Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Vet Res ; 55(1): 46, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38589976

RESUMEN

Pasteurella multocida is an important zoonotic respiratory pathogen capable of infecting a diverse range of hosts, including humans, farm animals, and wild animals. However, the precise mechanisms by which P. multocida compromises the pulmonary integrity of mammals and subsequently induces systemic infection remain largely unexplored. In this study, based on mouse and rabbit models, we found that P. multocida causes not only lung damage but also bacteremia due to the loss of lung integrity. Furthermore, we demonstrated that bacteremia is an important aspect of P. multocida pathogenesis, as evidenced by the observed multiorgan damage and systemic inflammation, and ultimately found that this systemic infection leads to a cytokine storm that can be mitigated by IL-6-neutralizing antibodies. As a result, we divided the pathogenesis of P. multocida into two phases: the pulmonary infection phase and the systemic infection phase. Based on unbiased RNA-seq data, we discovered that P. multocida-induced apoptosis leads to the loss of pulmonary epithelial integrity. These findings have been validated in both TC-1 murine lung epithelial cells and the lungs of model mice. Conversely, the administration of Ac-DEVD-CHO, an apoptosis inhibitor, effectively restored pulmonary epithelial integrity, significantly mitigated lung damage, inhibited bacteremia, attenuated the cytokine storm, and reduced mortality in mouse models. At the molecular level, we demonstrated that the FAK-AKT-FOXO1 axis is involved in P. multocida-induced lung epithelial cell apoptosis in both cells and animals. Thus, our research provides crucial information with regard to the pathogenesis of P. multocida as well as potential treatment options for this and other respiratory bacterial diseases.


Asunto(s)
Bacteriemia , Infecciones por Pasteurella , Pasteurella multocida , Enfermedades de los Roedores , Humanos , Animales , Conejos , Ratones , Infecciones por Pasteurella/veterinaria , Infecciones por Pasteurella/microbiología , Proteínas Proto-Oncogénicas c-akt , Síndrome de Liberación de Citoquinas/patología , Síndrome de Liberación de Citoquinas/veterinaria , Pulmón/patología , Bacteriemia/veterinaria , Bacteriemia/patología , Apoptosis , Mamíferos , Proteína Forkhead Box O1
2.
Vet Res ; 55(1): 31, 2024 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-38493147

RESUMEN

Pasteurella multocida is an opportunistic zoonotic pathogen that primarily causes fatal respiratory diseases, such as pneumonia and respiratory syndromes. However, the precise mechanistic understanding of how P. multocida disrupts the epithelial barrier in mammalian lung remains largely unknown. In this study, using unbiased RNA-seq analysis, we found that the evolutionarily conserved Hippo-Yap pathway was dysregulated after P. multocida infection. Given the complexity of P. multocida infection associated with lung injury and systemic inflammatory processes, we employed a combination of cell culture models, mouse models, and rabbit models to investigate the dynamics of the Hippo-Yap pathway during P. multocida infection. Our findings reveal that P. multocida infection activates the Hippo-Yap pathway both in vitro and in vivo, by upregulating the upstream factors p-Mst1/2, p-Lats1, and p-Yap, and downregulating the downstream effectors Birc5, Cyr61, and Slug. Conversely, pharmacological inhibition of the Hippo pathway by XMU-MP-1 significantly rescued pulmonary epithelial cell apoptosis in vitro and reduced lung injury, systemic inflammation, and mouse mortality in vivo. Mechanistic studies revealed that P. multocida induced up-regulation of Rassf1 expression, and Rassf1 enhanced Hippo-Yap pathway through phosphorylation. Accordingly, in vitro knockdown of Rassf1 significantly enhanced Yap activity and expression of Yap downstream factors and reduced apoptosis during P. multocida infection. P. multocida-infected rabbit samples also showed overexpression of Rassf1, p-Lats1, and p-Yap, suggesting that P. multocida activates the Rassf1-Hippo-Yap pathway. These results elucidate the pathogenic role of the Rassf1-Hippo-Yap pathway in P. multocida infection and suggest that this pathway has the potential to be a drug target for the treatment of pasteurellosis.


