Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Cell ; 184(13): 3438-3451.e10, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34139177

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been spreading worldwide, causing a global pandemic. Bat-origin RaTG13 is currently the most phylogenetically related virus. Here we obtained the complex structure of the RaTG13 receptor binding domain (RBD) with human ACE2 (hACE2) and evaluated binding of RaTG13 RBD to 24 additional ACE2 orthologs. By substituting residues in the RaTG13 RBD with their counterparts in the SARS-CoV-2 RBD, we found that residue 501, the major position found in variants of concern (VOCs) 501Y.V1/V2/V3, plays a key role in determining the potential host range of RaTG13. We also found that SARS-CoV-2 could induce strong cross-reactive antibodies to RaTG13 and identified a SARS-CoV-2 monoclonal antibody (mAb), CB6, that could cross-neutralize RaTG13 pseudovirus. These results elucidate the receptor binding and host adaption mechanisms of RaTG13 and emphasize the importance of continuous surveillance of coronaviruses (CoVs) carried by animal reservoirs to prevent another spillover of CoVs.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Sitios de Unión/fisiología , COVID-19/metabolismo , Quirópteros/virología , SARS-CoV-2/patogenicidad , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , COVID-19/inmunología , Quirópteros/inmunología , Quirópteros/metabolismo , Especificidad del Huésped/inmunología , Humanos , Filogenia , Unión Proteica/fisiología , Receptores Virales/metabolismo , SARS-CoV-2/inmunología , Alineación de Secuencia
2.
Nature ; 567(7746): 109-112, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30787439

RESUMEN

Zoonotic influenza A viruses of avian origin can cause severe disease in individuals, or even global pandemics, and thus pose a threat to human populations. Waterfowl and shorebirds are believed to be the reservoir for all influenza A viruses, but this has recently been challenged by the identification of novel influenza A viruses in bats1,2. The major bat influenza A virus envelope glycoprotein, haemagglutinin, does not bind the canonical influenza A virus receptor, sialic acid or any other glycan1,3,4, despite its high sequence and structural homology with conventional haemagglutinins. This functionally uncharacterized plasticity of the bat influenza A virus haemagglutinin means the tropism and zoonotic potential of these viruses has not been fully determined. Here we show, using transcriptomic profiling of susceptible versus non-susceptible cells in combination with genome-wide CRISPR-Cas9 screening, that the major histocompatibility complex class II (MHC-II) human leukocyte antigen DR isotype (HLA-DR) is an essential entry determinant for bat influenza A viruses. Genetic ablation of the HLA-DR α-chain rendered cells resistant to infection by bat influenza A virus, whereas ectopic expression of the HLA-DR complex in non-susceptible cells conferred susceptibility. Expression of MHC-II from different bat species, pigs, mice or chickens also conferred susceptibility to infection. Notably, the infection of mice with bat influenza A virus resulted in robust virus replication in the upper respiratory tract, whereas mice deficient for MHC-II were resistant. Collectively, our data identify MHC-II as a crucial entry mediator for bat influenza A viruses in multiple species, which permits a broad vertebrate tropism.


Asunto(s)
Quirópteros/virología , Antígenos de Histocompatibilidad Clase II/metabolismo , Especificidad del Huésped , Virus de la Influenza A/inmunología , Virus de la Influenza A/fisiología , Zoonosis/inmunología , Zoonosis/virología , Animales , Proteína 9 Asociada a CRISPR , Sistemas CRISPR-Cas , Pollos/genética , Pollos/inmunología , Quirópteros/genética , Quirópteros/inmunología , Quirópteros/metabolismo , Femenino , Perfilación de la Expresión Génica , Antígenos HLA-DR/genética , Antígenos HLA-DR/inmunología , Antígenos HLA-DR/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Especificidad del Huésped/genética , Especificidad del Huésped/inmunología , Humanos , Masculino , Ratones , Ratones Noqueados , Sistema Respiratorio/virología , Porcinos/genética , Porcinos/inmunología , Tropismo Viral/genética , Tropismo Viral/inmunología , Replicación Viral , Zoonosis/genética , Zoonosis/metabolismo
3.
PLoS Pathog ; 17(4): e1009501, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33836016

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein mediates infection of cells expressing angiotensin-converting enzyme 2 (ACE2). ACE2 is also the viral receptor of SARS-CoV (SARS-CoV-1), a related coronavirus that emerged in 2002-2003. Horseshoe bats (genus Rhinolophus) are presumed to be the original reservoir of both viruses, and a SARS-like coronavirus, RaTG13, closely related to SARS-CoV-2, has been identified in one horseshoe-bat species. Here we characterize the ability of the S-protein receptor-binding domains (RBDs) of SARS-CoV-1, SARS-CoV-2, pangolin coronavirus (PgCoV), RaTG13, and LyRa11, a bat virus similar to SARS-CoV-1, to bind a range of ACE2 orthologs. We observed that the PgCoV RBD bound human ACE2 at least as efficiently as the SARS-CoV-2 RBD, and that both RBDs bound pangolin ACE2 efficiently. We also observed a high level of variability in binding to closely related horseshoe-bat ACE2 orthologs consistent with the heterogeneity of their RBD-binding regions. However five consensus horseshoe-bat ACE2 residues enhanced ACE2 binding to the SARS-CoV-2 RBD and neutralization of SARS-CoV-2 pseudoviruses by an enzymatically inactive immunoadhesin form of human ACE2 (hACE2-NN-Fc). Two of these mutations impaired neutralization of SARS-CoV-1 pseudoviruses. An hACE2-NN-Fc variant bearing all five mutations neutralized both SARS-CoV-2 pseudovirus and infectious virus more efficiently than wild-type hACE2-NN-Fc. These data suggest that SARS-CoV-1 and -2 originate from distinct bat species, and identify a more potently neutralizing form of soluble ACE2.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , COVID-19/inmunología , COVID-19/virología , Quirópteros/metabolismo , SARS-CoV-2/genética , Animales , COVID-19/genética , Quirópteros/genética , Especificidad del Huésped/genética , Especificidad del Huésped/inmunología , Humanos , Modelos Moleculares , Mutación , Unión Proteica/genética , Unión Proteica/fisiología , Receptores Virales/metabolismo , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo
4.
PLoS Pathog ; 16(2): e1008330, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32084248

RESUMEN

Both the replication and transcription of the influenza virus are catalyzed by the viral polymerase complex. The polymerases of most avian influenza A viruses have poor performance in mammalian cells, which is considered to be one of the important species barriers. Pigs have been long considered as important intermediate hosts for interspecies transmission of the avian influenza virus, because of their susceptibility to infection with both avian and mammalian influenza viruses. However, the molecular basis of influenza polymerase adaptation in pigs remains largely unknown. ANP32A and ANP32B proteins have been identified as playing fundamental roles in influenza virus replication and host range determination. In this study, we found that swine ANP32A (swANP32A), unlike swine ANP32B or other mammalian ANP32A or B, shows stronger supporting activity to avian viral polymerase. Knockout of ANP32A in pig cells PK15 dramatically reduced avian influenza polymerase activity and viral infectivity, suggesting a unique feature of swANP32A in supporting avian influenza viral polymerase. This species-specific activity is mapped to two key sites, 106V and 156S, in swANP32A. Interestingly, the amino acid 106V is unique to pigs among all the vertebrate species studied, and when combined with 156S, exhibits positive epistasis in pigs. Mutation of 106V and 156S to the signature found in ANP32As from other mammalian species weakened the interaction between swANP32A and chicken viral polymerase, and reduced polymerase activity. Understanding the molecular basis of ANP32 proteins may help to discover new antiviral targets and design avian influenza resistant genome edited pigs.


Asunto(s)
Virus de la Influenza A/genética , Gripe Humana/genética , Porcinos/virología , Animales , Pollos , Especificidad del Huésped/genética , Especificidad del Huésped/inmunología , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H7N9 del Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Gripe Aviar/genética , Gripe Humana/virología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Infecciones por Orthomyxoviridae , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Porcinos/genética , Porcinos/metabolismo , Proteínas Virales/metabolismo , Replicación Viral
5.
J Virol ; 94(4)2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31776281

RESUMEN

A novel genus within the Orthomyxoviridae family was identified in the United States and named influenza D virus (IDV). Bovines have been proposed to be the primary host, and three main viral lineages (D/OK-like, D/660-like, and D/Japan-like) have been described. Experimental infections had previously been performed in swine, ferrets, calves, and guinea pigs in order to study IDV pathogenesis. We developed a murine experimental model to facilitate the study of IDV pathogenesis and the immune response. DBA/2 mice were inoculated with 105 50% tissue culture infective dose (TCID50) of D/bovine/France/5920/2014 (D/OK-like). No clinical signs or weight loss were observed. Viral replication was observed mainly in the upper respiratory tract (nasal turbinates) but also in the lower respiratory tract of infected mice, with a peak at 4 days postinfection. Moreover, the virus was also detected in the intestines. All infected mice seroconverted by 14 days postinfection. Transcriptomic analyses demonstrated that IDV induced the activation of proinflammatory genes, such as gamma interferon (IFN-γ) and CCL2. Inoculation of NF-κB-luciferase and Ifnar1-/- mice demonstrated that IDV induced mild inflammation and that a type I interferon response was not necessary in IDV clearance. Adaptation of IDV by serial passages in mice was not sufficient to induce disease or increased pathogenesis. Taken together, present data and comparisons with the calf model show that our mouse model allows for the study of IDV replication and fitness (before selected viruses may be inoculated on calves) and also of the immune response.IMPORTANCE Influenza D virus (IDV), a new genus of Orthomyxoviridae family, presents a large host range and a worldwide circulation. The pathogenicity of this virus has been studied in the calf model. The mouse model is frequently used to enable a first assessment of a pathogen's fitness, replication, and pathogenesis for influenza A and B viruses. We showed that DBA/2 mice are a relevant in vivo model for the study of IDV replication. This model will allow for rapid IDV fitness and replication evaluation and will enable phenotypic comparisons between isolated viruses. It will also allow for a better understanding of the immune response induced after IDV infection.


Asunto(s)
Especificidad del Huésped/inmunología , Infecciones por Orthomyxoviridae/inmunología , Thogotovirus/patogenicidad , Animales , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Infecciones del Sistema Respiratorio/virología , Seroconversión , Replicación Viral/inmunología
6.
PLoS Pathog ; 15(5): e1007710, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31145755

RESUMEN

Modified vaccinia virus Ankara (MVA) is the leading poxvirus vector for development of vaccines against diverse infectious diseases. This distinction is based on high expression of proteins and good immunogenicity despite an inability to assemble infectious progeny in human cells, which together promote efficacy and safety. Nevertheless, the basis for the host-range restriction is unknown despite past systematic attempts to identify the relevant missing viral gene(s). The search for host-range factors is exacerbated by the large number of deletions, truncations and mutations that occurred during the long passage history of MVA in chicken embryo fibroblasts. By whole genome sequencing of a panel of recombinant host-range extended (HRE) MVAs generated by marker rescue with 40 kbp segments of vaccinia virus DNA, we identified serine protease inhibitor 1 (SPI-1) as one of several candidate host-range factors present in those viruses that gained the ability to replicate in human cells. Electron microscopy revealed that the interruption of morphogenesis in human cells infected with MVA occurred at a similar stage as that of a vaccinia virus strain WR SPI-1 deletion mutant. Moreover, the introduction of the SPI-1 gene into the MVA genome led to more than a 2-log enhancement of virus spread in human diploid MRC-5 cells, whereas deletion of the gene diminished the spread of HRE viruses by similar extents. Furthermore, MRC-5 cells stably expressing SPI-1 also enhanced replication of MVA. A role for additional host range genes was suggested by the restoration of MVA replication to a lower level relative to HRE viruses, particularly in other human cell lines. Although multiple sequence alignments revealed genetic changes in addition to SPI-1 common to the HRE MVAs, no evidence for their host-range function was found by analysis thus far. Our finding that SPI-1 is host range factor for MVA should simplify use of high throughput RNAi or CRISPR/Cas single gene methods to identify additional viral and human restriction elements.


Asunto(s)
Especificidad del Huésped/inmunología , Inhibidores de Serina Proteinasa/inmunología , Virus Vaccinia/fisiología , Vaccinia/virología , Vacunas Virales/inmunología , Replicación Viral , Células A549 , Vectores Genéticos/inmunología , Humanos , Inhibidores de Serina Proteinasa/genética , Vaccinia/inmunología , Vaccinia/prevención & control
7.
J Gen Virol ; 101(6): 599-608, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32213247

RESUMEN

Infection of chicken coronavirus infectious bronchitis virus (IBV) is initiated by binding of the viral heavily N-glycosylated attachment protein spike to the alpha-2,3-linked sialic acid receptor Neu5Ac. Previously, we have shown that N-glycosylation of recombinantly expressed receptor binding domain (RBD) of the spike of IBV-M41 is of critical importance for binding to chicken trachea tissue. Here we investigated the role of N-glycosylation of the RBD on receptor specificity and virus replication in the context of the virus particle. Using our reverse genetics system we were able to generate recombinant IBVs for nine-out-of-ten individual N-glycosylation mutants. In vitro growth kinetics of these viruses were comparable to the virus containing the wild-type M41-S1. Furthermore, Neu5Ac binding by the recombinant viruses containing single N-glycosylation site knock-out mutations matched the Neu5Ac binding observed with the recombinant RBDs. Five N-glycosylation mutants lost the ability to bind Neu5Ac and gained binding to a different, yet unknown, sialylated glycan receptor on host cells. These results demonstrate that N-glycosylation of IBV is a determinant for receptor specificity.


Asunto(s)
Infecciones por Coronavirus/inmunología , Especificidad del Huésped/inmunología , Virus de la Bronquitis Infecciosa/química , Dominios Proteicos , Receptores Virales/inmunología , Glicoproteína de la Espiga del Coronavirus/química , Animales , Línea Celular , Embrión de Pollo , Infecciones por Coronavirus/virología , Glicosilación , Virus de la Bronquitis Infecciosa/inmunología , Riñón/citología , Riñón/embriología , Unión Proteica , Receptores de Superficie Celular/metabolismo , Receptores Virales/metabolismo , Proteínas Recombinantes , Glicoproteína de la Espiga del Coronavirus/metabolismo , Tropismo Viral/inmunología , Acoplamiento Viral , Replicación Viral
8.
Nat Rev Neurosci ; 16(7): 389-402, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26087680

RESUMEN

Nociceptors and immune cells both protect the host from potential threats to homeostasis. There is growing evidence for bidirectional signalling between these two systems, and the underlying mechanisms are beginning to be elucidated. An understanding is emerging of how both the adaptive and innate immune systems can activate and sensitize nociceptors, and, reciprocally, how nociceptors modulate immune cells. In this Review, we discuss how these interactions can be adaptive and useful to the organism but also consider when such signalling might be maladaptive and pathophysiological, contributing to immune-mediated diseases and persistent pain states.


Asunto(s)
Inmunidad Adaptativa/inmunología , Especificidad del Huésped/inmunología , Inmunidad Celular/inmunología , Inmunidad Innata/inmunología , Nociceptores/inmunología , Animales , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Enfermedades del Sistema Inmune/inmunología , Enfermedades del Sistema Inmune/metabolismo , Nociceptores/metabolismo , Dolor/inmunología , Dolor/metabolismo
9.
Infect Immun ; 87(12)2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31548317

RESUMEN

Most Salmonella serovars cause disease in many host species, while a few serovars have evolved to be host specific. Very little is known about the mechanisms that contribute to Salmonella host specificity. We compared the interactions between chicken primary macrophages (CDPM) and host-generalist serovar Salmonella enterica serovar Typhimurium, host-adapted Salmonella enterica serovar Dublin, and avian host-specific Salmonella enterica serovar Gallinarum. S Gallinarum was taken up in lower numbers by CDPM than S Typhimurium and S Dublin; however, a higher survival rate was observed for this serovar. In addition, S Typhimurium and S Dublin caused substantially higher levels of cell death to the CDPM, while significantly higher concentrations of NO were produced by S Gallinarum-infected cells. Global transcriptome analysis performed 2 h postinfection showed that S Gallinarum infection triggered a more comprehensive response in CDPM with 1,114 differentially expressed genes (DEGs) compared to the responses of S Typhimurium (625 DEGs) and S Dublin (656 DEGs). Comparable levels of proinflammation responses were observed in CDPM infected by these three different serovars at the initial infection phase, but a substantially quicker reduction in levels of interleukin-1ß (IL-1ß), CXCLi1, and CXCLi2 gene expression was detected in the S Gallinarum-infected macrophages than that of two other groups as infections proceeded. KEGG cluster analysis for unique DEGs after S Gallinarum infection showed that the JAK-STAT signaling pathway was top enriched, indicating a specific role for this pathway in response to S Gallinarum infection of CDPM. Together, these findings provide new insights into the interaction between Salmonella and the host and increase our understanding of S Gallinarum host specificity.


Asunto(s)
Especificidad del Huésped/inmunología , Macrófagos/inmunología , Enfermedades de las Aves de Corral/inmunología , Salmonelosis Animal/inmunología , Salmonella typhimurium/inmunología , Animales , Bovinos , Células Cultivadas , Pollos , Interleucina-1beta/inmunología , Óxido Nítrico/metabolismo , Enfermedades de las Aves de Corral/microbiología , Salmonella typhimurium/clasificación
10.
Med Mycol ; 56(suppl_1): 126-143, 2018 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-29538731

RESUMEN

The description of cryptic species with different pathogenic potentials has changed the perspectives on sporotrichosis. Sporothrix schenckii causes a benign chronic subcutaneous mycosis, Sporothrix brasiliensis is highly virulent, and Sporothrix globosa mainly causes fixed cutaneous lesions. Furthermore, S. brasiliensis is the prevalent species related to cat-transmitted sporotrichosis. Sources of infection, transmission, and distribution patterns also differ between species, and variability differs between species because of different degrees of clonality. The present review article will cover several aspects of the biology of clinically relevant agents of sporotrichosis, including epidemiological aspects of emerging species. Genomic information of Sporothrix spp. is also discussed. The cell wall is an essential structure for cell viability, interaction with the environment, and the host immune cells and contains several macromolecules involved in virulence. Due to its importance, aspects of glycosylation and cell wall polysaccharides are reviewed. Recent genome data and bioinformatics analyses helped to identify specific enzymes of the biosynthetic glycosylation routes, with no homologs in mammalian cells, which can be putative targets for development of antifungal drugs. A diversity of molecular techniques is available for the recognition of the clinically relevant species of Sporothrix. Furthermore, antigens identified as diagnostic markers and putative vaccine candidates are described. Cell-mediated immunity plays a key role in controlling infection, but Sporothrix species differ in their interaction with the host. The adaptive branch of the immune response is essential for appropriate control of infection.


Asunto(s)
Sporothrix/fisiología , Esporotricosis/diagnóstico , Esporotricosis/inmunología , Animales , Antígenos Fúngicos/inmunología , Pared Celular/química , Pared Celular/metabolismo , Genoma Fúngico , Especificidad del Huésped/inmunología , Humanos , Técnicas de Diagnóstico Molecular , Sporothrix/clasificación , Sporothrix/inmunología , Esporotricosis/microbiología , Esporotricosis/transmisión , Virulencia
11.
J Gen Virol ; 98(3): 311-321, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27902345

RESUMEN

Canine distemper virus (CDV) has emerged as a significant disease of wildlife, which is highly contagious and readily transmitted between susceptible hosts. Initially described as an infectious disease of domestic dogs, it is now recognized as a global multi-host pathogen, infecting and causing mass mortalities in a wide range of carnivore species. The last decade has seen the effect of numerous CDV outbreaks in various wildlife populations. Prevention of CDV requires a clear understanding of the potential hosts in danger of infection as well as the dynamic pathways CDV uses to gain entry to its host cells and its ability to initiate viral shedding and disease transmission. We review recent research conducted on CDV infections in wildlife, including the latest findings on the causes of host specificity and cellular receptors involved in distemper pathogenesis.


Asunto(s)
Animales Salvajes/virología , Virus del Moquillo Canino , Moquillo/virología , Perros/virología , Especificidad del Huésped/inmunología , Animales , Brotes de Enfermedades/prevención & control , Moquillo/diagnóstico , Moquillo/epidemiología , Moquillo/inmunología , Virus del Moquillo Canino/química , Virus del Moquillo Canino/genética , Virus del Moquillo Canino/patogenicidad , Virus del Moquillo Canino/ultraestructura
12.
J Biol Chem ; 290(21): 13440-53, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25837248

RESUMEN

Lipid A in LPS activates innate immunity through the Toll-like receptor 4 (TLR4)-MD-2 complex on host cells. Variation in lipid A has significant consequences for TLR4 activation and thus may be a means by which Gram-negative bacteria modulate host immunity. However, although even minor changes in lipid A structure have been shown to affect downstream immune responses, the mechanism by which the TLR4-MD-2 receptor complex recognizes these changes is not well understood. We previously showed that strain BP338 of the human pathogen Bordetella pertussis, the causative agent of whooping cough, modifies its lipid A by the addition of glucosamine moieties that promote TLR4 activation in human, but not mouse, macrophages. Using site-directed mutagenesis and an NFκB reporter assay screen, we have identified several charged amino acid residues in TLR4 and MD-2 that are important for these species-specific responses; some of these are novel for responses to penta-acyl B. pertussis LPS, and their mutation does not affect the response to hexa-acylated Escherichia coli LPS or tetra-acylated lipid IVA. We additionally show evidence that suggests that recognition of penta-acylated B. pertussis lipid A is dependent on uncharged amino acids in TLR4 and MD-2 and that this is true for both human and mouse TLR4-MD-2 receptors. Taken together, we have demonstrated that the TLR4-MD-2 receptor complex recognizes variation in lipid A molecules using multiple sites for receptor-ligand interaction and propose that host-specific immunity to a particular Gram-negative bacterium is, at least in part, mediated by very subtle tuning of one of the earliest interactions at the host-pathogen interface.


Asunto(s)
Aminoácidos/química , Bordetella pertussis/inmunología , Especificidad del Huésped/inmunología , Lípido A/química , Lípido A/inmunología , Antígeno 96 de los Linfocitos/metabolismo , Receptor Toll-Like 4/metabolismo , Aminoácidos/inmunología , Aminoácidos/metabolismo , Animales , Western Blotting , Células Cultivadas , Glucosamina/metabolismo , Células HEK293 , Humanos , Inmunidad Innata/inmunología , Lípido A/metabolismo , Antígeno 96 de los Linfocitos/genética , Antígeno 96 de los Linfocitos/inmunología , Ratones , Mutagénesis Sitio-Dirigida , Mutación/genética , Conformación Proteica , Transducción de Señal , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
13.
J Virol ; 89(22): 11507-22, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26339062

RESUMEN

UNLABELLED: Infections of domestic and wild birds with low-pathogenic avian influenza viruses (LPAIVs) have been associated with protective immunity to subsequent infection. However, the degree and duration of immunity in wild birds from previous LPAIV infection, by the same or a different subtype, are poorly understood. Therefore, we inoculated H13N2 (A/black-headed gull/Netherlands/7/2009) and H16N3 (A/black-headed gull/Netherlands/26/2009) LPAIVs into black-headed gulls (Chroicocephalus ridibundus), their natural host species, and measured the long-term immune response and protection against one or two reinfections over a period of >1 year. This is the typical interval between LPAIV epizootics in wild birds. Reinfection with the same virus resulted in progressively less virus excretion, with complete abrogation of virus excretion after two infections for H13 but not H16. However, reinfection with the other virus affected neither the level nor duration of virus excretion. Virus excretion by immunologically naive birds did not differ in total levels of excreted H13 or H16 virus between first- and second-year birds, but the duration of H13 excretion was shorter for second-year birds. Furthermore, serum antibody levels did not correlate with protection against LPAIV infection. LPAIV-infected gulls showed no clinical signs of disease. These results imply that the epidemiological cycles of H13 and H16 in black-headed gulls are relatively independent from each other and depend mainly on infection of first-year birds. IMPORTANCE: Low-pathogenic avian influenza viruses (LPAIVs) circulate mainly in wild water birds but are occasionally transmitted to other species, including humans, where they cause subclinical to fatal disease. To date, the effect of LPAIV-specific immunity on the epidemiology of LPAIV in wild birds is poorly understood. In this study, we investigated the effect of H13 and H16 LPAIV infection in black-headed gulls on susceptibility and virus excretion of subsequent infection with the same or the other virus within the same breeding season and between breeding seasons. These are the only two LPAIV hemagglutinin subtypes predominating in this species. The findings suggest that H13 and H16 LPAIV cycles in black-headed gull populations are independent of each other, indicate the importance of first-year birds in LPAIV epidemiology, and emphasize the need for alternatives to avian influenza virus (AIV)-specific serum antibodies as evidence of past LPAIV infection and correlates of protection against LPAIV infection in wild birds.


Asunto(s)
Charadriiformes/virología , Resistencia a la Enfermedad/inmunología , Hemaglutininas Virales/inmunología , Virus de la Influenza A/inmunología , Gripe Aviar/inmunología , Factores de Edad , Animales , Anticuerpos Antivirales/sangre , Protección Cruzada/inmunología , Susceptibilidad a Enfermedades , Hemaglutininas Virales/clasificación , Especificidad del Huésped/genética , Especificidad del Huésped/inmunología , Inmunidad Humoral/inmunología , Inmunización , Virus de la Influenza A/genética , Gripe Aviar/epidemiología , Gripe Aviar/virología , Datos de Secuencia Molecular , Recurrencia , Esparcimiento de Virus/inmunología
14.
Biologicals ; 44(4): 252-256, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27230301

RESUMEN

A novel triple reasserted H1N2 virus A/swine/Shanghai/1/2007 (SH07) was isolated from nasal swabs of weaned pig showing clinical symptoms of coughing and sneezing. To explore the virus characteristics, mice, chickens and pigs were selected for pathogenicity study. Pigs inoculated intranasally with 10(6) TCID50 SH07 showed clinical symptoms with coughing and sneezing, but no death. The virus nuclear acid was detected in many tissues using real-time PCR, which was mainly distributed in respiratory system particularly in the lungs. The virus was low-pathogenic to chickens with 10(6) TCID50 dose inoculation either via intramuscular or intranasal routes. However virus nuclear acid detection and virus isolation confirmed that the virus can also be found in nasal and rectum. When virus was inoculated into mice by intramuscular or intranasal routes we observed 100% and 80% lethality respectively. The third generation of samples passaged on MDCK cell were SIV positive in indirect immunofluorescence assay (IFA) using antiserum against H1N2 SIV. Furthermore, the lungs of mice showed obvious lesion with interstitial pneumonia. Data in our study suggest that SH07 is preferentially pathogenic to mammals rather than birds although it is a reasserting virus with the fragments from swine, human and avian origin.


Asunto(s)
Subtipo H1N2 del Virus de la Influenza A/inmunología , Gripe Aviar/inmunología , Infecciones por Orthomyxoviridae/inmunología , Enfermedades de los Porcinos/inmunología , Animales , Pollos , Perros , Especificidad del Huésped/inmunología , Humanos , Subtipo H1N2 del Virus de la Influenza A/genética , Subtipo H1N2 del Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Pulmón/inmunología , Pulmón/virología , Células de Riñón Canino Madin Darby , Ratones , Microscopía Fluorescente , Cavidad Nasal/inmunología , Cavidad Nasal/virología , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Orthomyxoviridae/virología , Recto/inmunología , Recto/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos , Enfermedades de los Porcinos/virología , Virulencia/inmunología
15.
J Gen Virol ; 95(Pt 6): 1193-1210, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24584475

RESUMEN

Typical avian influenza A viruses are restricted from replicating efficiently and causing disease in humans. However, an avian virus can become adapted to humans by mutating or recombining with currently circulating human viruses. These viruses have the potential to cause pandemics in an immunologically naïve human population. It is critical that we understand the molecular basis of host-range restriction and how this can be overcome. Here, we review our current understanding of the mechanisms by which influenza viruses adapt to replicate efficiently in a new host. We predominantly focus on the influenza polymerase, which remains one of the least understood host-range barriers.


Asunto(s)
Virus de la Influenza A/patogenicidad , Adaptación Fisiológica , Animales , Aves , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/fisiología , Genes Virales , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/fisiología , Especificidad del Huésped/genética , Especificidad del Huésped/inmunología , Especificidad del Huésped/fisiología , Humanos , Inmunidad Innata , Virus de la Influenza A/genética , Virus de la Influenza A/fisiología , Gripe Aviar/inmunología , Gripe Aviar/virología , Gripe Humana/inmunología , Gripe Humana/virología , Mutación , Neuraminidasa/genética , Neuraminidasa/fisiología , Nucleoproteínas/genética , Nucleoproteínas/fisiología , Proteínas Virales/genética , Proteínas Virales/fisiología
17.
PLoS Pathog ; 7(7): e1002142, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21829348

RESUMEN

Chikungunya virus (CHIKV) is a reemerging mosquito-borne pathogen that has recently caused devastating urban epidemics of severe and sometimes chronic arthralgia. As with most other mosquito-borne viral diseases, control relies on reducing mosquito populations and their contact with people, which has been ineffective in most locations. Therefore, vaccines remain the best strategy to prevent most vector-borne diseases. Ideally, vaccines for diseases of resource-limited countries should combine low cost and single dose efficacy, yet induce rapid and long-lived immunity with negligible risk of serious adverse reactions. To develop such a vaccine to protect against chikungunya fever, we employed a rational attenuation mechanism that also prevents the infection of mosquito vectors. The internal ribosome entry site (IRES) from encephalomyocarditis virus replaced the subgenomic promoter in a cDNA CHIKV clone, thus altering the levels and host-specific mechanism of structural protein gene expression. Testing in both normal outbred and interferon response-defective mice indicated that the new vaccine candidate is highly attenuated, immunogenic and efficacious after a single dose. Furthermore, it is incapable of replicating in mosquito cells or infecting mosquitoes in vivo. This IRES-based attenuation platform technology may be useful for the predictable attenuation of any alphavirus.


Asunto(s)
Infecciones por Alphavirus , Virus Chikungunya , Especificidad del Huésped/inmunología , Vacunas Virales , Infecciones por Alphavirus/genética , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/prevención & control , Infecciones por Alphavirus/virología , Animales , Vectores Artrópodos/inmunología , Vectores Artrópodos/virología , Fiebre Chikungunya , Virus Chikungunya/genética , Virus Chikungunya/inmunología , Chlorocebus aethiops , Especificidad del Huésped/genética , Ratones , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Células Vero , Vacunas Virales/genética , Vacunas Virales/inmunología
18.
Diabetes Obes Metab ; 15 Suppl 3: 61-70, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24003922

RESUMEN

Over the last decade, the research community has revealed the role of a new organ: the intestinal microbiota. It is considered as a symbiont that is part of our organism since, at birth, it educates the immune system and contributes to the development of the intestinal vasculature and most probably the nervous system. With the advent of new generation sequencing techniques, a catalogue of genes that belong to this microbiome has been established that lists more than 5 million non-redundant genes called the metagenome. Using germ free mice colonized with the microbiota from different origins, it has been formally demonstrated that the intestinal microbiota causes the onset of metabolic diseases. Further to the role of point mutations in our genome, the microbiota can explain the on-going worldwide pandemic of obesity and diabetes, its dissemination and family inheritance, as well as the diversity of the associated metabolic phenotypes. More recently, the discovery of bacterial DNA within host tissues, such as the liver, the adipose tissue and the blood, which establishes a tissue microbiota, introduces new opportunities to identify targets and predictive biomarkers based on the host to microbiota interaction, as well as to define new strategies for pharmacological, immunomodulatory vaccines and nutritional applications.


Asunto(s)
Metabolismo/fisiología , Metagenoma/fisiología , Microbiota/fisiología , Animales , Comunicación Celular/fisiología , Especificidad del Huésped/inmunología , Humanos , Intestinos/inmunología , Intestinos/microbiología , Enfermedades Metabólicas/microbiología , Ratones
19.
J Immunol ; 186(1): 230-41, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21098219

RESUMEN

Graft-versus-host disease (GVHD) is initiated by APCs that prime alloreactive donor T cells. In antipathogen responses, Ag-bearing APCs receive signals through pattern-recognition receptors, including TLRs, which induce the expression of costimulatory molecules and production of inflammatory cytokines, which in turn mold the adaptive T cell response. However, in allogeneic hematopoietic stem cell transplantation (alloSCT), there is no specific pathogen, alloantigen is ubiquitous, and signals that induce APC maturation are undefined. To investigate APC activation in GVHD, we used recipient mice with hematopoietic cells genetically deficient in pathways critical for APC maturation in models in which host APCs are absolutely required. Strikingly, CD8-mediated and CD4-mediated GVHD were similar whether host APCs were wild-type or deficient in MyD88, TRIF, or MyD88 and TRIF, which excludes essential roles for TLRs and IL-1ß, the key product of inflammasome activation. Th1 differentiation was if anything augmented when APCs were MyD88/TRIF(-/-), and T cell production of IFN-γ did not require host IL-12. GVHD was also intact when APCs lacked the type I IFNR, which amplifies APC activation pathways that induce type I IFNs. Thus in GVHD, alloreactive T cells can be activated when pathways critical for antipathogen T cell responses are impaired.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Hematopoyesis/inmunología , Inmunidad Innata , Transducción de Señal/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/microbiología , Células Presentadoras de Antígenos/virología , Trasplante de Médula Ósea/inmunología , Trasplante de Médula Ósea/patología , Enfermedad Injerto contra Huésped/microbiología , Enfermedad Injerto contra Huésped/virología , Hematopoyesis/genética , Especificidad del Huésped/genética , Especificidad del Huésped/inmunología , Inmunidad Innata/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Quimera por Radiación/inmunología , Transducción de Señal/genética , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/fisiología
20.
Adv Exp Med Biol ; 783: 33-44, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23468102

RESUMEN

The host response to mycobacterial infection including tuberculosis depends on genetically controlled host and bacterial factors and their interaction. A largely unknown aspect of this interaction is whether disease results from an additive and independent effect of host and pathogen or from specific host-pathogen combinations. The preferential association of specific mycobacterial strains with specific ethnic groups provided tentative evidence in favor of host-pathogen specificity in tuberculosis and is consistent with the hypothesis of host-mycobacterial co-adaptation. Substantial evidence for specificity has now been provided by animal models and human case-control association studies. These studies indicate that differences in the host response to infection are at least in part due to specific combinations of host genetic factors and genetic and phenotypic characteristics of the infecting mycobacterial strain.


Asunto(s)
Especificidad del Huésped , Mycobacterium tuberculosis/fisiología , Tuberculosis/microbiología , Animales , Araquidonato 5-Lipooxigenasa/genética , Araquidonato 5-Lipooxigenasa/fisiología , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/fisiología , Etnicidad/genética , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/fisiología , Predisposición Genética a la Enfermedad , Salud Global , Especificidad del Huésped/inmunología , Humanos , Lectina de Unión a Manosa/genética , Lectina de Unión a Manosa/fisiología , Ratones , Ratones Endogámicos , Mycobacterium bovis/patogenicidad , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/patogenicidad , Especificidad de la Especie , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Tuberculosis/etnología , Tuberculosis/genética , Tuberculosis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA