identified the binding sites for collagen and echovirus 1 in the α2 I domain. We used CHO cells expressing mutated α2ß1 to identify amino acids involved in binding to human and animal rotaviruses. Residues where mutation affected rotavirus binding were located in several exposed loops and adjacent regions of the α2 I domain. Binding by all rotaviruses was eliminated by mutations in the activation-responsive αC-α6 and αF helices. This is a novel feature that distinguishes rotavirus from other α2ß1 ligands. Mutation of residues that co-ordinate the metal ion (Ser-153, Thr-221, and Glu-256 in α2 and Asp-130 in ß1) and nearby amino acids (Ser-154, Gln-215, and Asp-219) also inhibited rotavirus binding. The importance of most of these residues was greatest for binding by human rotaviruses. These mutations inhibit collagen binding to α2ß1 (apart from Glu-256) but do not affect echovirus binding. Overall, residues where mutation affected both rotavirus and collagen recognition are located at one side of the metal ion-dependent adhesion site, whereas those important for collagen alone cluster nearby. Mutations eliminating rotavirus and echovirus binding are distinct, consistent with the respective preference of these viruses for activated or inactive α2ß1. In contrast, rotavirus and collagen utilize activated α2ß1 and show an overlap in α2ß1 residues important for binding.


Asunto(s)
Integrina alfa2beta1/metabolismo , Infecciones por Rotavirus/metabolismo , Rotavirus/metabolismo , Animales , Sitios de Unión , Células CHO , Cricetinae , Cricetulus , Enterovirus Humano B/genética , Enterovirus Humano B/metabolismo , Infecciones por Enterovirus/genética , Infecciones por Enterovirus/metabolismo , Humanos , Integrina alfa2beta1/genética , Mutación , Mapeo Peptídico/métodos , Rotavirus/genética , Infecciones por Rotavirus/genética
13.
J Virol ; 85(20): 10509-17, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21849465

RESUMEN

Antibodies that neutralize rotavirus infection target outer coat proteins VP4 and VP7 and inhibit viral entry. The structure of a VP7-Fab complex (S. T. Aoki, et al., Science 324:1444-1447, 2009) led us to reclassify epitopes into two binding regions at inter- and intrasubunit boundaries of the calcium-dependent trimer. It further led us to show that antibodies binding at the intersubunit boundary inhibit uncoating of the virion outer layer. We have now tested representative antibodies for each of the defined structural epitope regions and find that antibodies recognizing epitopes in either binding region neutralize by cross-linking VP7 trimers. Antibodies that bind at the intersubunit junction neutralize as monovalent Fabs, while those that bind at the intrasubunit region require divalency. The VP7 structure has also allowed us to design a disulfide cross-linked VP7 mutant which recoats double-layered particles (DLPs) as efficiently as does wild-type VP7 but which yields particles defective in cell entry as determined both by lack of infectivity and by loss of α-sarcin toxicity in the presence of recoated particles. We conclude that dissociation of the VP7 trimer is an essential step in viral penetration into cells.


Asunto(s)
Antígenos Virales/metabolismo , Proteínas de la Cápside/metabolismo , Rotavirus/fisiología , Internalización del Virus , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Antígenos Virales/química , Antígenos Virales/inmunología , Proteínas de la Cápside/química , Proteínas de la Cápside/inmunología , Línea Celular , Disulfuros/metabolismo , Macaca mulatta
14.
PLoS Pathog ; 6(5): e1000898, 2010 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-20485572

RESUMEN

Many bacterial pathogens utilize a type III secretion system to deliver multiple effector proteins into host cells. Here we found that the type III effectors, NleE from enteropathogenic E. coli (EPEC) and OspZ from Shigella, blocked translocation of the p65 subunit of the transcription factor, NF-kappaB, to the host cell nucleus. NF-kappaB inhibition by NleE was associated with decreased IL-8 expression in EPEC-infected intestinal epithelial cells. Ectopically expressed NleE also blocked nuclear translocation of p65 and c-Rel, but not p50 or STAT1/2. NleE homologues from other attaching and effacing pathogens as well OspZ from Shigella flexneri 6 and Shigella boydii, also inhibited NF-kappaB activation and p65 nuclear import; however, a truncated form of OspZ from S. flexneri 2a that carries a 36 amino acid deletion at the C-terminus had no inhibitory activity. We determined that the C-termini of NleE and full length OspZ were functionally interchangeable and identified a six amino acid motif, IDSY(M/I)K, that was important for both NleE- and OspZ-mediated inhibition of NF-kappaB activity. We also established that NleB, encoded directly upstream from NleE, suppressed NF-kappaB activation. Whereas NleE inhibited both TNFalpha and IL-1beta stimulated p65 nuclear translocation and IkappaB degradation, NleB inhibited the TNFalpha pathway only. Neither NleE nor NleB inhibited AP-1 activation, suggesting that the modulatory activity of the effectors was specific for NF-kappaB signaling. Overall our data show that EPEC and Shigella have evolved similar T3SS-dependent means to manipulate host inflammatory pathways by interfering with the activation of selected host transcriptional regulators.


Asunto(s)
Escherichia coli O157/metabolismo , Proteínas de Escherichia coli/metabolismo , Shigella boydii/metabolismo , Shigella flexneri/metabolismo , Factor de Transcripción ReIA/metabolismo , Factores de Virulencia/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Células CACO-2 , Disentería Bacilar/inmunología , Disentería Bacilar/metabolismo , Disentería Bacilar/microbiología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Escherichia coli O157/patogenicidad , Células HeLa , Humanos , Proteínas I-kappa B/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Subunidad p50 de NF-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-rel/metabolismo , ARN Mensajero/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Shigella boydii/patogenicidad , Shigella flexneri/patogenicidad , Activación Transcripcional/fisiología , Virulencia
15.
Nat Chem Biol ; 5(2): 91-3, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19109595

RESUMEN

We used NMR spectroscopy, molecular modeling and infectivity competition assays to investigate the key interactions between the spike protein (VP8(*)) from 'sialidase-insensitive' human Wa and 'sialidase-sensitive' porcine CRW-8 rotaviruses and the glycans of gangliosides G(M1) and G(D1a). Our data provide strong evidence that N-acetylneuraminic acid is a key determinant for binding of these rotaviruses. This is in contrast to the widely accepted paradigm that sialic acids are irrelevant in host cell recognition by sialidase-insensitive rotaviruses.


Asunto(s)
Ácido N-Acetilneuramínico/metabolismo , Rotavirus/patogenicidad , Espectroscopía de Resonancia Magnética , Rotavirus/metabolismo
16.
J Gen Virol ; 91(Pt 8): 2007-2018, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20392902

RESUMEN

Intestinal epithelial cell death following rotavirus infection is associated with villus atrophy and gastroenteritis. Roles for both apoptosis and necrosis in cytocidal activity within rotavirus-infected epithelial cells have been proposed. Additionally, inactivated rotavirus has been reported to induce diarrhoea in infant mice. We further examined the death mechanisms induced in epithelial cell lines following rotavirus infection or inactivated rotavirus exposure. Monolayer integrity changes in MA104, HT-29 and partially differentiated Caco-2 cells following inactivated rotavirus exposure or RRV or CRW-8 rotavirus infection paralleled cell metabolic activity and viability reductions. MA104 cell exposure to rotavirus dsRNA also altered monolayer integrity. Inactivated rotaviruses induced delayed cell function losses that were unrelated to apoptosis. Phosphatidylserine externalization, indicating early apoptosis, occurred in MA104 and HT-29 but not in partially differentiated Caco-2 cells by 11 h after infection. Rotavirus activation of phosphatidylinositol 3-kinase partially protected MA104 and HT-29 cells from early apoptosis. In contrast, activation of the stress-activated protein kinase JNK by rotavirus did not influence apoptosis induction in these cells. RRV infection produced DNA fragmentation, indicating late-stage apoptosis, in fully differentiated Caco-2 cells only. These studies show that the apoptosis initiation and cell death mechanism induced by rotavirus infection depend on cell type and degree of differentiation. Early stage apoptosis resulting from rotavirus infection is probably counter-balanced by virus-induced phosphatidylinositol 3-kinase activation. The ability of inactivated rotaviruses and rotavirus dsRNA to perturb monolayer integrity supports a potential role for these rotavirus components in disease pathogenesis.


Asunto(s)
Muerte Celular , Células Epiteliales/virología , ARN Viral/genética , Rotavirus/patogenicidad , Proteínas Virales/toxicidad , Animales , Línea Celular , Supervivencia Celular , Humanos , Macaca mulatta , Transfección
17.
J Virol ; 83(10): 4942-51, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19244315

RESUMEN

A vital arm of the innate immune response to viral infection is the induction and subsequent antiviral effects of interferon (IFN). Rotavirus reduces type I IFN induction in infected cells by the degradation of IFN regulatory factors. Here, we show that the monkey rotavirus RRV and human rotavirus Wa also block gene expression induced by type I and II IFNs through a mechanism allowing signal transducer and activator of transcription 1 (STAT1) and STAT2 activation but preventing their nuclear accumulation. In infected cells, this may allow rotavirus to block the antiviral actions of IFN produced early in infection or by activated immune cells. As the intracellular expression of rotavirus nonstructural proteins NSP1, NSP3, and NSP4 individually did not inhibit IFN-stimulated gene expression, their involvement in this process is unlikely. RRV and Wa rotaviruses also prevented the tumor necrosis factor alpha-stimulated nuclear accumulation of NF-kappaB and NF-kappaB-driven gene expression. In addition, NF-kappaB was activated by rotavirus infection, confirming earlier findings by others. As NF-kappaB is important for the induction of IFN and other cytokines during viral infection, this suggests that rotavirus prevents cellular transcription as a means to evade host responses. To our knowledge, this is the first report of the use of this strategy by a double-stranded RNA virus.


Asunto(s)
FN-kappa B/inmunología , Infecciones por Rotavirus/inmunología , Rotavirus/inmunología , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT2/inmunología , Animales , Células CACO-2 , Expresión Génica , Genes Reporteros , Humanos , Inmunidad Innata , Interferones/inmunología , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Mensajero/metabolismo , Rotavirus/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas no Estructurales Virales/metabolismo
18.
Glycobiology ; 19(3): 194-200, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18974199

RESUMEN

The rotavirus spike protein VP4 mediates attachment to host cells and subsequent membrane penetration. The VP8(*) domain of VP4 forms the spike tips and is proposed to recognize host-cell surface glycans. For sialidase-sensitive rotaviruses such as rhesus (RRV), this recognition involves terminal sialic acids. We show here that the RRV VP8(*)(64-224) protein competes with RRV infection of host cells, demonstrating its relevance to infection. In addition, we observe that the amino acids revealed by X-ray crystallography to be in direct contact with the bound sialic acid derivative methyl alpha-D-N-acetylneuraminide, and that are highly conserved amongst sialidase-sensitive rotaviruses, are residues that are also important in interactions with host-cell carbohydrates. Residues Arg101 and Ser190 of the RRV VP8(*) carbohydrate-binding site were mutated to assess their importance for binding to the sialic acid derivative and their competition with RRV infection of host cells. The crystallographic structure of the Arg(101)Ala mutant crystallized in the presence of the sialic acid derivative was determined at 295 K to a resolution of 1.9 A. Our multidisciplinary study using X-ray crystallography, saturation transfer difference nuclear magnetic resonance spectroscopy, isothermal titration calorimetry, and competitive virus infectivity assays to investigate RRV wild-type and mutant VP8(*) proteins has provided the first evidence that the carbohydrate-binding cavity in RRV VP8(*) is used for host-cell recognition, and this interaction is not only with the sialic acid portion but also with other parts of the glycan structure.


Asunto(s)
Aminoácidos/genética , Mutación , Ácido N-Acetilneuramínico/química , Rotavirus/química , Proteínas no Estructurales Virales/química , Secuencia de Aminoácidos , Sitios de Unión , Calorimetría , Carbohidratos/química , Cristalografía por Rayos X , Enlace de Hidrógeno , Ligandos , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Agua/química , Difracción de Rayos X
19.
J Virol ; 82(1): 148-60, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17942548

RESUMEN

Changes in the interactions between intestinal cells and their surrounding environment during virus infection have not been well documented. The growth and survival of intestinal epithelial cells, the main targets of rotavirus infection, are largely dependent on the interaction of cell surface integrins with the extracellular matrix. In this study, we detected alterations in cellular integrin expression following rotavirus infection, identified the signaling components required, and analyzed the subsequent effects on cell binding to the matrix component collagen. After rotavirus infection of intestinal cells, expression of alpha2beta1 and beta2 integrins was up-regulated, whereas that of alphaVbeta3, alphaVbeta5, and alpha5beta1 integrins, if present, was down-regulated. This differential regulation of integrins was reflected at the transcriptional level. It was unrelated to the use of integrins as rotavirus receptors, as both integrin-using and integrin-independent viruses induced integrin regulation. Using pharmacological agents that inhibit kinase activity, integrin regulation was shown to be dependent on phosphatidylinositol 3-kinase (PI3K) but independent of the activities of the mitogen-activated protein kinases p38 and ERK1/2, and cyclooxygenase-2. Replication-dependent activation of the PI3K/Akt pathway was observed following infection of intestinal and nonintestinal cell lines. Rotavirus activation of PI3K was important for regulation of alpha2beta1 expression. Blockade of integrin regulation by PI3K inhibition led to decreased adherence of infected intestinal cells to collagen and a concomitant decrease in virus titer. These findings indicate that rotavirus-induced PI3K activation causes regulation of integrin expression in intestinal cells, leading to prolonged adherence of infected cells to collagen and increased virus production.


Asunto(s)
Adhesión Celular , Células Epiteliales/virología , Integrinas/biosíntesis , Intestinos/virología , Fosfatidilinositol 3-Quinasas/metabolismo , Rotavirus/fisiología , Replicación Viral , Línea Celular , Colágeno/metabolismo , Ciclooxigenasa 2/metabolismo , Regulación de la Expresión Génica , Células HT29 , Humanos , Intestinos/citología , Proteínas Quinasas Activadas por Mitógenos/metabolismo
20.
J Virol ; 82(13): 6139-49, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18417562

RESUMEN

Infection modulates type 1 diabetes, a common autoimmune disease characterized by the destruction of insulin-producing islet beta cells in the pancreas. Childhood rotavirus infections have been associated with exacerbations in islet autoimmunity. Nonobese diabetic (NOD) mice develop lymphocytic islet infiltration (insulitis) and then clinical diabetes, whereas NOD8.3 TCR mice, transgenic for a T-cell receptor (TCR) specific for an important islet autoantigen, show more rapid diabetes onset. Oral infection of infant NOD mice with the monkey rotavirus strain RRV delays diabetes development. Here, the effect of RRV infection on diabetes development once insulitis is established was determined. NOD and NOD8.3 TCR mice were inoculated with RRV aged > or = 12 and 5 weeks, respectively. Diabetes onset was significantly accelerated in both models (P < 0.024), although RRV infection was asymptomatic and confined to the intestine. The degree of diabetes acceleration was related to the serum antibody titer to RRV. RRV-infected NOD mice showed a possible trend toward increased insulitis development. Infected males showed increased CD8(+) T-cell proportions in islets. Levels of beta-cell major histocompatibility complex class I expression and islet tumor necrosis factor alpha mRNA were elevated in at least one model. NOD mouse exposure to mouse rotavirus in a natural experiment also accelerated diabetes. Thus, rotavirus infection after beta-cell autoimmunity is established affects insulitis and exacerbates diabetes. A possible mechanism involves increased exposure of beta cells to immune recognition and activation of autoreactive T cells by proinflammatory cytokines. The timing of infection relative to mouse age and degree of insulitis determines whether diabetes onset is delayed, unaltered, or accelerated.


Asunto(s)
Diabetes Mellitus Tipo 1/etiología , Islotes Pancreáticos/inmunología , Infecciones por Rotavirus/complicaciones , Factores de Edad , Análisis de Varianza , Animales , Anticuerpos Antivirales/sangre , Linfocitos T CD8-positivos/inmunología , Citometría de Flujo , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA