Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 18(8): e1010745, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-36037230

RESUMEN

In vivo bioluminescence imaging facilitates the non-invasive visualization of biological processes in living animals. This system has been used to track virus infections mostly in mice and ferrets; however, until now this approach has not been applied to pathogens in avian species. To visualize the infection of an important avian pathogen, we generated Marek's disease virus (MDV) recombinants expressing firefly luciferase during lytic replication. Upon characterization of the recombinant viruses in vitro, chickens were infected and the infection visualized in live animals over the course of 14 days. The luminescence signal was consistent with the known spatiotemporal kinetics of infection and the life cycle of MDV, and correlated well with the viral load measured by qPCR. Intriguingly, this in vivo bioimaging approach revealed two novel sites of MDV replication, the beak and the skin of the feet covered in scales. Feet skin infection was confirmed using a complementary fluorescence bioimaging approach with MDV recombinants expressing mRFP or GFP. Infection was detected in the intermediate epidermal layers of the feet skin that was also shown to produce infectious virus, regardless of the animals' age at and the route of infection. Taken together, this study highlights the value of in vivo whole body bioimaging in avian species by identifying previously overlooked sites of replication and shedding of MDV in the chicken host.


Asunto(s)
Herpesviridae , Herpesvirus Gallináceo 2 , Enfermedad de Marek , Animales , Pollos , Hurones , Ratones
2.
Proc Natl Acad Sci U S A ; 117(1): 337-345, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31871151

RESUMEN

Out of the 14 avian ß-defensins identified in the Gallus gallus genome, only 3 are present in the chicken egg, including the egg-specific avian ß-defensin 11 (Gga-AvBD11). Given its specific localization and its established antibacterial activity, Gga-AvBD11 appears to play a protective role in embryonic development. Gga-AvBD11 is an atypical double-sized defensin, predicted to possess 2 motifs related to ß-defensins and 6 disulfide bridges. The 3-dimensional NMR structure of the purified Gga-AvBD11 is a compact fold composed of 2 packed ß-defensin domains. This fold is the archetype of a structural family, dubbed herein as avian-double-ß-defensins (Av-DBD). We speculate that AvBD11 emanated from a monodomain gene ancestor and that similar events might have occurred in arthropods, leading to another structural family of less compact DBDs. We show that Gga-AvBD11 displays antimicrobial activities against gram-positive and gram-negative bacterial pathogens, the avian protozoan Eimeria tenella, and avian influenza virus. Gga-AvBD11 also shows cytotoxic and antiinvasive activities, suggesting that it may not only be involved in innate protection of the chicken embryo, but also in the (re)modeling of embryonic tissues. Finally, the contribution of either of the 2 Gga-AvBD11 domains to these biological activities was assessed, using chemically synthesized peptides. Our results point to a critical importance of the cationic N-terminal domain in mediating antibacterial, antiparasitic, and antiinvasive activities, with the C-terminal domain potentiating the 2 latter activities. Strikingly, antiviral activity in infected chicken cells, accompanied by marked cytotoxicity, requires the full-length protein.


Asunto(s)
Proteínas Aviares/genética , Embrión de Pollo/inmunología , Pollos/fisiología , Desarrollo Embrionario/inmunología , beta-Defensinas/genética , Secuencia de Aminoácidos , Animales , Proteínas Aviares/ultraestructura , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/veterinaria , Bioensayo , Embrión de Pollo/crecimiento & desarrollo , Embrión de Pollo/microbiología , Embrión de Pollo/parasitología , Coccidiosis/inmunología , Coccidiosis/parasitología , Coccidiosis/veterinaria , Eimeria tenella/inmunología , Evolución Molecular , Genoma , Inmunidad Innata/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Aviar/inmunología , Gripe Aviar/virología , Resonancia Magnética Nuclear Biomolecular , Filogenia , Dominios Proteicos/genética , Dominios Proteicos/inmunología
3.
Am J Respir Cell Mol Biol ; 65(4): 378-389, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34102087

RESUMEN

Excessive lung inflammation and airway epithelial damage are hallmarks of human inflammatory lung diseases, such as cystic fibrosis (CF). Enhancement of innate immunity provides protection against pathogens while reducing lung-damaging inflammation. However, the mechanisms underlying innate immunity-mediated protection in the lung remain mysterious, in part because of the lack of appropriate animal models for these human diseases. TLR5 (Toll-like receptor 5) stimulation by its specific ligand, the bacterial protein flagellin, has been proposed to enhance protection against several respiratory infectious diseases, although other cellular events, such as calcium signaling, may also control the intensity of the innate immune response. Here, we investigated the molecular events prompted by stimulation with flagellin and its role in regulating innate immunity in the lung of the pig, which is anatomically and genetically more similar to humans than rodent models. We found that flagellin treatment modulated NF-κB signaling and intracellular calcium homeostasis in airway epithelial cells. Flagellin pretreatment reduced the NF-κB nuclear translocation and the expression of proinflammatory cytokines to a second flagellin stimulus as well as to Pseudomonas aeruginosa infection. Moreover, in vivo administration of flagellin decreased the severity of P. aeruginosa-induced pneumonia. Then we confirmed these beneficial effects of flagellin in a pathological model of CF by using ex vivo precision-cut lung slices from a CF pigz model. These results provide evidence that flagellin treatment contributes to a better regulation of the inflammatory response in inflammatory lung diseases such as CF.


Asunto(s)
Flagelina/farmacología , Inflamación/tratamiento farmacológico , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad , Animales , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Flagelina/inmunología , Flagelina/metabolismo , Inmunidad Innata/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Transducción de Señal/efectos de los fármacos , Porcinos
4.
Genet Sel Evol ; 53(1): 44, 2021 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-33957861

RESUMEN

BACKGROUND: In all organisms, life-history traits are constrained by trade-offs, which may represent physiological limitations or be related to energy resource management. To detect trade-offs within a population, one promising approach is the use of artificial selection, because intensive selection on one trait can induce unplanned changes in others. In chickens, the breeding industry has achieved remarkable genetic progress in production and feed efficiency over the last 60 years. However, this may have been accomplished at the expense of other important biological functions, such as immunity. In the present study, we used three experimental lines of layer chicken-two that have been divergently selected for feed efficiency and one that has been selected for increased antibody response to inactivated Newcastle disease virus (ND3)-to explore the impact of improved feed efficiency on animals' immunocompetence and, vice versa, the impact of improved antibody response on animals' growth and feed efficiency. RESULTS: There were detectable differences between the low (R+) and high (R-) feed-efficiency lines with respect to vaccine-specific antibody responses and counts of monocytes, heterophils, and/or T cell population. The ND3 line presented reduced body weight and feed intake compared to the control line. ND3 chickens also demonstrated an improved antibody response against a set of commercial viral vaccines, but lower blood leucocyte counts. CONCLUSIONS: This study demonstrates the value of using experimental chicken lines that are divergently selected for RFI or for a high antibody production, to investigate the modulation of immune parameters in relation to growth and feed efficiency. Our results provide further evidence that long-term selection for the improvement of one trait may have consequences on other important biological functions. Hence, strategies to ensure optimal trade-offs among competing functions will ultimately be required in multi-trait selection programs in livestock.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales/genética , Pollos/genética , Enfermedades de las Aves de Corral/genética , Selección Artificial , Animales , Peso Corporal , Pollos/crecimiento & desarrollo , Pollos/inmunología , Rasgos de la Historia de Vida , Enfermedades de las Aves de Corral/inmunología
5.
Vet Res ; 51(1): 2, 2020 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-31924278

RESUMEN

The avian respiratory tract is a common entry route for many pathogens and an important delivery route for vaccination in the poultry industry. Immune responses in the avian lung have mostly been studied in vivo due to the lack of robust, relevant in vitro and ex vivo models mimicking the microenvironment. Precision-cut lung slices (PCLS) have the major advantages of maintaining the 3-dimensional architecture of the lung and includes heterogeneous cell populations. PCLS have been obtained from a number of mammalian species and from chicken embryos. However, as the embryonic lung is physiologically undifferentiated and immunologically immature, it is less suitable to examine complex host-pathogen interactions including antimicrobial responses. Here we prepared PCLS from immunologically mature chicken lungs, tested different culture conditions, and found that serum supplementation has a detrimental effect on the quality of PCLS. Viable cells in PCLS remained present for ≥ 40 days, as determined by viability assays and sustained motility of fluorescent mononuclear phagocytic cells. The PCLS were responsive to lipopolysaccharide stimulation, which induced the release of nitric oxide, IL-1ß, type I interferons and IL-10. Mononuclear phagocytes within the tissue maintained phagocytic activity, with live cell imaging capturing interactions with latex beads and an avian pathogenic Escherichia coli strain. Finally, the PCLS were also shown to be permissive to infection with low pathogenic avian influenza viruses. Taken together, immunologically mature chicken PCLS provide a suitable model to simulate live organ responsiveness and cell dynamics, which can be readily exploited to examine host-pathogen interactions and inflammatory responses.


Asunto(s)
Pollos , Interacciones Huésped-Patógeno/inmunología , Pulmón/inmunología , Enfermedades de las Aves de Corral/inmunología , Medicina Veterinaria/métodos , Animales , Pollos/inmunología , Lipopolisacáridos/metabolismo , Pulmón/microbiología , Pulmón/parasitología , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/parasitología
6.
Avian Pathol ; 48(2): 121-134, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30556415

RESUMEN

It is well established that the endothelium plays a prominent role in the pathogenesis of various infectious diseases in mammals. However, little is known about the role of endothelial cells (EC) as targets for avian pathogens and their contribution to the pathogenesis of infectious diseases in galliform birds. First, we explored the innate immune response of primary chicken aortic endothelial cells (pchAEC), obtained from 18-day-old embryos, to stimulation with pathogen-associated molecular patterns or recombinant chicken interferons (type I, II and III IFNs). In spite of the abundant expression of a number of innate immune receptors, marked cytokine responses to stimulation with pathogen-associated molecular patterns were only seen in pchAEC treated with the TLR3 agonist polyI:C (pI:C) and the MDA5 agonist liposome-complexed polyI:C (L-pI:C), as was assessed by quantitative PCR and luciferase-based IFN-I/NFκB reporter assays. Treatments of pchAEC with IFN-α, IFN-γ and IFN-λ resulted in STAT1-phosphorylation/activation, as was revealed by immunoblotting. Next, we demonstrated that pchAEC are susceptible to infection with a variety of poultry pathogens, including Marek's disease virus (MDV), infectious bursal disease virus (IBDV), avian pathogenic Escherichia coli (APEC) and Eimeria tenella. Our data highlight that chicken EC are potential targets for viral, bacterial and protozoan pathogens in gallinaceous poultry and may partake in the inflammatory and antimicrobial response. The pchAEC infection model used herein will allow further studies interrogating avian pathogen interactions with vascular EC. RESEARCH HIGHLIGHTS Use of a well-defined primary chicken aortic endothelial cell (pchAEC) culture model for studying avian host-pathogen interactions. pchAEC are responsive to innate immune stimulation with viral pathogen-associated molecular patterns and chicken type I, II and III interferons. pchAEC are susceptible to infections with economically important poultry pathogens, including MDV, IBDV, APEC and Eimeria tenella.


Asunto(s)
Interacciones Huésped-Patógeno , Inmunidad Innata , Interferones/metabolismo , Enfermedades de las Aves de Corral/inmunología , Animales , Línea Celular Tumoral , Células Cultivadas , Embrión de Pollo , Pollos , Células Endoteliales/inmunología , Endotelio/inmunología , Femenino , Inflamación/microbiología , Inflamación/parasitología , Inflamación/veterinaria , Interferones/genética , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/parasitología
7.
BMC Microbiol ; 18(1): 138, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30340527

RESUMEN

BACKGROUND: Endothelial cells play a major role in highly pathogenic avian influenza (HPAI) virus pathogenesis in gallinaceous poultry species (e.g. chicken, turkey and quail). Upon infection of gallinaceous poultry with HPAI viruses, endothelial cells throughout the body become rapidly infected, leading to systemic dissemination of the virus, disseminated intravascular coagulation, oedema and haemorrhaging. In contrast, the pathogenesis of HPAI viruses in most wild bird species (e.g. duck, goose and gull species) is not associated with endothelial tropism. Indeed, viral antigen is not found in the endothelial cells of most wild bird species following infection with HPAI viruses. This differential endothelial cell tropism in avian species is poorly understood, mainly due to the absence of appropriate cell culture systems. RESULTS: Here, we describe the isolation and purification of primary duck endothelial cells from the aorta or bone marrow of Pekin duck embryos. Cells were differentiated in the presence of vascular endothelial growth factor and, if needed, enriched via fluorescent-activated cell sorting based on the uptake of acetylated low-density lipoprotein. The expression of von Willebrand factor, a key marker of endothelial cells, was confirmed by polymerase chain reaction. Monocultures of duck endothelial cells, either derived from the aorta or the bone marrow, were susceptible to infection with an H5N1 HPAI virus but to a much lesser extent than chicken endothelial cells. CONCLUSIONS: The methods described herein to isolate and purify duck endothelial cells from the aorta or bone marrow could also be applied to obtain microvascular endothelial cells from other tissues and organs, such as the lung or the intestine, and represent a valuable tool to study the pathogenesis of avian viruses.


Asunto(s)
Técnicas de Cultivo de Célula , Células Endoteliales/virología , Subtipo H5N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Animales , Antígenos Virales , Aorta/citología , Aorta/virología , Células de la Médula Ósea/virología , Células Cultivadas , Patos/virología , Citometría de Flujo , Gripe Aviar/virología , Enfermedades de las Aves de Corral/virología , Factor A de Crecimiento Endotelial Vascular/farmacología
8.
Virol J ; 15(1): 55, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29587792

RESUMEN

BACKGROUND: Non-structural protein NS1 of influenza A viruses harbours several determinants of pathogenicity and host-range. However it is still unclear to what extent each of its two structured domains (i.e. RNA-binding domain, RBD, and effector domain, ED) contribute to its various activities. METHODS: To evaluate the respective contributions of the two domains, we genetically engineered two variants of an H7N1 low pathogenicity avian influenza virus harbouring amino-acid substitutions that impair the functionality of either domain. The RBD- and ED-mutant viruses were compared to their wt- counterpart in vivo and in vitro, notably in chicken infection and avian cell culture models. RESULTS: The double substitution R38A-K41A in the RBD dramatically reduced the pathogenicity and replication potential of the virus, whereas the substitution A149V that was considered to abrogate the IFN-antagonistic activity of the effector domain entailed much less effects. While all three viruses initiated the viral life cycle in avian cells, replication of the R38A-K41A virus was severely impaired. This defect was associated with a delayed synthesis of nucleoprotein NP and a reduced accumulation of NS1, which was found to reach a concentration of about 30 micromol.L- 1 in wt-infected cells at 8 h post-infection. When overexpressed in avian lung epithelial cells, both the wt-NS1 and 3841AA-NS1, but not the A149V-NS1, reduced the poly(I:C)-induced activation of the IFN-sensitive chicken Mx promoter. Unexpectedly, the R38A-K41A substitution in the recombinant RBD did not alter its in vitro affinity for a model dsRNA. When overexpressed in avian cells, both the wt- and A149V-NS1s, as well as the individually expressed wt-RBD to a lesser extent, enhanced the activity of the reconstituted viral RNA-polymerase in a minireplicon assay. CONCLUSIONS: Collectively, our data emphasized the critical importance and essential role of the RNA-binding domain in essential steps of the virus replication cycle, notably expression and translation of viral mRNAs.


Asunto(s)
Subtipo H7N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H7N1 del Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Motivos de Unión al ARN/fisiología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/fisiología , Sustitución de Aminoácidos , Animales , Línea Celular , Embrión de Pollo , Pollos , Modelos Animales de Enfermedad , Perros , Expresión Génica , Regulación Viral de la Expresión Génica , Subtipo H7N1 del Virus de la Influenza A/genética , Células de Riñón Canino Madin Darby , Motivos de Unión al ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas Virales/biosíntesis , Virulencia/genética
9.
Arch Virol ; 160(1): 161-71, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25298182

RESUMEN

Gallid herpesvirus 2 (GaHV-2) is the alphaherpesvirus responsible for Marek's disease (MD), a T-cell lymphoma of chickens. The virulence of the GaHV-2 field strain is steadily increasing, but MD is still controlled by the CVI988/Rispens vaccine. We tried to determine distinguishing traits of the CVI988/Rispens vaccine by focusing on the 5' end region of the latency-associated transcript (5'LAT). It includes a variable number of 60-bp tandem repeats depending on the GaHV-2 strain. By analyzing six batches of vaccine, we showed that CVI988/Rispens consisted of a population of 5'LAT molecular subtypes, all with deletions and lacking 60-bp tandem repeat motifs, with two major subtypes that probably constitute CVI988/Rispens markers. Serial passages in cell culture led to a substantial change in the frequency of CVI988/Rispens 5'LAT subtypes, with non-deleted subtypes harboring up to four 60-bp repeats emerging during the last few passages. Dynamic changes in the distribution of 5'LAT-deleted subtypes were also detected after infection of chickens. By contrast, the 5'LAT region of the oncogenic clonal RB-1B strain, which was investigated at every step from the isolation of the clonal bacmid RB-1B DNA to the isolation of the ovarian lymphoma cell line, consisted of non-deleted 5'LAT subtypes harboring at least two 60-bp repeats. Thus, vaccine and oncogenic GaHV-2 strains consist of specific populations of viral genomes that are constantly evolving in vivo and in vitro and providing potential markers for epidemiological surveys.


Asunto(s)
Evolución Molecular , Regulación Viral de la Expresión Génica/fisiología , Variación Genética , Herpesvirus Gallináceo 2/clasificación , Enfermedad de Marek/virología , Proteínas Virales/metabolismo , Vacunas Virales/inmunología , Animales , Pollos , Herpesvirus Gallináceo 2/genética , Enfermedad de Marek/prevención & control , Organismos Libres de Patógenos Específicos , Proteínas Virales/genética
10.
J Gen Virol ; 95(Pt 6): 1233-1243, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24694396

RESUMEN

Currently circulating H5N1 influenza viruses have undergone a complex evolution since the appearance of their progenitor A/Goose/Guangdong/1/96 in 1996. After the eradication of the H5N1 viruses that emerged in Hong Kong in 1997 (HK/97 viruses), new genotypes of H5N1 viruses emerged in the same region in 2000 that were more pathogenic for both chickens and mice than HK/97 viruses. These, as well as virtually all highly pathogenic H5N1 viruses since 2000, harbour a deletion of aa 80-84 in the unstructured region of the non-structural (NS) protein NS1 linking its RNA-binding domain to its effector domain. NS segments harbouring this mutation have since been found in non-H5N1 viruses and we asked whether this 5 aa deletion could have a general effect not limited to the NS1 of H5N1 viruses. We genetically engineered this deletion in the NS segment of a duck-origin avian H1N1 virus, and compared the in vivo and in vitro properties of the WT and NSdel8084 viruses. In experimentally infected chickens, the NSdel8084 virus showed both an increased replication potential and an increased pathogenicity. This in vivo phenotype was correlated with a higher replicative efficiency in vitro, both in embryonated eggs and in a chicken lung epithelial cell line. Our data demonstrated that the increased replicative potential conferred by this small deletion was a general feature not restricted to NS1 from H5N1 viruses and suggested that viruses acquiring this mutation may be selected positively in the future.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Embrión de Pollo , Pollos , Citocinas/genética , ADN Viral/genética , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/química , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/fisiología , Gripe Aviar/inmunología , Gripe Aviar/patología , Gripe Aviar/virología , Interferón Tipo I/biosíntesis , Pulmón/patología , Pulmón/virología , Ratones , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Eliminación de Secuencia , Especificidad de la Especie , Carga Viral , Proteínas no Estructurales Virales/genética , Virulencia/genética , Virulencia/fisiología , Replicación Viral/genética , Replicación Viral/fisiología
11.
Dev Comp Immunol ; 151: 105093, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37951324

RESUMEN

The innate immune response relies on the ability of host cells to rapidly detect and respond to microbial nucleic acids. Toll-like receptors (TLRs), a class of pattern recognition receptors (PRRs), play a fundamental role in distinguishing self from non-self at the molecular level. In this study, we focused on TLR21, an avian TLR that recognizes DNA motifs commonly found in bacterial genomic DNA, specifically unmethylated CpG motifs. TLR21 is believed to act as a functional homologue to mammalian TLR9. By analysing TLR21 signalling in chickens, we sought to elucidate avian TLR21 activation outputs in parallel to that of other nucleic acid species. Our analyses revealed that chicken TLR21 (chTLR21) triggers the activation of NF-κB and induces a potent type-I interferon response in chicken macrophages, similar to the signalling cascades observed in mammalian TLR9 activation. Notably, the transcription of interferon beta (IFNB) by chTLR21 was found to be dependent on both NF-κB and IRF7 signalling, but independent of the TBK1 kinase, a distinctive feature of mammalian TLR9 signalling. These findings highlight the conservation of critical signalling components and downstream responses between avian TLR21 and mammalian TLR9, despite their divergent evolutionary origins. These insights into the evolutionarily conserved mechanisms of nucleic acid sensing contribute to the broader understanding of host-pathogen interactions across species.


Asunto(s)
Interferón Tipo I , Ácidos Nucleicos , Animales , Pollos , Receptor Toll-Like 9 , FN-kappa B , Oligodesoxirribonucleótidos , Mamíferos
12.
PLoS Pathog ; 7(10): e1002333, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22046133

RESUMEN

Telomerase reverse transcriptase (TERT) and telomerase RNA (TR) represent the enzymatically active components of telomerase. In the complex, TR provides the template for the addition of telomeric repeats to telomeres, a protective structure at the end of linear chromosomes. Human TR with a mutation in the template region has been previously shown to inhibit proliferation of cancer cells in vitro. In this report, we examined the effects of a mutation in the template of a virus encoded TR (vTR) on herpesvirus-induced tumorigenesis in vivo. For this purpose, we used the oncogenic avian herpesvirus Marek's disease virus (MDV) as a natural virus-host model for lymphomagenesis. We generated recombinant MDV in which the vTR template sequence was mutated from AATCCCAATC to ATATATATAT (vAU5) by two-step Red-mediated mutagenesis. Recombinant viruses harboring the template mutation replicated with kinetics comparable to parental and revertant viruses in vitro. However, mutation of the vTR template sequence completely abrogated virus-induced tumor formation in vivo, although the virus was able to undergo low-level lytic replication. To confirm that the absence of tumors was dependent on the presence of mutant vTR in the telomerase complex, a second mutation was introduced in vAU5 that targeted the P6.1 stem loop, a conserved region essential for vTR-TERT interaction. Absence of vTR-AU5 from the telomerase complex restored virus-induced lymphoma formation. To test if the attenuated vAU5 could be used as an effective vaccine against MDV, we performed vaccination-challenge studies and determined that vaccination with vAU5 completely protected chickens from lethal challenge with highly virulent MDV. Taken together, our results demonstrate 1) that mutation of the vTR template sequence can completely abrogate virus-induced tumorigenesis, likely by the inhibition of cancer cell proliferation, and 2) that this strategy could be used to generate novel vaccine candidates against virus-induced lymphoma.


Asunto(s)
Herpesvirus Gallináceo 2/patogenicidad , Linfoma de Células T/veterinaria , Enfermedad de Marek/virología , Mutación , ARN/genética , Telomerasa/genética , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Pollos , Regulación Leucémica de la Expresión Génica , Regulación Viral de la Expresión Génica , Herpesvirus Gallináceo 2/enzimología , Herpesvirus Gallináceo 2/genética , Interacciones Huésped-Patógeno , Linfoma de Células T/genética , Linfoma de Células T/virología , Enfermedad de Marek/genética , ARN Viral/análisis , Moldes Genéticos , Vacunación/veterinaria
13.
J Exp Med ; 203(5): 1307-17, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16651385

RESUMEN

Telomerase is a ribonucleoprotein complex consisting of two essential core components: a reverse transcriptase and an RNA subunit (telomerase RNA [TR]). Dysregulation of telomerase has been associated with cell immortalization and oncogenesis. Marek's disease herpesvirus (MDV) induces a malignant T cell lymphoma in chickens and harbors in its genome two identical copies of a viral TR (vTR) with 88% sequence identity to chicken TR. MDV mutants lacking both copies of vTR were significantly impaired in their ability to induce T cell lymphomas, although lytic replication in vivo was unaffected. Tumor incidences were reduced by >60% in chickens infected with vTR- viruses compared with animals inoculated with MDV harboring at least one intact copy of vTR. Lymphomas in animals infected with the vTR- viruses were also significantly smaller in size and less disseminated. Constitutive expression of vTR in the chicken fibroblast cell line DF-1 resulted in a phenotype consistent with transformation as indicated by morphological alteration, enhanced anchorage-independent cell growth, cell growth beyond saturation density, and increased expression levels of integrin alpha v. We concluded that vTR plays a critical role in MDV-induced T cell lymphomagenesis. Furthermore, our results provide the first description of tumor-promoting effects of TR in a natural virus-host infection model.


Asunto(s)
Transformación Celular Viral , Linfoma de Células T/enzimología , Mardivirus/enzimología , Telomerasa/metabolismo , Proteínas Virales/metabolismo , Animales , Línea Celular , Pollos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/virología , Regulación Leucémica de la Expresión Génica/fisiología , Regulación Viral de la Expresión Génica/fisiología , Genoma , Cadenas alfa de Integrinas/biosíntesis , Cadenas beta de Integrinas/biosíntesis , Linfoma de Células T/genética , Linfoma de Células T/patología , Linfoma de Células T/virología , Mardivirus/genética , Enfermedad de Marek/enzimología , Enfermedad de Marek/genética , Enfermedad de Marek/patología , Enfermedad de Marek/virología , Mutación , Homología de Secuencia de Aminoácido , Telomerasa/genética , Proteínas Virales/genética
14.
PLoS Pathog ; 6(8): e1001073, 2010 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-20865127

RESUMEN

Telomerase is a ribonucleoprotein complex involved in the maintenance of telomeres, a protective structure at the distal ends of chromosomes. The enzyme complex contains two main components, telomerase reverse transcriptase (TERT), the catalytic subunit, and telomerase RNA (TR), which serves as a template for the addition of telomeric repeats (TTAGGG)(n). Marek's disease virus (MDV), an oncogenic herpesvirus inducing fatal lymphoma in chickens, encodes a TR homologue, viral TR (vTR), which significantly contributes to MDV-induced lymphomagenesis. As recent studies have suggested that TRs possess functions independently of telomerase activity, we investigated if the tumor-promoting properties of MDV vTR are dependent on formation of a functional telomerase complex. The P6.1 stem-loop of TR is known to mediate TR-TERT complex formation and we show here that interaction of vTR with TERT and, consequently, telomerase activity was efficiently abrogated by the disruption of the vTR P6.1 stem-loop (P6.1mut). Recombinant MDV carrying the P6.1mut stem-loop mutation were generated and tested for their behavior in the natural host in vivo. In contrast to viruses lacking vTR, all animals infected with the P6.1mut viruses developed MDV-induced lymphomas, but onset of tumor formation was significantly delayed. P6.1mut viruses induced enhanced metastasis, indicating functionality of non-complexed vTR in tumor dissemination. We discovered that RPL22, a cellular factor involved in T-cell development and virus-induced transformation, directly interacts with wild-type and mutant vTR and is, consequently, relocalized to the nucleoplasm. Our study provides the first evidence that expression of TR, in this case encoded by a herpesvirus, is pro-oncogenic in the absence of telomerase activity.


Asunto(s)
Herpesvirus Gallináceo 2/enzimología , Enfermedad de Marek/enzimología , ARN Viral/metabolismo , ARN/metabolismo , Telomerasa/metabolismo , Animales , Dominio Catalítico/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Pollos , Cromosomas Artificiales Bacterianos , Herpesvirus Gallináceo 2/genética , Enfermedad de Marek/genética , Enfermedad de Marek/patología , Mutación , ARN Viral/efectos adversos , ARN Viral/genética , Telomerasa/genética
15.
Front Immunol ; 13: 956670, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36268022

RESUMEN

Research in mammals has evidenced that proper colonization of the gut by a complex commensal microbial community, the gut microbiota (GM), is critical for animal health and wellbeing. It greatly contributes to the control of infectious processes through competition in the microbial environment while supporting proper immune system development and modulating defence mechanisms at distant organ sites such as the lung: a concept named 'gut-lung axis'. While recent studies point to a role of the GM in boosting immunity and pathogen resilience also in poultry, the mechanisms underlying this role are largely unknown. In spite of this knowledge gap, GM modulation approaches are today considered as one of the most promising strategies to improve animal health and welfare in commercial poultry production, while coping with the societal demand for responsible, sustainable and profitable farming systems. The majority of pathogens causing economically important infectious diseases in poultry are targeting the respiratory and/or gastrointestinal tract. Therefore, a better understanding of the role of the GM in the development and function of the mucosal immune system is crucial for implementing measures to promote animal robustness in commercial poultry production. The importance of early gut colonization in the chicken has been overlooked or neglected in industrial poultry production systems, where chicks are hampered from acquiring a complex GM from the hen. Here we discuss the concept of strengthening mucosal immunity in the chicken through GM modulation approaches favouring immune system development and functioning along the gut-lung axis, which could be put into practice through improved farming systems, early-life GM transfer, feeding strategies and pre-/probiotics. We also provide original data from experiments with germ-free and conventional chickens demonstrating that the gut-lung axis appears to be functional in chickens. These key principles of mucosal immunity are likely to be relevant for a variety of avian diseases and are thus of far-reaching importance for the poultry sector worldwide.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades de las Aves de Corral , Animales , Femenino , Pollos , Inmunidad Innata , Aves de Corral , Pulmón , Mamíferos
16.
Front Microbiol ; 13: 958307, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35875580

RESUMEN

The continuous spread of African swine fever virus (ASFV) in Europe and Asia represents a major threat to livestock health, with billions of dollars of income losses and major perturbations of the global pig industry. One striking feature of African swine fever (ASF) is the existence of different forms of the disease, ranging from acute with mortality rates approaching 100% to chronic, with mild clinical manifestations. These differences in pathogenicity have been linked to genomic alterations present in attenuated ASFV strains (and absent in virulent ones) and differences in the immune response of infected animals. In this mini-review, we summarized current knowledge on the connection between ASFV pathogenicity and the innate immune response induced in infected hosts, with a particular focus on the pathways involved in ASFV detection. Indeed, recent studies have highlighted the key role of the DNA sensor cGAS in ASFV sensing. We discussed what other pathways may be involved in ASFV sensing and inflammasome activation and summarized recent findings on the viral ASFV genes involved in the modulation of the interferon (IFN) and nuclear factor kappa B (NF-κB) pathways.

17.
J Neuroinflammation ; 7: 7, 2010 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-20109187

RESUMEN

BACKGROUND: Sudden limb paresis is a common problem in White Leghorn flocks, affecting about 1% of the chicken population before achievement of sexual maturity. Previously, a similar clinical syndrome has been reported as being caused by inflammatory demyelination of peripheral nerve fibres. Here, we investigated in detail the immunopathology of this paretic syndrome and its possible resemblance to human neuropathies. METHODS: Neurologically affected chickens and control animals from one single flock underwent clinical and neuropathological examination. Peripheral nervous system (PNS) alterations were characterised using standard morphological techniques, including nerve fibre teasing and transmission electron microscopy. Infiltrating cells were phenotyped immunohistologically and quantified by flow cytometry. The cytokine expression pattern was assessed by quantitative real-time PCR (qRT-PCR). These investigations were accomplished by MHC genotyping and a PCR screen for Marek's disease virus (MDV). RESULTS: Spontaneous paresis of White Leghorns is caused by cell-mediated, inflammatory demyelination affecting multiple cranial and spinal nerves and nerve roots with a proximodistal tapering. Clinical manifestation coincides with the employment of humoral immune mechanisms, enrolling plasma cell recruitment, deposition of myelin-bound IgG and antibody-dependent macrophageal myelin-stripping. Disease development was significantly linked to a 539 bp microsatellite in MHC locus LEI0258. An aetiological role for MDV was excluded. CONCLUSIONS: The paretic phase of avian inflammatory demyelinating polyradiculoneuritis immunobiologically resembles the late-acute disease stages of human acute inflammatory demyelinating polyneuropathy, and is characterised by a Th1-to-Th2 shift.


Asunto(s)
Modelos Animales de Enfermedad , Síndrome de Guillain-Barré/patología , Síndrome de Guillain-Barré/fisiopatología , Síndrome de Guillain-Barré/veterinaria , Animales , Antígenos CD/metabolismo , Pollos , Femenino , Citometría de Flujo , Ganglios Espinales/patología , Ganglios Espinales/ultraestructura , Genotipo , Síndrome de Guillain-Barré/virología , Humanos , Modelos Logísticos , Complejo Mayor de Histocompatibilidad/genética , Microscopía Electrónica de Transmisión/métodos , Neurosífilis , Lectinas de Plantas , Médula Espinal/patología , Médula Espinal/ultraestructura , Nervios Espinales/patología , Nervios Espinales/ultraestructura , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
18.
Arch Virol ; 155(11): 1823-37, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20680360

RESUMEN

Mdv1-miR-M4 is one of 25 microRNAs (miRNAs) expressed by Marek's disease virus (MDV-1), an oncogenic alphaherpesvirus that induces fatal T-cell lymphoma in chickens. Mdv1-miR-M4 was shown to be the second functional viral ortholog of miR-155, a cellular miRNA that plays a crucial role in several physiological and pathological processes in lymphocyte biology. In this study, we investigated a panel of putative mdv1-miR-M4 targets involved in gene networks affecting both cellular and viral life cycles. Using luciferase reporter assays, we showed that mdv1-miR-M4-5P and miR-155 efficiently targeted a common set of 3' untranslated regions (3'UTR) of six cellular genes (GPM6B, RREB1, c-Myb, MAP3K7IP2, PU.1 and C/EBP). In addition, we also investigated the interactions between mdv1-miR-M4-5P and mdv1-miR-M43P and viral mRNAs encoding UL28 and UL32 in both reporter and western blot assays. Mdv1-miR-M4 specifically inhibited the translation of these two viral proteins, which are involved in the cleavage/packaging of herpesvirus DNA.


Asunto(s)
Fibroblastos/metabolismo , Regulación de la Expresión Génica/fisiología , Mardivirus/genética , Mardivirus/metabolismo , MicroARNs/metabolismo , Animales , Secuencia de Bases , Línea Celular , Pollos , Leucocitos/metabolismo , MicroARNs/genética
19.
Viruses ; 12(9)2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32867106

RESUMEN

The non-structural protein NS1 of influenza A viruses is an RNA-binding protein of which its activities in the infected cell contribute to the success of the viral cycle, notably through interferon antagonism. We have previously shown that NS1 strongly binds RNA aptamers harbouring virus-specific sequence motifs (Marc et al., Nucleic Acids Res. 41, 434-449). Here, we started out investigating the putative role of one particular virus-specific motif through the phenotypic characterization of mutant viruses that were genetically engineered from the parental strain WSN. Unexpectedly, our data did not evidence biological importance of the putative binding of NS1 to this specific motif (UGAUUGAAG) in the 3'-untranslated region of its own mRNA. Next, we sought to identify specificity determinants in the NS1-RNA interaction through interaction assays in vitro with several RNA ligands and through solving by X-ray diffraction the 3D structure of several complexes associating NS1's RBD with RNAs of various affinities. Our data show that the RBD binds the GUAAC motif within double-stranded RNA helices with an apparent specificity that may rely on the sequence-encoded ability of the RNA to bend its axis. On the other hand, we showed that the RBD binds to the virus-specific AGCAAAAG motif when it is exposed in the apical loop of a high-affinity RNA aptamer, probably through a distinct mode of interaction that still requires structural characterization. Our data are consistent with more than one mode of interaction of NS1's RBD with RNAs, recognizing both structure and sequence determinants.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/química , Subtipo H7N1 del Virus de la Influenza A/química , ARN Viral/química , ARN/química , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Regiones no Traducidas 3' , Animales , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/metabolismo , Secuencia de Bases , Línea Celular , Humanos , Modelos Moleculares , Conformación de Ácido Nucleico , Unión Proteica , Dominios Proteicos , ARN/metabolismo , ARN Bicatenario/química , ARN Bicatenario/metabolismo , ARN Mensajero/química , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Técnica SELEX de Producción de Aptámeros
20.
Nat Rev Microbiol ; 4(4): 283-94, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16541136

RESUMEN

Marek's disease virus (MDV) is an oncogenic herpesvirus that causes various clinical syndromes in its natural host, the chicken. MDV has long been of interest as a model organism, particularly with respect to the pathogenesis and immune control of virus-induced lymphoma in an easily accessible small-animal system. Recent advances in MDV genetics and the determination of the chicken genome sequence, aided by functional genomics, have begun to dramatically increase our understanding not only of lytic MDV replication, but also of the factors and mechanisms leading to latency and tumour formation. This new information is helping to elucidate cellular signalling pathways that have undergone convergent evolution and are perturbed by different viruses, and emphasizes the value of MDV as a comparative biomedical model. Furthermore, the door is now open for rational and efficient engineering of new vaccines against one of the most important and widespread infectious diseases in chickens.


Asunto(s)
Mardivirus/patogenicidad , Enfermedad de Marek/fisiopatología , Enfermedades de las Aves de Corral/virología , Animales , Modelos Animales de Enfermedad , Mardivirus/genética , Enfermedad de Marek/epidemiología , Enfermedad de Marek/prevención & control , Aves de Corral , Enfermedades de las Aves de Corral/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA