Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Emerg Microbes Infect ; 10(1): 2199-2201, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34749583

RESUMEN

We report pilot studies to evaluate the susceptibility of common domestic livestock (cattle, sheep, goat, alpaca, rabbit, and horse) to intranasal infection with SARS-CoV-2. None of the infected animals shed infectious virus via nasal, oral, or faecal routes, although viral RNA was detected in several animals. Further, neutralizing antibody titres were low or non-existent one month following infection. These results suggest that domestic livestock are unlikely to contribute to SARS-CoV-2 epidemiology.


Asunto(s)
COVID-19/veterinaria , Especificidad del Huésped , Ganado/virología , SARS-CoV-2/patogenicidad , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , COVID-19/virología , Camélidos del Nuevo Mundo/virología , Bovinos/virología , Chlorocebus aethiops , Reservorios de Enfermedades/virología , Cabras/virología , Caballos/virología , Especificidad del Huésped/inmunología , Humanos , Cavidad Nasal/virología , ARN Viral/análisis , Conejos/virología , Recto/virología , Sistema Respiratorio/virología , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Ovinos/virología , Especificidad de la Especie , Células Vero , Esparcimiento de Virus , Vísceras/virología
2.
Front Immunol ; 12: 769543, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790204

RESUMEN

Many pathogens encode proteases that serve to antagonize the host immune system. In particular, viruses with a positive-sense single-stranded RNA genome [(+)ssRNA], including picornaviruses, flaviviruses, and coronaviruses, encode proteases that are not only required for processing viral polyproteins into functional units but also manipulate crucial host cellular processes through their proteolytic activity. Because these proteases must cleave numerous polyprotein sites as well as diverse host targets, evolution of these viral proteases is expected to be highly constrained. However, despite this strong evolutionary constraint, mounting evidence suggests that viral proteases such as picornavirus 3C, flavivirus NS3, and coronavirus 3CL, are engaged in molecular 'arms races' with their targeted host factors, resulting in host- and virus-specific determinants of protease cleavage. In cases where protease-mediated cleavage results in host immune inactivation, recurrent host gene evolution can result in avoidance of cleavage by viral proteases. In other cases, such as recently described examples in NLRP1 and CARD8, hosts have evolved 'tripwire' sequences that mimic protease cleavage sites and activate an immune response upon cleavage. In both cases, host evolution may be responsible for driving viral protease evolution, helping explain why viral proteases and polyprotein sites are divergent among related viruses despite such strong evolutionary constraint. Importantly, these evolutionary conflicts result in diverse protease-host interactions even within closely related host and viral species, thereby contributing to host range, zoonotic potential, and pathogenicity of viral infection. Such examples highlight the importance of examining viral protease-host interactions through an evolutionary lens.


Asunto(s)
Sistema Inmunológico/inmunología , Proteasas Virales/inmunología , Animales , Evolución Molecular , Especificidad del Huésped/genética , Especificidad del Huésped/inmunología , Humanos , Proteasas Virales/genética , Proteínas Virales/genética , Proteínas Virales/inmunología
3.
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
4.
Nat Rev Microbiol ; 19(10): 639-653, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34045709

RESUMEN

Developing general principles of host-microorganism interactions necessitates a robust understanding of the eco-evolutionary processes that structure microbiota. Phylosymbiosis, or patterns of microbiome composition that can be predicted by host phylogeny, is a unique framework for interrogating these processes. Identifying the contexts in which phylosymbiosis does and does not occur facilitates an evaluation of the relative importance of different ecological processes in shaping the microbial community. In this Review, we summarize the prevalence of phylosymbiosis across the animal kingdom on the basis of the current literature and explore the microbial community assembly processes and related host traits that contribute to phylosymbiosis. We find that phylosymbiosis is less prevalent in taxonomically richer microbiomes and hypothesize that this pattern is a result of increased stochasticity in the assembly of complex microbial communities. We also note that despite hosting rich microbiomes, mammals commonly exhibit phylosymbiosis. We hypothesize that this pattern is a result of a unique combination of mammalian traits, including viviparous birth, lactation and the co-evolution of haemochorial placentas and the eutherian immune system, which compound to ensure deterministic microbial community assembly. Examining both the individual and the combined importance of these traits in driving phylosymbiosis provides a new framework for research in this area moving forward.


Asunto(s)
Microbioma Gastrointestinal , Interacciones Microbiota-Huesped/genética , Especificidad del Huésped/genética , Mamíferos/microbiología , Filogenia , Simbiosis/genética , Animales , Interacciones Microbiota-Huesped/inmunología , Interacciones Microbiota-Huesped/fisiología , Especificidad del Huésped/inmunología , Especificidad del Huésped/fisiología , Mamíferos/inmunología , Ratones , ARN Ribosómico 16S , Simbiosis/fisiología
5.
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
6.
Sci Rep ; 11(1): 3493, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33568758

RESUMEN

Yersinia ruckeri is the causative agent of enteric redmouth disease (ERM) which causes economically significant losses in farmed salmonids, especially Atlantic salmon (Salmo salar) and rainbow trout (Oncorhynchus mykiss, Walbaum). However, very little is known about the genetic relationships of disease-causing isolates in these two host species or about factors responsible for disease. Phylogenetic analyses of 16 representative isolates based on the nucleotide sequences of 19 housekeeping genes suggests that pathogenic Atlantic salmon and rainbow trout isolates represent distinct host-specific lineages. However, the apparent phylogenies of certain isolates has been influenced by horizontal gene transfer and recombinational exchange. Splits decomposition analysis demonstrated a net-like phylogeny based on the housekeeping genes, characteristic of recombination. Comparative analysis of the distribution of individual housekeeping gene alleles across the isolates demonstrated evidence of genomic mosaicism and recombinational exchange involving certain Atlantic salmon and rainbow trout isolates. Comparative nucleotide sequence analysis of the key outer membrane protein genes ompA and ompF revealed that the corresponding gene trees were both non-congruent with respect to the housekeeping gene phylogenies providing evidence that horizontal gene transfer has influenced the evolution of both these surface protein-encoding genes. Analysis of inferred amino acid sequence variation in OmpA identified a single variant, OmpA.1, that was present in serotype O1 and O8 isolates representing typical pathogenic strains in rainbow trout and Atlantic salmon, respectively. In particular, the sequence of surface-exposed loop 3 differed by seven amino acids to that of other Y. ruckeri isolates. These findings suggest that positive selection has likely influenced the presence of OmpA.1 in these isolates and that loop 3 may play an important role in virulence. Amino acid sequence variation of OmpF was greater than that of OmpA and was similarly restricted mainly to the surface-exposed loops. Two OmpF variants, OmpF.1 and OmpF.2, were associated with pathogenic rainbow trout and Atlantic salmon isolates, respectively. These OmpF proteins had very similar amino acid sequences suggesting that positive evolutionary pressure has also favoured the selection of these variants in pathogenic strains infecting both species.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Oncorhynchus mykiss/genética , Yersiniosis/virología , Yersinia ruckeri/virología , Animales , Enfermedades de los Peces/virología , Especificidad del Huésped/inmunología , Filogenia , Serogrupo , Virulencia/genética , Virulencia/fisiología
7.
JCI Insight ; 6(2)2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33491669

RESUMEN

Pneumocystis is an important opportunistic fungus that causes pneumonia in children and immunocompromised individuals. Recent genomic data show that divergence of major surface glycoproteins may confer speciation and host range selectivity. On the other hand, immune clearance between mice and humans is well correlated. Thus, we hypothesized that humanize mice may provide information about human immune responses involved in controlling Pneumocystis infection. CD34-engrafted huNOG-EXL mice controlled fungal burdens to a greater extent than nonengrafted mice. Moreover, engrafted mice generated fungal-specific IgM. Fungal control was associated with a transcriptional signature that was enriched for genes associated with nonopsonic recognition of trophs (CD209) and asci (CLEC7A). These same genes were downregulated in CD4-deficient mice as well as twins with bare lymphocyte syndrome with Pneumocystis pneumonia.


Asunto(s)
Neumonía por Pneumocystis/inmunología , Animales , Anticuerpos Antifúngicos/biosíntesis , Antígenos CD34/metabolismo , Moléculas de Adhesión Celular/genética , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Xenoinjertos , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Especificidad del Huésped/inmunología , Humanos , Inmunoglobulina G/biosíntesis , Inmunoglobulina M/biosíntesis , Lectinas Tipo C/genética , Pulmón/inmunología , Pulmón/microbiología , Ratones , Ratones Transgénicos , Pneumocystis/inmunología , Pneumocystis/patogenicidad , Neumonía por Pneumocystis/genética , Neumonía por Pneumocystis/microbiología , Receptores de Superficie Celular/genética , Especificidad de la Especie
8.
Sci Rep ; 10(1): 6779, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-32322086

RESUMEN

Novel disease emergence is often associated with changes in pathogen traits that enable pathogen colonisation, persistence and transmission in the novel host environment. While understanding the mechanisms underlying disease emergence is likely to have critical implications for preventing infectious outbreaks, such knowledge is often based on studies of viral pathogens, despite the fact that bacterial pathogens may exhibit very different life histories. Here, we investigate the ability of epizootic outbreak strains of the bacterial pathogen, Mycoplasma gallisepticum, which jumped from poultry into North American house finches (Haemorhous mexicanus), to interact with model avian cells. We found that house finch epizootic outbreak strains of M. gallisepticum displayed a greater ability to adhere to, invade, persist within and exit from cultured chicken embryonic fibroblasts, than the reference virulent (R_low) and attenuated (R_high) poultry strains. Furthermore, unlike the poultry strains, the house finch epizootic outbreak strain HF_1994 displayed a striking lack of cytotoxicity, even exerting a cytoprotective effect on avian cells. Our results suggest that, at epizootic outbreak in house finches, M. gallisepticum was particularly adept at using the intra-cellular environment, which may have facilitated colonisation, dissemination and immune evasion within the novel finch host. Whether this high-invasion phenotype is similarly displayed in interactions with house finch cells, and whether it contributed to the success of the host shift, remains to be determined.


Asunto(s)
Pinzones/inmunología , Especificidad del Huésped/inmunología , Mycoplasma gallisepticum/inmunología , Aves de Corral/inmunología , Animales , Línea Celular , Pinzones/microbiología , Interacciones Microbiota-Huesped/inmunología , Infecciones por Mycoplasma/inmunología , Infecciones por Mycoplasma/microbiología , Mycoplasma gallisepticum/fisiología , Aves de Corral/microbiología
9.
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
10.
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
11.
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
12.
Ecohealth ; 16(4): 726-733, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31664588

RESUMEN

Mosquito-borne flaviviruses (MBFVs) are of public and animal health concern because they cause millions of human deaths annually and impact domestic animals and wildlife globally. MBFVs are phylogenetically divided into two clades, one is transmitted by Aedes mosquitoes (Ae-MBFVs) associated with mammals and the other by Culex mosquitoes (Cx-MBFVs) associated with birds. However, this assumption has not been evaluated. Here, we synthesized 79 published reports of MBFVs from wild mammals, estimating their host. Then, we tested whether the host specificity was biased to sampling and investigation efforts or to phylogenetic relationships using a viral phylogenetic tree drawn from analyzing whole flavivirus genomes obtained in GenBank. We found in total 18 flaviviruses, nine related to Aedes spp. and nine to Culex spp. infecting 129 mammal species. Thus, this supports that vectors are transmitting MBFV across available host clades and that ornithophilic mosquitoes are readily infecting mammals. Although most of the mosquito species are generalists in their host-feeding preferences, we also found a certain degree of MBFV's specificity, as most of them infect closely related mammal species. The present study integrates knowledge regarding MBFVs, and it may help to understand their transmission dynamics between viruses, vectors, and mammal hosts.


Asunto(s)
Interacciones Microbiota-Huesped/inmunología , Especificidad del Huésped/genética , Especificidad del Huésped/inmunología , Mosquitos Vectores/virología , Fiebre del Nilo Occidental/inmunología , Fiebre del Nilo Occidental/transmisión , Virus del Nilo Occidental/aislamiento & purificación , Aedes/virología , Animales , Animales Domésticos/virología , Culex/virología , Interacciones Microbiota-Huesped/genética , Mamíferos/genética , Mamíferos/virología
13.
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
14.
Front Immunol ; 10: 1688, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31379874

RESUMEN

Endoparasitoid wasps, which lay eggs inside the bodies of other insects, use various strategies to protect their offspring from the host immune response. The hymenopteran species of the genus Leptopilina, parasites of Drosophila, rely on the injection of a venom which contains proteins and peculiar vesicles (hereafter venosomes). We show here that the injection of purified L. boulardi venosomes is sufficient to impair the function of the Drosophila melanogaster lamellocytes, a hemocyte type specialized in the defense against wasp eggs, and thus the parasitic success of the wasp. These venosomes seem to have a unique extracellular biogenesis in the wasp venom apparatus where they acquire specific secreted proteins/virulence factors and act as a transport system to deliver these compounds into host lamellocytes. The level of venosomes entry into lamellocytes of different Drosophila species was correlated with the rate of parasitism success of the wasp, suggesting that this venosome-cell interaction may represent a new evolutionary level of host-parasitoid specificity.


Asunto(s)
Drosophila melanogaster/inmunología , Vesículas Extracelulares/inmunología , Especificidad del Huésped/inmunología , Factores de Virulencia/inmunología , Venenos de Avispas/inmunología , Avispas/inmunología , Animales , Hemocitos/inmunología , Interacciones Huésped-Parásitos
15.
PLoS One ; 14(7): e0219000, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31269085

RESUMEN

In accordance with the Red Queen hypothesis, the lower genotypic diversity in clonally reproducing species should make them easier targets for pathogen infection, especially when closely related sexually reproducing species occur in close proximity. We analyzed two populations of clonal P. formosa and their sexual parental species P. mexicana by correlating individual parasite infection with overall and immune genotype. Our study revealed lower levels of overall genotypic diversity and marginally fewer MHC class I alleles in P. formosa individuals compared to sexually reproducing P. mexicana. Parasite load, however, differed only between field sites but not between species. We hypothesize that this might be due to slightly higher genotypic diversity in P. formosa at the innate immune system (toll like receptor 8) which is likely due to the species' hybrid origin. In consequence, it appears that clonal individuals do not necessarily suffer a disadvantage compared to sexual individuals when fighting parasite infection.


Asunto(s)
Poecilia/fisiología , Poecilia/parasitología , Reproducción Asexuada/genética , Reproducción Asexuada/inmunología , Inmunidad Adaptativa/genética , Alelos , Animales , Femenino , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Genes MHC Clase I , Variación Genética , Genotipo , Especificidad del Huésped/genética , Especificidad del Huésped/inmunología , Inmunidad Innata/genética , Masculino , México , Modelos Genéticos , Modelos Inmunológicos , Carga de Parásitos , Poecilia/genética , Reproducción/genética , Reproducción/inmunología , Especificidad de la Especie , Receptor Toll-Like 8/genética , Receptor Toll-Like 8/inmunología
16.
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
17.
Sci Rep ; 9(1): 6134, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30992458

RESUMEN

Mastitis is the most economically important disease affecting dairy cattle worldwide. Staphylococcus aureus is a highly prevalent cause of mastitis, causing infections ranging from sub-clinical to gangrenous. However, the interaction between the genotype of the infecting strain of S. aureus and the host response remains largely uncharacterised. To better understand the variation in presentation and outcomes of S. aureus-mediated bovine mastitis, we studied the interaction of a panel of mastitis isolates from several prominent bovine-associated lineages with bovine mammary epithelial cells (bMEC) and neutrophils. Significant differences in immune gene expression by infected primary or immortalised bMEC, or their elaboration of neutrophil chemoattractants, were observed and were dependent on the lineage of the infecting strain. Differences were also apparent in the invasiveness of S. aureus strains and their ability to survive killing by neutrophils. Our results demonstrate that a range of immune responses occur, suggesting the importance of S. aureus strain in dictating mastitis disease course. S. aureus lineages may therefore have adopted differing strategies for exploitation of the intramammary niche. Consequently, improved diagnosis of infecting lineage may enable better prognosis for S. aureus mastitis and reduce morbidity and economic loss.


Asunto(s)
Genotipo , Especificidad del Huésped/genética , Interacciones Huésped-Patógeno/genética , Mastitis Bovina/inmunología , Staphylococcus aureus/inmunología , Animales , Bovinos , Células Cultivadas , Quimiotaxis/inmunología , Citocinas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Femenino , Regulación de la Expresión Génica/inmunología , Granulocitos/inmunología , Granulocitos/metabolismo , Especificidad del Huésped/inmunología , Interacciones Huésped-Patógeno/inmunología , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/microbiología , Glándulas Mamarias Animales/patología , Mastitis Bovina/microbiología , Mastitis Bovina/patología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Cultivo Primario de Células , Staphylococcus aureus/genética , Staphylococcus aureus/aislamiento & purificación
18.
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
19.
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
20.
Microbes Infect ; 20(2): 101-110, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29024796

RESUMEN

Streptobacillus (S.) moniliformis is a rat-associated zoonotic pathogen that occasionally causes disease in other species. We investigated the working hypothesis that intranasal infection might lead to different immune responses in C57BL/6 and BALB/c mice associated with distinct pathologies. This study confirmed with 75% mortality the known high susceptibility of C57BL/6 mice to Streptobacillus moniliformis infection in comparison to BALB/c mice which did not develop signs of disease. Main pathologies in C57BL/6 mice were purulent to necrotizing lymphadenitis and pneumonia. Significant seroconversion was recorded in surviving mice of both strains. Differentiation of IgG-subclasses revealed mean ratios of IgG2b to IgG1 below 0.5 in sera of all mice prior to infection and of BALB/c mice post infection. In contrast, C57BL/6 mice had a mean IgG2b/IgG1 ratio of 2.5 post infection indicating a Th1 immune response in C57BL/6 versus a Th2 response in BALB/c mice. Evaluation of different sentinel systems revealed that cultural and serological investigations of these animals might not be sufficient to detect infection. In summary, an intranasal S. moniliformis infection model in C57BL/6 mice leading to purulent to necrotizing inflammations in the lung, the lymph nodes and other organs associated with a Th1 immune response is described.


Asunto(s)
Infecciones por Fusobacterium/inmunología , Infecciones por Fusobacterium/patología , Streptobacillus , Animales , Modelos Animales de Enfermedad , Femenino , Infecciones por Fusobacterium/microbiología , Especificidad del Huésped/inmunología , Inmunoglobulina G/sangre , Inflamación/inmunología , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Streptobacillus/fisiología , Células TH1/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA