Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Recognit ; 31(9): e2718, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29687510

RESUMO

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.


Assuntos
Proteínas do Capsídeo/genética , Ácidos Neuramínicos/química , Rotavirus/genética , Proteínas não Estruturais Virais/genética , Animais , Proteínas do Capsídeo/química , Carboidratos/química , Carboidratos/genética , Bovinos , Cães , Epitopos/genética , Epitopos/imunologia , Ligação Proteica/genética , Domínios Proteicos/genética , Rotavirus/química , Suínos , Proteínas não Estruturais Virais/química
2.
Sci Rep ; 8(1): 67, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29311575

RESUMO

Detection of viral infection by host cells leads to secretion of type I interferon, which induces antiviral gene expression. The class I major histocompatibility complex (MHCI) is required for viral antigen presentation and subsequent infected cell killing by cytotoxic T lymphocytes. STAT1 activation by interferon can induce NLRC5 expression, promoting MHCI expression. Rotavirus, an important pathogen, blocks interferon signalling through inhibition of STAT1 nuclear translocation. We assessed MHCI expression in HT-29 intestinal epithelial cells following rotavirus infection. MHCI levels were upregulated in a partially type I interferon-dependent manner in bystander cells lacking rotavirus antigen, but not in infected cells. MHCI and NLRC5 mRNA expression also was elevated in bystander, but not infected, cells, suggesting a transcriptional block in infected cells. STAT1 was activated in bystander and infected cells, but showed nuclear localisation in bystander cells only. Overall, the lack of MHCI upregulation in rotavirus-infected cells may be at least partially due to rotavirus blockade of interferon-induced STAT1 nuclear translocation. The reduced MHCI protein levels in infected cells support the existence of an additional, non-transcriptional mechanism that reduces MHCI expression. It is possible that rotavirus also may suppress MHCI expression in vivo, which might limit T cell-mediated killing of rotavirus-infected enterocytes.


Assuntos
Antígenos Virais/imunologia , Expressão Gênica , Genes MHC Classe I , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Infecções por Rotavirus/genética , Infecções por Rotavirus/imunologia , Rotavirus/imunologia , Animais , Antígenos Virais/genética , Biomarcadores , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Humanos , Interferon Tipo I/metabolismo , Mucosa Intestinal/virologia , Camundongos , Infecções por Rotavirus/virologia , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Replicação Viral
3.
Sci Rep ; 6: 29697, 2016 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-27405244

RESUMO

Rotavirus infection is associated with childhood progression to type 1 diabetes. Infection by monkey rotavirus RRV accelerates diabetes onset in non-obese diabetic (NOD) mice, which relates to regional lymph node infection and a T helper 1-specific immune response. When stimulated ex vivo with RRV, plasmacytoid dendritic cells (pDCs) from naïve NOD mice secrete type I interferon, which induces the activation of bystander lymphocytes, including islet-autoreactive T cells. This is our proposed mechanism for diabetes acceleration by rotaviruses. Here we demonstrate bystander lymphocyte activation in RRV-infected NOD mice, which showed pDC activation and strong upregulation of interferon-dependent gene expression, particularly within lymph nodes. The requirement for type I interferon signalling was analysed using NOD mice lacking a functional type I interferon receptor (NOD.IFNAR1(-/-) mice). Compared with NOD mice, NOD.IFNAR1(-/-) mice showed 8-fold higher RRV titers in lymph nodes and 3-fold higher titers of total RRV antibody in serum. However, RRV-infected NOD.IFNAR1(-/-) mice exhibited delayed pDC and lymphocyte activation, no T helper 1 bias in RRV-specific antibodies and unaltered diabetes onset when compared with uninfected controls. Thus, the type I interferon signalling induced by RRV infection is required for bystander lymphocyte activation and accelerated type 1 diabetes onset in genetically susceptible mice.


Assuntos
Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/imunologia , Interferon Tipo I/imunologia , Infecções por Rotavirus/imunologia , Rotavirus/imunologia , Transdução de Sinais/imunologia , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/virologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/virologia , Interferon Tipo I/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Infecções por Rotavirus/genética , Infecções por Rotavirus/patologia , Transdução de Sinais/genética , Células Th1/imunologia , Células Th1/patologia
4.
Curr Opin Virol ; 15: 90-6, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26363995

RESUMO

Rotaviruses are major etiologic agents of severe gastroenteritis in human and animals, infecting the mature intestinal epithelium. Their attachment to host cell glycans is mediated through the virion spike protein. This is considered to be crucial for successful host cell invasion by rotaviruses. Recent studies have greatly expanded our understanding of the diversity of glycans commonly recognized by rotaviruses, to include the ganglioside GM1a and histo-blood group antigens. Here, these new findings are integrated with advances in knowledge of spike protein structure, rotavirus entry mechanisms and innate intestinal immunity to provide an overview of the variety of rotavirus glycan receptors and their roles in cell penetration, host tropism and pathogenesis.


Assuntos
Polissacarídeos/fisiologia , Receptores Virais/fisiologia , Infecções por Rotavirus/virologia , Rotavirus/fisiologia , Animais , Especificidade de Hospedeiro , Humanos , Imunidade Inata , Polissacarídeos/imunologia , Polissacarídeos/metabolismo , Receptores Virais/imunologia , Receptores Virais/metabolismo , Rotavirus/imunologia , Rotavirus/metabolismo , Internalização do Vírus
5.
J Gen Virol ; 96(12): 3519-3524, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26404393

RESUMO

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.


Assuntos
Mitocôndrias/fisiologia , Rotavirus/metabolismo , Proteínas não Estruturais Virais/fisiologia , Animais , Linhagem Celular , Regulação Viral da Expressão Gênica/fisiologia , Células HEK293 , Humanos , Transporte Proteico , Rotavirus/genética , Replicação Viral
6.
Chembiochem ; 16(15): 2176-81, 2015 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-26250751

RESUMO

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.


Assuntos
Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Especificidade de Hospedeiro , Receptores Virais/metabolismo , Infecções por Rotavirus/virologia , Rotavirus/química , Sequência de Aminoácidos , Animais , Carboidratos/farmacologia , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína/efeitos dos fármacos , Receptores Virais/química , Rotavirus/genética , Rotavirus/metabolismo , Alinhamento de Sequência
7.
Virus Res ; 208: 89-97, 2015 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-26079065

RESUMO

Rotavirus infection is a major cause of life-threatening infantile gastroenteritis. The innate immune system provides an immediate mechanism of suppressing viral replication and is necessary for an effective adaptive immune response. Innate immunity involves host recognition of viral infection and establishment of a powerful antiviral state through the expression of pro-inflammatory cytokines such as type-1 interferon (IFN). Macrophages, the front-line cells of innate immunity, produce IFN and other cytokines in response to viral infection. However, the role of macrophages during rotavirus infection is not well defined. We demonstrate here that RRV rotavirus triggers the production of proinflammatory cytokines from mouse bone marrow-derived macrophages. IFN and antiviral cytokine production was abolished in rotavirus-infected MAVS (-/-) macrophages. This indicates that rotavirus triggers innate immunity in macrophages through RIG-I and/or MDA5 viral recognition, and MAVS signaling is essential for cytokine responses in macrophages. Rotavirus induced IFN expression in both wild type and MDA5 (-/-) macrophages, showing that MDA5 is not essential for IFN secretion following infection, and RIG-I and MDA5 may act redundantly in promoting rotavirus recognition. Interestingly, rotavirus neither stimulated mitogen-activated protein kinases p38 and JNK nor activated the NLRP3 inflammasome, demonstrating that these components might not be involved in innate responses to rotavirus infection in macrophages. Our results indicate that rotavirus elicits intracellular signaling in macrophages, resulting in the induction of IFN and antiviral cytokines, and advance our understanding of the involvement of these cells in innate responses against rotavirus.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas de Transporte/imunologia , Inflamassomos/imunologia , MAP Quinase Quinase 4/imunologia , Macrófagos/imunologia , Infecções por Rotavirus/imunologia , Rotavirus/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Transporte/genética , Humanos , Imunidade Inata , Inflamassomos/genética , MAP Quinase Quinase 4/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Rotavirus/imunologia , Infecções por Rotavirus/genética , Infecções por Rotavirus/virologia , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/genética
8.
J Biol Chem ; 290(19): 12403-14, 2015 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-25814665

RESUMO

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.


Assuntos
Antígenos de Superfície/química , Glicolipídeos/química , Glicoproteínas/química , Glicoproteínas de Membrana/química , Proteínas do Leite/química , Peptídeos/química , Infecções por Rotavirus/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Bovinos , Membrana Celular/metabolismo , Sobrevivência Celular , Equidae , Cavalos , Humanos , Concentração Inibidora 50 , Integrinas/química , Gotículas Lipídicas , Leite , Dados de Sequência Molecular , Proteômica , Rotavirus/metabolismo , Infecções por Rotavirus/tratamento farmacológico , Homologia de Sequência de Aminoácidos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
9.
Diabetologia ; 58(6): 1149-59, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25794781

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Ativação Linfocitária/imunologia , Animais , Autoantígenos/imunologia , Efeito Espectador , Citocinas/metabolismo , Células Dendríticas/citologia , Diabetes Mellitus Tipo 1/virologia , Enterovirus/imunologia , Humanos , Inflamação , Vírus da Influenza A/imunologia , Camundongos , Mimetismo Molecular , Pâncreas/virologia , Peptídeos/química , Polimorfismo Genético , Receptores de Reconhecimento de Padrão/metabolismo , Rotavirus/imunologia , Linfócitos T/citologia , Viroses/complicações , Viroses/imunologia
10.
J Gen Virol ; 96(Pt 7): 1768-76, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25701827

RESUMO

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.


Assuntos
NF-kappa B/antagonistas & inibidores , Rotavirus/imunologia , Proteínas não Estruturais Virais/metabolismo , Proteínas Contendo Repetições de beta-Transducina/antagonistas & inibidores , Animais , Linhagem Celular , Células Epiteliais/imunologia , Células Epiteliais/virologia , Humanos , Evasão da Resposta Imune , Proteólise , Suínos
11.
Nat Commun ; 6: 5907, 2015 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-25556995

RESUMO

Histo-blood group antigens (HBGAs) have been proposed as rotavirus receptors. H type-1 and Lewis(b) antigens have been reported to bind VP8* from major human rotavirus genotypes P[4], P[6] and P[8], while VP8* from a rarer P[14] rotavirus recognizes A-type HBGAs. However, the role and significance of HBGA receptors in rotavirus pathogenesis remains uncertain. Here we report that P[14] rotavirus HAL1166 and the related P[9] human rotavirus K8 bind to A-type HBGAs, although neither virus engages the HBGA-specific α1,2-linked fucose moiety. Notably, human rotaviruses DS-1 (P[4]) and RV-3 (P[6]) also use A-type HBGAs for infection, with fucose involvement. However, human P[8] rotavirus Wa does not recognize A-type HBGAs. Furthermore, the common human rotaviruses that we have investigated do not use Lewis(b) and H type-1 antigens. Our results indicate that A-type HBGAs are receptors for human rotaviruses, although rotavirus strains vary in their ability to recognize these antigens.


Assuntos
Sistema ABO de Grupos Sanguíneos/metabolismo , Infecções por Rotavirus/fisiopatologia , Rotavirus/metabolismo , Internalização do Vírus , Humanos , Espectroscopia de Ressonância Magnética , Simulação de Dinâmica Molecular , Estrutura Molecular , Oligossacarídeos/química , Oligossacarídeos/metabolismo , Infecções por Rotavirus/metabolismo
12.
Virology ; 468-470: 504-509, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25262468

RESUMO

T cell-receptor transgenic NOD8.3 mice provide a model for spontaneous type 1 diabetes development. Infection of 5 week-old NOD8.3 mice with Rhesus monkey rotavirus (RRV) accelerates the onset of their diabetes. This acceleration requires virus replication and relates to the presence and level of serum anti-rotavirus antibodies, but the role of individual RRV genes is unknown. Here we assessed the importance for diabetes acceleration of the RRV genes encoding VP4 and VP7, by infecting NOD8.3 mice with parental and reassortant rotaviruses. Diabetes was accelerated by reassortant rotaviruses containing RRV VP7 on a UK rotavirus genetic background, but not by parental UK or a UK reassortant containing RRV VP4 without VP7. Diabetes acceleration by reassortant rotaviruses containing RRV VP7 depended on the development of a high serum anti-rotavirus antibody titer. This study shows that VP7, together with an elevated anti-rotavirus antibody response, contributes to the acceleration of diabetes onset by RRV.


Assuntos
Anticorpos Antivirais/imunologia , Antígenos Virais/metabolismo , Proteínas do Capsídeo/metabolismo , Diabetes Mellitus , Rotavirus/fisiologia , Animais , Antígenos Virais/genética , Glicemia , Proteínas do Capsídeo/genética , Linhagem Celular , Feminino , Regulação Viral da Expressão Gênica , Masculino , Camundongos , Camundongos Transgênicos , Vírus Reordenados , Organismos Livres de Patógenos Específicos
13.
J Gen Virol ; 95(Pt 8): 1723-1733, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24814927

RESUMO

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.


Assuntos
Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Interferons/antagonistas & inibidores , Rotavirus/fisiologia , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT2/antagonistas & inibidores , Linhagem Celular , Humanos , Interferons/imunologia , Carioferinas/metabolismo , Rotavirus/imunologia
14.
PLoS Pathog ; 10(3): e1003998, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24676425

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/virologia , Ativação Linfocitária/imunologia , Glicoproteínas de Membrana/imunologia , Infecções por Rotavirus/imunologia , Transdução de Sinais , Receptor 7 Toll-Like/imunologia , Animais , Diabetes Mellitus Tipo 1/imunologia , Citometria de Fluxo , Interferons/biossíntese , Interferons/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Rotavirus/imunologia , Transdução de Sinais/imunologia
15.
J Virol ; 88(8): 4558-71, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24501414

RESUMO

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.


Assuntos
Gangliosídeos/metabolismo , Integrina alfa2beta1/metabolismo , Ácidos Neuramínicos/metabolismo , Receptores Virais/metabolismo , Infecções por Rotavirus/metabolismo , Rotavirus/fisiologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Humanos , Integrina alfa2beta1/genética , Ácido N-Acetilneuramínico/metabolismo , Ligação Proteica , Receptores Virais/genética , Rotavirus/genética , Infecções por Rotavirus/genética , Infecções por Rotavirus/virologia
16.
Virus Res ; 179: 73-84, 2014 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-24246305

RESUMO

Rotavirus infection has been proposed to enhance progression towards type 1 diabetes in at-risk children. Rhesus monkey rotavirus (RRV) accelerates diabetes onset in non-obese diabetic (NOD) and T cell receptor transgenic NOD8.3 mice. Infected NOD mice show virus spread to pancreatic lymph nodes (PLN) and mesenteric lymph nodes (MLN), induction of a serum T helper 1-biased specific antibody response and proinflammatory cytokine mRNA expression in PLN and islets. Here, we analysed the effects of RRV infection on intestinal responses and the activation of antigen presenting cells (APC), T cells and B cells in PLN, MLN, spleen and islets. Diabetes acceleration by RRV was associated with minimal immune activation in Peyer's patches. Increased proinflammatory cytokine expression by APC, including dendritic cells, was observed exclusively in the PLN, while cytokine expression by T cells was detected in islets, PLN, MLN and spleen. RRV infection of NOD8.3 mice increased IFNγ expression by CD8(+) T cells, which primarily recognise an islet autoantigen. A peptide corresponding to RRV VP7 amino acids 5-13, with sequence similarity to this islet autoantigen, did not induce activation or proliferation of NOD8.3 mouse T cells. RRV infection of NOD mice elevated B cell MHC I expression in PLN and MLN, and increased the B cell-mediated proliferation of islet antigen-specific CD8(+) T cells. These studies demonstrate that RRV infection of NOD mice activates APC, T cells and B cells at sites where autoreactive lymphocytes accumulate, in association with proinflammatory cytokine expression and an increased capacity to present antigen. Taken together with previous findings, these data support a possible role for bystander activation in type 1 diabetes acceleration by RRV.


Assuntos
Apresentação de Antígeno , Linfócitos B/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/virologia , Genes MHC Classe I , Infecções por Rotavirus/imunologia , Rotavirus/fisiologia , Linfócitos T/imunologia , Animais , Proliferação de Células , Citocinas/genética , Citocinas/imunologia , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Progressão da Doença , Feminino , Humanos , Ilhotas Pancreáticas/imunologia , Linfonodos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Rotavirus/imunologia , Infecções por Rotavirus/genética , Infecções por Rotavirus/patologia , Infecções por Rotavirus/virologia , Baço/imunologia , Linfócitos T/citologia , Regulação para Cima
17.
PLoS One ; 8(3): e59182, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23554993

RESUMO

Rotaviruses are implicated as a viral trigger for the acceleration of type 1 diabetes in children. Infection of adult non-obese diabetic (NOD) mice with rotavirus strain RRV accelerates diabetes development, whereas RRV infection in infant NOD mice delays diabetes onset. In this study of infant mice, RRV titers and lymphocyte populations in the intestine, mesenteric lymph nodes (MLN) and thymus of NOD mice were compared with those in diabetes-resistant BALB/c and C57BL/6 mice. Enhanced intestinal RRV infection occurred in NOD mice compared with the other mouse strains. This was associated with increases in the frequency of CD8αß TCRαß intraepithelial lymphocytes, and their PD-L1 expression. Virus spread to the MLN and T cell numbers there also were greatest in NOD mice. Thymic RRV infection is shown here in all mouse strains, often in combination with alterations in T cell ontogeny. Infection lowered thymocyte numbers in infant NOD and C57BL/6 mice, whereas thymocyte production was unaltered overall in infant BALB/c mice. In the NOD mouse thymus, effector CD4(+) T cell numbers were reduced by infection, whereas regulatory T cell numbers were maintained. It is proposed that maintenance of thymic regulatory T cell numbers may contribute to the increased suppression of inflammatory T cells in response to a strong stimulus observed in pancreatic lymph nodes of adult mice infected as infants. These findings show that rotavirus replication is enhanced in diabetes-prone mice, and provide evidence that thymic T cell alterations may contribute to the delayed diabetes onset following RRV infection.


Assuntos
Diabetes Mellitus Tipo 1/patologia , Infecções por Rotavirus/patologia , Subpopulações de Linfócitos T/patologia , Linfócitos T/patologia , Timo/patologia , Fatores Etários , Animais , Animais Recém-Nascidos , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/virologia , Feminino , Expressão Gênica , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno/imunologia , Intestinos/imunologia , Intestinos/patologia , Intestinos/virologia , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/virologia , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Rotavirus/imunologia , Infecções por Rotavirus/complicações , Infecções por Rotavirus/imunologia , Infecções por Rotavirus/virologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/virologia , Linfócitos T/imunologia , Linfócitos T/virologia , Timo/imunologia , Timo/virologia
18.
J Gen Virol ; 94(Pt 6): 1151-1160, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23486667

RESUMO

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.


Assuntos
Imunidade Inata , Infecções por Rotavirus/imunologia , Rotavirus/imunologia , Animais , Humanos , Camundongos , Rotavirus/fisiologia , Infecções por Rotavirus/genética , Infecções por Rotavirus/virologia
19.
J Virol ; 86(24): 13456-66, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23035213

RESUMO

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.


Assuntos
Ácidos Neuramínicos/metabolismo , Receptores Virais/metabolismo , Rotavirus/fisiologia , Ácidos Siálicos/metabolismo , Sequência de Bases , Calorimetria , Cristalografia por Raios X , Primers do DNA , Citometria de Fluxo , Modelos Moleculares , Ácidos Neuramínicos/química , Ressonância Magnética Nuclear Biomolecular , Ácidos Siálicos/química
20.
J Mol Biol ; 413(5): 929-39, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21945555

RESUMO

Rotaviruses ubiquitously infect children under the age of 5, being responsible for more than half a million diarrhoeal deaths each year worldwide. Host cell oligosaccharides containing sialic acid(s) are critical for attachment by rotaviruses. However, to date, no detailed three-dimensional atomic model showing the exact rotavirus interactions with these glycoconjugate receptors has been reported. Here, we present the first crystallographic structures of the rotavirus carbohydrate-recognizing protein VP8* in complex with ganglioside G(M3) glycans. In combination with assessment of the inhibition of rotavirus infectivity by N-acetyl and N-glycolyl forms of this ganglioside, our results reveal key details of rotavirus-ganglioside G(M3) glycan recognition. In addition, they show a direct correlation between the carbohydrate specificities exhibited by VP8* from porcine and by monkey rotaviruses and the respective infectious virus particles. These novel results also indicate the potential binding interactions of rotavirus VP8* with other sialic acid-containing gangliosides.


Assuntos
Gangliosídeos/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Polissacarídeos/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Infecções por Rotavirus/metabolismo , Rotavirus/patogenicidade , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Animais , Cristalografia por Raios X , Haplorrinos , Espectroscopia de Ressonância Magnética , Modelos Químicos , Conformação Proteica , Infecções por Rotavirus/virologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...