Asunto(s)
Lesión Pulmonar , Pasteurella multocida , Enfermedades de los Roedores , Ratones , Animales , Conejos , Vía de Señalización Hippo , Transducción de Señal , Lesión Pulmonar/veterinaria , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Pulmón/metabolismo , Apoptosis , Proliferación Celular , Mamíferos
3.
BMC Vet Res ; 20(1): 94, 2024 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-38461234

RESUMEN

Pasteurella multocida type A (PmA) mainly causes respiratory diseases such as pneumonia in bovines, leading to great economic losses to the breeding industry. At present, there is still no effective commercial vaccine against PmA infection. In this study, a mutant strain (PmCQ2Δ4555-4580) with brand-new phenotypes was obtained after serially passaging at 42 °C. Whole genome resequencing and PCR analysis showed that PmCQ2Δ4555-4580 missed six genes, including PmCQ2_004555, PmCQ2_004560, PmCQ2_004565, PmCQ2_004570, PmCQ2_004575, and PmCQ2_004580. Importantly, the virulence of PmCQ2Δ4555-4580 was reduced by approximately 2.8 × 109 times in mice. Notably, live PmCQ2Δ4555-4580 could provide 100%, 100% and 40% protection against PmA, PmB and PmF, respectively; and inactivated PmCQ2Δ4555-4580 could provide 100% and 87.5% protection against PmA and PmB. Interestingly, immune protection-related proteins were significantly upregulated in PmCQ2Δ4555-4580 based on RNA-seq and bioinformatics analysis. Meaningfully, by in vitro expression, purification and in vivo immunization, 12 proteins had different degrees of immune protective effects. Among them, PmCQ2_008205, PmCQ2_010435, PmCQ2_008190, and PmCQ2_004170 had the best protective effect, the protection rates against PmA were 50%, 40%, 30%, and 30%, respectively, and the protective rates against PmB were 62.5%, 42.9%, 37.5%, and 28.6%, respectively. Collectively, PmCQ2Δ4555-4580 is a potential vaccine candidate for the prevention of Pasteurellosis involving in high expression of immune protective related proteins.


Asunto(s)
Enfermedades de los Bovinos , Infecciones por Pasteurella , Pasteurella multocida , Enfermedades de los Roedores , Animales , Ratones , Bovinos , Pasteurella multocida/genética , Vacunas Atenuadas , Infecciones por Pasteurella/prevención & control , Infecciones por Pasteurella/veterinaria , Inmunización/veterinaria , Vacunación/veterinaria , Vacunas Bacterianas
4.
Vet Res ; 54(1): 73, 2023 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-37684678

RESUMEN

Pasteurella multocida is a gram-negative bacterium that causes serious diseases in a wide range of animal species. Inflammasomes are intracellular multimolecular protein complexes that play a critical role in host defence against microbial infection. Our previous study showed that bovine P. multocida type A (PmCQ2) infection induces NLRP3 inflammasome activation. However, the exact mechanism underlying PmCQ2-induced NLRP3 inflammasome activation is not clear. Here, we show that NLRP3 inflammasome activation is positively regulated by a scaffold protein called receptor for activated C kinase 1 (RACK1). This study shows that RACK1 expression was downregulated by PmCQ2 infection in primary mouse peritoneal macrophages and mouse tissues, and overexpression of RACK1 prevented PmCQ2-induced cell death and reduced the numbers of adherent and invasive PmCQ2, indicating a modulatory role of RACK1 in the cell death that is induced by P. multocida infection. Next, RACK1 knockdown by siRNA significantly attenuated PmCQ2-induced NLRP3 inflammasome activation, which was accompanied by a reduction in the protein expression of interleukin (IL)-1ß, pro-IL-1ß, caspase-1 and NLRP3 as well as the formation of ASC specks, while RACK1 overexpression by pcDNA3.1-RACK1 plasmid transfection significantly promoted PmCQ2-induced NLRP3 inflammasome activation; these results showed that RACK1 is essential for NLRP3 inflammasome activation. Furthermore, RACK1 knockdown decreased PmCQ2-induced NF-κB activation, but RACK1 overexpression had the opposite effect. In addition, the immunofluorescence staining and immunoprecipitation results showed that RACK1 colocalized with NLRP3 and that NEK7 and interacted with these proteins. However, inhibition of potassium efflux significantly attenuated the RACK1-NLRP3-NEK7 interaction. Our study demonstrated that RACK1 plays an important role in promoting NLRP3 inflammasome activation by regulating NF-κB and promoting NLRP3 inflammasome assembly.


Asunto(s)
Enfermedades de los Bovinos , Infecciones por Pasteurella , Pasteurella multocida , Animales , Bovinos , Ratones , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , FN-kappa B , Infecciones por Pasteurella/veterinaria , Receptores de Cinasa C Activada
5.
Vet Res ; 53(1): 81, 2022 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-36224650

RESUMEN

Pasteurella multocida (P. multocida) can cause severe respiratory disease in cattle, resulting in high mortality and morbidity. Inflammasomes are multiprotein complexes in the cytoplasm that recognize pathogens and play an important role in the host defense against microbial infection. In this study, the mechanism of P. multocida-induced NLRP6 inflammasome activation was investigated in vitro and in vivo. Firstly, P. multocida induced severe inflammation with a large number of inflammatory cells infiltrating the lungs of WT and Nlrp6-/- mice. Nlrp6-/- mice were more susceptible to P. multocida infection and they had more bacterial burden in the lungs. Then, the recruitment of macrophages and neutrophils in the lungs was investigated and the results show that the number of immune cells was significantly decreased in Nlrp6-/- mice. Subsequently, NLRP6 was shown to regulate P. multocida-induced inflammatory cytokine secretion including IL-1ß and IL-6 both in vivo and in vitro while TNF-α secretion was not altered. Moreover, NLRP6 was found to mediate caspase-1 activation and ASC oligomerization, resulting in IL-1ß secretion. Furthermore, NLRP6 inflammasome mediated the gene expression of chemokines including CXCL1, CXCL2 and CXCR2 which drive the activation of NLRP3 inflammasomes. Finally, NLRP3 protein expression was detected to be abrogated in P. multocida-infected Nlrp6-/- macrophages, indicating the synergic effect of NLRP6 and NLRP3. Our study demonstrates that NLRP6 inflammasome plays an important role in the host against P. multocida infection and contributes to the development of immune therapeutics against P. multocida.


Asunto(s)
Inflamasomas , Pasteurella multocida , Receptores de Superficie Celular/metabolismo , Animales , Caspasa 1 , Caspasas , Interleucina-1beta/metabolismo , Interleucina-6 , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Factor de Necrosis Tumoral alfa
6.
Microb Pathog ; 159: 105145, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34411653

RESUMEN

Pasteurella multocida (P. multocida) is a Gram-negative bacterium which causes diseases in poultry, livestock, and humans, resulting in huge economic losses. P. multocida serovar A CQ6 (PmCQ6) is a naturally occurring attenuated strain with a thin capsule. Thus, we aimed to explore why this strain is less virulent and produces less capsule compared with P. multocida serovar A strain CQ2 (PmCQ2). Analysis of capsular polysaccharide synthesis genes in PmCQ6 revealed that, compared with PmCQ2, there was only a single point mutation in the initiation codon sequence of the hyaC gene. To test whether this point mutation caused capsular deficiency and reduced virulence, we rescued this hyaC mutation and observed a restoration of capsule production and higher virulence. Transcriptome analysis showed that the hyaC point mutation led to a downregulation of capsule synthesis and/or iron utilization related-genes. Taken together, the results indicate that the start codon mutation of hyaC is an important factor affecting the capsule synthesis and virulence of PmCQ6.


Asunto(s)
Infecciones por Pasteurella , Pasteurella multocida , Uridina Difosfato Glucosa Deshidrogenasa/genética , Humanos , Infecciones por Pasteurella/veterinaria , Pasteurella multocida/enzimología , Pasteurella multocida/genética , Mutación Puntual , Serogrupo , Virulencia/genética
7.
Vet Res ; 52(1): 140, 2021 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-34801081

RESUMEN

QseC, a histidine sensor kinase of the QseBC two-component system, acts as a global regulator of bacterial stress resistance, biofilm formation, and virulence. The function of QseC in some bacteria is well understood, but not in Pasteurella multocida. We found that deleting qseC in P. multocida serotype A:L3 significantly down-regulated bacterial virulence. The mutant had significantly reduced capsule production but increased resistance to oxidative stress and osmotic pressure. Deleting qseC led to a significant increase in qseB expression. Transcriptome sequencing analysis showed that 1245 genes were regulated by qseC, primarily those genes involved in capsule and LPS biosynthesis and export, biofilm formation, and iron uptake/utilization, as well as several immuno-protection related genes including ompA, ptfA, plpB, vacJ, and sodA. In addition to presenting strong immune protection against P. multocida serotypes A:L1 and A:L3 infection, live ΔqseC also exhibited protection against P. multocida serotype B:L2 and serotype F:L3 infection in a mouse model. The results indicate that QseC regulates capsular production and virulence in P. multocida. Furthermore, the qseC mutant can be used as an attenuated vaccine against P. multocida strains of multiple serotypes.


Asunto(s)
Infecciones por Pasteurella , Pasteurella multocida , Enfermedades de los Roedores , Virulencia , Animales , Ratones , Infecciones por Pasteurella/prevención & control , Infecciones por Pasteurella/veterinaria , Pasteurella multocida/genética , Enfermedades de los Roedores/prevención & control , Virulencia/genética
8.
Foodborne Pathog Dis ; 18(10): 712-717, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33493405

RESUMEN

Salmonella is considered one of the leading causes for foodborne diseases in humans. Pork and its products contaminated with Salmonella are increasingly recognized as an important source of human salmonellosis. The aim of this study was to investigate the antimicrobial resistance and prevalence of integrons in Salmonella isolates from pig farms. In total, 92 of 724 (12.7%) samples were Salmonella-positive, including 64 (15.0%) from fecal samples, 27 (12.6%) from floor samples, 1 (4.5%) from water samples, and 0 from feed and air samples. These isolates showed the highest resistance to tetracycline (85.9%), followed by trimethoprim (67.4%), ampicillin (60.9%), and chloramphenicol (51.1%). In addition, 51 isolates carried the complete class 1 integron, most of which (42/51) harbored antibiotic resistance cassettes. A total of six gene cassettes including orfF, est-X, dfrA1+aadA1, aadA1, dfrA12+aadA2, and sat were identified, in which the most prevalent one was orfF (29.4%). Furthermore, all 19 class 1 integron-positive isolates harboring dfr genes showed resistance to trimethoprim (SXT), suggesting that the trimethoprim resistance gene (dfr) may contribute to the emergence of SXT resistance phenotype. Therefore, considering the significance of integrons and related resistance genes for public health, special measures should be taken to control Salmonella spp. on the pig farms and to prevent spread of integrons and associated resistance genes.


Asunto(s)
Antibacterianos , Integrones , Animales , Antibacterianos/farmacología , Farmacorresistencia Bacteriana/genética , Farmacorresistencia Bacteriana Múltiple/genética , Granjas , Integrones/genética , Salmonella/genética , Porcinos
9.
Int J Mol Sci ; 22(17)2021 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-34502556

RESUMEN

c-Jun N-terminal kinases (JNKs) are among the most crucial mitogen-activated protein kinases (MAPKs) and regulate various cellular processes, including cell proliferation, apoptosis, autophagy, and inflammation. Microbes heavily rely on cellular signaling pathways for their effective replication; hence, JNKs may play important roles in infectious diseases. In this review, we describe the basic signaling properties of MAPKs and JNKs in apoptosis, autophagy, and inflammasome activation. Furthermore, we discuss the roles of JNKs in various infectious diseases induced by viruses, bacteria, fungi, and parasites, as well as their potential to serve as targets for the development of therapeutic agents for infectious diseases. We expect this review to expand our understanding of the JNK signaling pathway's role in infectious diseases and provide important clues for the prevention and treatment of infectious diseases.


Asunto(s)
Enfermedades Transmisibles/enzimología , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Animales , Apoptosis , Autofagia , Enfermedades Transmisibles/metabolismo , Enfermedades Transmisibles/patología , Humanos , Fosforilación
10.
Int J Mol Sci ; 22(8)2021 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-33918100

RESUMEN

Streptococcus pneumoniae (S. pneumoniae) causes severe pulmonary diseases, leading to high morbidity and mortality. It has been reported that inflammasomes such as NLR family pyrin domain containing 3 (NLRP3) and absent in melanoma 2 (AIM2) play an important role in the host defense against S. pneumoniae infection. However, the role of NLRP6 in vivo and in vitro against S. pneumoniae remains unclear. Therefore, we investigated the role of NLRP6 in regulating the S. pneumoniae-induced inflammatory signaling pathway in vitro and the role of NLRP6 in the host defense against S. pneumoniae in vivo by using NLRP6-/- mice. The results showed that the NLRP6 inflammasome regulated the maturation and secretion of IL-1ß, but it did not affect the induction of IL-1ß transcription in S. pneumoniae-infected macrophages. Furthermore, the activation of caspase-1, caspase-11, and gasdermin D (GSDMD) as well as the oligomerization of apoptosis-associated speck-like protein (ASC) were also mediated by NLRP6 in S. pneumoniae-infected macrophages. However, the activation of NLRP6 reduced the expression of NF-κB and ERK signaling pathways in S. pneumoniae-infected macrophages. In vivo study showed that NLRP6-/- mice had a higher survival rate, lower number of bacteria, and milder inflammatory response in the lung compared with wild-type (WT) mice during S. pneumoniae infection, indicating that NLRP6 plays a negative role in the host defense against S. pneumoniae. Furthermore, increased bacterial clearance in NLRP6 deficient mice was modulated by the recruitment of macrophages and neutrophils. Our study provides a new insight on S. pneumoniae-induced activation of NLRP6 and suggests that blocking NLRP6 could be considered as a potential therapeutic strategy to treat S. pneumoniae infection.


Asunto(s)
Interacciones Huésped-Patógeno , Inflamasomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Infecciones Neumocócicas/metabolismo , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/fisiología , Animales , Caspasa 1/metabolismo , Caspasas Iniciadoras/metabolismo , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Péptidos y Proteínas de Señalización Intracelular/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/patología , Transducción de Señal
11.
Infect Immun ; 87(12)2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31570555

RESUMEN

Pasteurella multocida causes a variety of infectious diseases in various species of mammals and birds, resulting in enormous economic loss to the modern livestock and poultry industry. However, the mechanism of host-pathogen interaction is unclear. Here, we found that l-serine levels were significantly decreased in murine lungs infected with P. multocida Exogenous l-serine supplementation significantly increased the survival rate of mice and decreased the colonization of P. multocida in the lungs of mice. Notably, l-serine decreased the macrophage- and neutrophil-mediated inflammatory responses in mice during P. multocida infection.


Asunto(s)
Macrófagos/inmunología , Neutrófilos/inmunología , Infecciones por Pasteurella/inmunología , Pasteurella multocida/inmunología , Serina/farmacología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno/inmunología , Inflamación/tratamiento farmacológico , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Infecciones por Pasteurella/microbiología , Infecciones por Pasteurella/patología , Serina/análisis
12.
Biochem Biophys Res Commun ; 513(3): 675-680, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-30982580

RESUMEN

Streptococcus pneumoniae is a pathogenic bacterium that can cause severe invasive diseases, such as pneumonia, otitis media and meningitis. The pro-inflammatory cytokine, IL-1ß, has been reported to play important role in host defense against S. pneumoniae. The mechanism of IL-1ß maturation and secretion in macrophages has been well studied. However, the precise mechanism of IL-1ß processing within neutrophils upon S. pneumoniae infection remains unclear. In this study, mouse peritoneal neutrophils from C57BL/6 WT and inflammasome components knockout mice were infected by S. pneumoniae in vitro. The results showed that NLRP3 inflammasome is critically involved in neutrophil IL-1ß secretion, while the AIM2 and NLRC4 inflammasomes were dispensable. Moreover, the upstream kinase, JNK, modulates ASC oligomerization and consequent caspase-1 activation and IL-1ß secretion. Additionally, neutrophil serine proteases also participate in IL-1ß secretion by mediating ASC oligomerization and caspase-1 activation. Taken together, these findings indicated that both the NLRP3 inflammasome-related pathway and neutrophil serine protease mediate IL-1ß processing upon S. pneumoniae infection.


Asunto(s)
Caspasa 1/inmunología , Interleucina-1beta/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Infecciones Neumocócicas/inmunología , Serina Proteasas/inmunología , Streptococcus pneumoniae/inmunología , Animales , Proteínas Adaptadoras de Señalización CARD/inmunología , Células Cultivadas , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Neutrófilos/virología , Infecciones Neumocócicas/virología
13.
Mol Cell Probes ; 44: 51-56, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30771482

RESUMEN

Enzootic nasal tumor virus 2 (ENTV-2), the aetiological agent of enzootic nasal adenocarcinoma in goats, is prevalent in China; resulting in substantial economic losses to the goat-breeding industry. Therefore, it is necessary to establish an efficient detection method for the diagnosis and prevention of ENTV-2 infection. More recently, EvaGreen is emerging as a novel alternative fluorescent dye for quantitative real-time PCR because of its low cost, specific amplification and high resolution. In this study, we developed a specific, sensitive, and cost-effective detection method-an EvaGreen-based real-time PCR assay for the detection of ENTV-2. This assay exhibited high specificity and sensitivity and was able to detect ENTV-2 at concentrations as low as 3.0 × 101 copies, which was more sensitive than the conventional PCR method (detection limit, 3.0 × 102 copies). In addition, the reproducibility test indicated that EvaGreen dye in our assay had a good reproducibility. In conclusion, we report that a highly sensitive, specific, and cost-effective EvaGreen-based real-time PCR assay is successful for the rapid detection of ENTV-2.


Asunto(s)
Betaretrovirus/genética , Betaretrovirus/aislamiento & purificación , Neoplasias Nasales/virología , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Zoonosis/virología , Animales , Cartilla de ADN/metabolismo , Cabras/virología , Límite de Detección , ARN Viral/genética , Estándares de Referencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
14.
Arch Virol ; 164(6): 1647-1650, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30877451

RESUMEN

Enzootic nasal adenocarcinoma (ENA) of goats, characterized by transformation of epithelial cells of the ethmoid turbinates, is caused by enzootic nasal tumor virus 2 (ENTV-2). ENTV-2 belongs to the genus Betaretrovirus and has extended its distribution globally with a high prevalence; however, the genetic diversity and genotypic distribution for ENTV-2 have not been analyzed systematically due to the limited availability of sequence data. In this study, an infection by ENTV-2 was detected by RT-PCR in Chongqing in July 2018, and the complete sequence of one strain (CQ1) was determined. Phylogenetic analysis indicated a high degree of genetic heterogeneity among ENTV-2 sequences, with the existence of two main lineages. Lineage 1 and 2 were composed of ENTV-2 from China and the UK, respectively. Although CQ1 was closely related to recent ENTV-2 strains collected in the neighboring provinces of Chongqing (Shaanxi and Sichuan), it formed a separate sublineage of lineage 1 (sublineage 1.3). This report will enhance our understanding of the epidemiology of ENTV-2 in China.


Asunto(s)
Betaretrovirus/clasificación , Técnicas de Genotipaje/métodos , Enfermedades de las Cabras/virología , Neoplasias Nasales/veterinaria , Análisis de Secuencia de ARN/métodos , Animales , Betaretrovirus/genética , Betaretrovirus/aislamiento & purificación , China , Variación Genética , Cabras , Neoplasias Nasales/virología , Filogenia , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Reino Unido
15.
BMC Vet Res ; 15(1): 456, 2019 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-31852466

RESUMEN

BACKGROUND: Salmonella is one of the most important foodborne pathogens, causing outbreaks of human salmonellosis worldwide. Owing to large scales of consumption markets, pork and poultry that contaminated by Salmonella could pose a tremendous threat to public health. The aim of this study was to investigate the contamination of Salmonella from chicken, pork and the environment in slaughtering and retail processes in Chongqing, China. RESULTS: A total of 115 Salmonella isolates were recovered from 1112 samples collected from pork, chicken and the environment. Compared with the isolation rate of samples from chicken (9.50%) and the environment (6.23%), samples from pork had a significant higher isolation rate (44.00%). The isolation rates in slaughterhouses (10.76%) and in supermarkets (10.07%) showed no statistical difference. Thirty different serotypes were identified among all the isolates. S. Derby (n = 26), S. London (n = 16) and S. Rissen (n = 12) were the dominant serotypes. Antimicrobial susceptibility testing revealed that 73.04% isolates were resistant to tetracycline, followed by 66.96% to ampicillin and 59.13% to doxycycline. More than half (50.43%) of the isolates were multidrug resistant (MDR), and most of the MDR isolates were from supermarkets. Multilocus sequence typing results showed 24 out of 115 isolates were ST40, which was the most prevalent. Furthermore, isolates from supermarkets had 20 different sequence types while isolates from slaughterhouses only had 8 different sequence types. CONCLUSION: Our study highlighted that Salmonella was more frequently isolated in pork production chain than that in chicken. Compared with isolates from slaughterhouses, isolates from supermarkets had more MDR profiles and represented a wider range of serotypes and sequence types, indicating that the retail process had more diverse sources of Salmonella contamination than that of slaughtering process.


Asunto(s)
Pollos/microbiología , Carne de Cerdo/microbiología , Salmonella/genética , Mataderos , Animales , China , Farmacorresistencia Bacteriana , Microbiología Ambiental , Manipulación de Alimentos , Microbiología de Alimentos , Tipificación de Secuencias Multilocus , Salmonella/efectos de los fármacos , Salmonella/aislamiento & purificación , Serotipificación , Sus scrofa
16.
Biochem Biophys Res Commun ; 507(1-4): 217-222, 2018 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-30446225

RESUMEN

Streptococcus pneumoniae is a pathogen of significant clinical importance worldwide that can cause severe invasive diseases, such as pneumonia, otitis media and meningitis. Inflammsomes has been reported to participate in host defense against S. pneumoniae infection. S. pneumoniae could induce the assembly of the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3)/absent in melanoma 2 (AIM2) inflammasome, which mediates the activation of caspase-1 and the subsequent maturation of Interleukin-1ß (IL-1ß). However, the precise signals that activate inflammasomes during pneumococcal infection remain to be fully elucidated. In the present study, primary mouse macrophages were selected as a cell model, and the effects of kinases on inflammasome activity induced by S. pneumoniae infection were examined by ELISA and western blotting after pretreatment with a kinase inhibitor. Here, we show that Syk and JNK signaling are required for S. pneumoniae-induced activation of the inflammasome. Inhibitors of Syk and JNK almost abolished the oligomerization of apoptosis-associated speck-like protein containing a caspase-activating and recruitment domain (ASC) and subsequent caspase-1 activation and IL-1ß secretion. Moreover, pneumolysin (PLY) participated in this process and was critical for Syk/JNK activation. These results suggested that the Syk/JNK signaling pathway may play a vital role in the inflammasome activation and modulate host immune responses against S. pneumoniae.


Asunto(s)
Inflamasomas/metabolismo , Sistema de Señalización de MAP Quinasas , Macrófagos/enzimología , Macrófagos/microbiología , Infecciones Neumocócicas/metabolismo , Streptococcus pneumoniae/fisiología , Estreptolisinas/metabolismo , Quinasa Syk/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Caspasa 1/metabolismo , Femenino , Interleucina-1beta/metabolismo , Macrófagos/patología , Ratones Endogámicos C57BL , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/patología
17.
Infect Immun ; 85(9)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28630064

RESUMEN

Pneumolysin (PLY), a major virulence factor of Streptococcus pneumoniae, is a pore-forming cytolysin that modulates host innate responses contributing to host defense against and pathogenesis of pneumococcal infections. Interleukin-1α (IL-1α) has been shown to be involved in tissue damage in a pneumococcal pneumonia model; however, the mechanism by which this cytokine is produced during S. pneumoniae infection remains unclear. In this study, we examined the role of PLY in IL-1α production. Although the strains induced similar levels of pro-IL-1α expression, wild-type S. pneumoniae D39, but not a deletion mutant of the ply gene (Δply), induced the secretion of mature IL-1α from host macrophages, suggesting that PLY is critical for the maturation and secretion of IL-1α during S. pneumoniae infection. Further experiments with calcium chelators and calpain inhibitors indicated that extracellular calcium ions and calpains (calcium-dependent proteases) facilitated the maturation and secretion of IL-1α from D39-infected macrophages. Moreover, we found that PLY plays a critical role in calcium influx and calpain activation, as elevated intracellular calcium levels and the degradation of the calpain substrate α-fodrin were detected in macrophages infected with D39 but not the Δply strain. These results suggested that PLY induces the influx of calcium in S. pneumoniae-infected macrophages, followed by calpain activation and subsequent IL-1α maturation and secretion.


Asunto(s)
Calpaína/metabolismo , Interacciones Huésped-Patógeno , Interleucina-1alfa/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Streptococcus pneumoniae/crecimiento & desarrollo , Estreptolisinas/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Células Cultivadas , Femenino , Ratones Endogámicos C57BL
18.
Eur J Immunol ; 46(2): 291-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26558536

RESUMEN

IL-17 mediates immune responses against extracellular pathogens, and it is associated with the development and pathogenesis of various autoimmune diseases. The expression of IL-17 is regulated by various intracellular signaling cascades. Recently, it has been shown that mechanistic target of rapamycin (mTOR) signaling, comprised mainly of mTORC1 signaling, plays a critical role in IL-17 expression. Here, we review the current knowledge regarding mechanisms by which mTORC1 regulates IL-17 expression. mTORC1 positively modulates IL-17 expression through several pathways, i.e. STAT3, -HIF-1α, -S6K1, and -S6K2. Amino acids (AAs) also regulate IL-17 expression by being the energy source for Th17 cells, and by activating mTORC1 signaling. Altogether, the AA-mTORC1-IL-17 axis has broad therapeutic implications for IL-17-associated diseases, such as EAE, allergies, and colitis.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Hipersensibilidad/inmunología , Interleucina-17/metabolismo , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Células Th17/inmunología , Animales , Enfermedades Autoinmunes/terapia , Regulación de la Expresión Génica , Humanos , Hipersensibilidad/terapia , Interleucina-17/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Terapia Molecular Dirigida , Transducción de Señal
19.
Amino Acids ; 49(12): 1945-1954, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28299479

RESUMEN

Dietary glutamine (Gln) or arginine (Arg) supplementation is beneficial for intestinal health; however, whether Gln or Arg may confer protection against Enterotoxigenic Escherichia coli (ETEC) infection is not known. To address this, we used an ETEC-infected murine model to investigate the protective effects of Gln and Arg. Experimentally, we pre-treated mice with designed diet of Gln or Arg supplementation prior to the oral ETEC infection and then assessed mouse mortality and intestinal bacterial burden. We also determined the markers of intestinal innate immunity in treated mice, including secretory IgA response (SIgA), mucins from goblet cells, as well as antimicrobial peptides from Paneth cells. ETEC colonized in mouse small intestine, including duodenum, jejunum, and ileum, and inhibited the mRNA expression of intestinal immune factors, such as polymeric immunoglobulin receptor (pIgR), cryptdin-related sequence 1C (CRS1C), and Reg3γ. We found that dietary Gln or Arg supplementation decreased bacterial colonization and promoted the activation of innate immunity (e.g., the mRNA expression of pIgR, CRS1C, and Reg3γ) in the intestine of ETEC-infected mice. Our results suggest that dietary arginine or glutamine supplementation may inhibit intestinal ETEC infection through intestinal innate immunity.


Asunto(s)
Arginina/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Glutamina/farmacología , Inmunidad Innata/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Animales , Antiinfecciosos/metabolismo , Carga Bacteriana/efectos de los fármacos , Suplementos Dietéticos , Escherichia coli Enterotoxigénica/efectos de los fármacos , Infecciones por Escherichia coli/inmunología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Íleon/efectos de los fármacos , Íleon/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/fisiopatología , Yeyuno/efectos de los fármacos , Yeyuno/inmunología , Ratones , Sustancias Protectoras/farmacología , Transducción de Señal/efectos de los fármacos
20.
J Pineal Res ; 62(3)2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28152213

RESUMEN

Melatonin affects a variety of physiological processes including circadian rhythms, cellular redox status, and immune function. Importantly, melatonin significantly influences T-cell-mediated immune responses, which are crucial to protect mammals against cancers and infections, but are associated with pathogenesis of many autoimmune diseases. This review focuses on our current understanding of the significance of melatonin in T-cell biology and the beneficial effects of melatonin in T-cell response-based diseases. In addition to expressing both membrane and nuclear receptors for melatonin, T cells have the four enzymes required for the synthesis of melatonin and produce high levels of melatonin. Meanwhile, melatonin is highly effective in modulating T-cell activation and differentiation, especially for Th17 and Treg cells, and also memory T cells. Mechanistically, the influence of melatonin in T-cell biology is associated with membrane and nuclear receptors as well as receptor-independent pathways, for example, via calcineurin. Several cell signaling pathways, including ERK1/2-C/EBPα, are involved in the regulatory roles of melatonin in T-cell biology. Through modulation in T-cell responses, melatonin exerts beneficial effects in various inflammatory diseases, such as type 1 diabetes, systemic lupus erythematosus, and multiple sclerosis. These findings highlight the importance of melatonin signaling in T-cell fate determination, and T cell-based immune pathologies.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Lupus Eritematoso Sistémico/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Proteínas Potenciadoras de Unión a CCAAT/inmunología , Diabetes Mellitus Tipo 1/patología , Humanos , Lupus Eritematoso Sistémico/patología , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Esclerosis Múltiple/patología , Linfocitos T Reguladores/patología , Células Th17/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA