Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
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.
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
4.
Vet Microbiol ; 259: 109130, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34052623

RESUMEN

Pseudorabies virus (PRV), an alphaherpesvirus, causes respiratory and reproductive diseases in pigs and severe nervous symptom in other susceptible hosts. Previous studies showed that PRV infection induced a systemic inflammatory response in mice, indicating that pro-inflammatory cytokines participated in viral neuropathy in mice. The pro-inflammatory cytokine IL-1ß is a key mediator of the inflammatory response and plays an important role in host-response to pathogens. However, the secretion of IL-1ß and its relationship with inflammasome activation during PRV infection remains poorly understood. In this study, we found that PRV infection caused significant secretion of several pro-inflammatory cytokines in macrophages and promoted IL-1ß secretion in an ATP-dependent manner. Furthermore, the expression of IL-1ß can be induced by only PRV infection and depended on NF-κB pathway activation, while the subsequent secretion of IL-1ß was mediated by ATP-induced P2 × 7R activation, loss of intracellular K+, and the subsequent NLRP3 inflammasome activation. By using a mouse infection model, we also found that ATP exacerbated clinical signs and death of mice infected by PRV in a NLRP3-dependent manner. These results indicate that ATP facilitates activation of NLRP3 inflammasome and enhances the pathogenicity of PRV in mice during its acute infection.


Asunto(s)
Adenosina Trifosfato/metabolismo , Herpesvirus Suido 1/metabolismo , Inflamasomas/metabolismo , Macrófagos/virología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Adenosina Trifosfato/inmunología , Animales , Células Cultivadas , Regulación de la Expresión Génica , Herpesvirus Suido 1/genética , Herpesvirus Suido 1/inmunología , Herpesvirus Suido 1/patogenicidad , Inflamasomas/genética , Inflamasomas/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Transducción de Señal
5.
Front Immunol ; 12: 649235, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34017331

RESUMEN

Staphylococcus aureus (S. aureus) is an important zoonotic food-borne pathogen causing severe invasive infections, such as sepsis, pneumonia, food poisoning, toxic shock syndrome and autoimmune diseases. Staphylococcal enterotoxin O (SEO) is a new type of enterotoxins of S. aureus with superantigenic and emetic activity. However, it is still unclear about SEO-induced host inflammatory response. Therefore, the mechanism of SEO-induced interleukin-1ß (IL-1ß) secretion in mouse neutrophils was investigated in this study. Our results showed that recombinant SEO had superantigenic activity with high level of gamma interferon (IFN-γ) production in mouse spleen cells and induced inflammatory cytokines expression including IL-1α, IL-1ß, IL-6 and TNF-α in neutrophils under the action of ATP. In addition, SEO-induced IL-1ß secretion was dependent on activation of Toll like receptor 4 (TLR4), nuclear factor kappa B (NF-κB) and c-jun N-terminal kinase (JNK) signaling pathways. However, SEO-induced IL-1ß secretion was abolished in the neutrophils of NLRP3-/- mice compared with those of wild type mice, indicating that activation of NLRP3 inflammasome mediated IL-1ß secretion during neutrophils stimulation with SEO under the action of ATP. Moreover, this process of SEO+ATP-induced IL-1ß secretion was dependent on potassium (K+) efflux. Taken together, our study suggests that activation of TLR4/JNK/NLRP3 inflammasome signaling pathway mediate maturation and secretion of IL-1ß and provides a new insight on S. aureus virulence factor-induced host immune response.


Asunto(s)
Adenosina Trifosfato/metabolismo , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neutrófilos/inmunología , Infecciones Estafilocócicas/inmunología , Animales , Modelos Animales de Enfermedad , Enterotoxinas/inmunología , Humanos , Inflamasomas/genética , Inflamasomas/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Neutrófilos/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factores de Virulencia/inmunología
6.
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
7.
Sci Adv ; 7(15)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33827820

RESUMEN

Accumulating evidence shows that nervous system governs host immune responses; however, how γ-aminobutyric acid (GABA)ergic system shapes the function of innate immune cells is poorly defined. Here, we demonstrate that GABA transporter (GAT2) modulates the macrophage function. GAT2 deficiency lowers the production of interleukin-1ß (IL-1ß) in proinflammatory macrophages. Mechanistically, GAT2 deficiency boosts the betaine/S-adenosylmethionine (SAM)/hypoxanthine metabolic pathway to inhibit transcription factor KID3 expression through the increased DNA methylation in its promoter region. KID3 regulates oxidative phosphorylation (OXPHOS) via targeting the expression of OXPHOS-related genes and is also critical for NLRP3-ASC-caspase-1 complex formation. Likewise, GAT2 deficiency attenuates macrophage-mediated inflammatory responses in vivo, including lipopolysaccharide-induced sepsis, infection-induced pneumonia, and high-fat diet-induced obesity. Together, we propose that targeting GABAergic system (e.g., GABA transporter) could provide previously unidentified therapeutic opportunities for the macrophage-associated diseases.


Asunto(s)
Lipopolisacáridos , Macrófagos , Caspasas/metabolismo , Proteínas Transportadoras de GABA en la Membrana Plasmática/genética , Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Expresión Génica , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/metabolismo
8.
Toxins (Basel) ; 13(1)2021 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-33477467

RESUMEN

Staphylococcus aureus is a Gram-positive opportunistic pathogen which causes infections in a variety of vertebrates. Virulence factors are the main pathogenesis of S. aureus as a pathogen, which induce the host's innate and adaptive immune responses. Toxic shock syndrome toxin 1 (TSST-1) is one of the most important virulence factors of S. aureus. However, the role of nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) in TSST-1-induced innate immune response is still unclear. Here, purified recombinant TSST-1 (rTSST-1) was prepared and used to stimulate mouse peritoneal macrophages. The results showed that under the action of adenosine-triphosphate (ATP), rTSST-1 significantly induced interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) production in mouse macrophages and the production was dose-dependent. In addition, rTSST-1+ATP-stimulated cytokine production in macrophage depends on the activation of toll like receptor 4 (TLR4), but not TLR2 on the cells. Furthermore, the macrophages of NLRP3-/- mice stimulated with rTSST-1+ATP showed significantly low levels of IL-1ß production compared to that of wild-type mice. These results demonstrated that TSST-1 can induce the expression of inflammatory cytokines in macrophages via the activation of the TLR4 and NLRP3 signaling pathways. Our study provides new information about the mechanism of the TSST-1-inducing host's innate immune responses.


Asunto(s)
Toxinas Bacterianas/inmunología , Citocinas/inmunología , Enterotoxinas/inmunología , Inflamasomas/inmunología , Macrófagos Peritoneales/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Superantígenos/inmunología , Animales , Proteínas Bacterianas/inmunología , Relación Dosis-Respuesta a Droga , Interacciones Huésped-Patógeno , Inmunidad Innata , Interleucina-1beta/inmunología , Ratones , Ratones Noqueados , Proteínas Recombinantes/inmunología , Transducción de Señal , Staphylococcus aureus/inmunología , Receptor Toll-Like 4/inmunología , Factores de Virulencia/inmunología
9.
Vet Microbiol ; 243: 108646, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32273022

RESUMEN

Pasteurella multocida is a gram-negative bacterial pathogen, which causes a large number of diseases in mammals, birds and human. Although the bacterium has been known for decades, the pathogenesis and the mechanisms of P. multocida induced host immunity are poorly understood. Recently, we have reported that nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome plays an important role in caspase-1 activation and IL-1ß secretion in macrophages infected with P. multocida. In this study, the inflammasome activation and IL-1ß secretion were further demonstrated by using high- and low-virulent bovine P. multocida isolates. The results showed that, comparing with macrophages infected with the high-virulent PmCQ2 isolates, the low-virulent PmCQ6 induced higher levels of NLRP3 transcription, caspase-1 activation and mature IL-1ß secretion. Furthermore, the capsule of the high-virulent PmCQ2 was much thicker than that of low-virulent PmCQ6, which indicating that capsular thickness might influence the bacteria colonization and NLRP3 inflammasome activation. The results suggested that differences in maturation of IL-1ß in macrophages upon high- and low- virulent P. multocida infection are critically dependent on the differential activation of NLRP3 inflammasome. This study provided more understanding for the host immune responses induced by P. multocida and further extended the knowledge of P. multocida virulence from the view of host innate immunity.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Inflamasomas/inmunología , Interleucina-1beta/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Pasteurella multocida/patogenicidad , Animales , Cápsulas Bacterianas/inmunología , Caspasa 1/inmunología , Inmunidad Innata , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Organismos Libres de Patógenos Específicos , Virulencia
10.
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.
Vet Microbiol ; 231: 207-213, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30955811

RESUMEN

Pasteurella multocida is a Gram-negative bacterium that is responsible for a variety of diseases in birds and mammals, including humans. We have previously reported that the P. multocida serotype A strain PmCQ2 causes severe lung pneumonia in bovines. Transcriptomic analysis showed that many genes related to the immune response were significantly upregulated in the lungs of mice infected with P. multocida compared with uninfected mice. However, the mechanism by which P. multocida induces host inflammatory cytokine secretion is poorly understood. In this study, the mechanism of caspase-1 activation and subsequent IL-1ß secretion in macrophages infected with P. multocida was elucidated. The nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome was shown to be involved in inducing this cellular response. Compared with wild-type macrophages, Nlrp3-/- macrophages exhibited a clear decrease in caspase-1 activation and IL-1ß secretion in response to P. multocida infection. Furthermore, spleen tyrosine kinase (Syk) was indicated to be involved in IL-1ß secretion, possibly by regulating the NLRP3 inflammasome. Our results provide new insight into the host proinflammatory immune response against P. multocida and the critical involvement of the NLRP3 inflammasome in this activity.


Asunto(s)
Caspasa 1/metabolismo , Interleucina-1beta/inmunología , Macrófagos/microbiología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Pasteurella multocida/inmunología , Animales , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Inflamasomas/inmunología , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Pasteurella multocida/genética , Fagocitosis , Transducción de Señal/inmunología , Quinasa Syk/metabolismo , Regulación hacia Arriba
13.
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
14.
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
15.
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
16.
Front Immunol ; 9: 1070, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29881379

RESUMEN

Betaine is known as trimethylglycine and is widely distributed in animals, plants, and microorganisms. Betaine is known to function physiologically as an important osmoprotectant and methyl group donor. Accumulating evidence has shown that betaine has anti-inflammatory functions in numerous diseases. Mechanistically, betaine ameliorates sulfur amino acid metabolism against oxidative stress, inhibits nuclear factor-κB activity and NLRP3 inflammasome activation, regulates energy metabolism, and mitigates endoplasmic reticulum stress and apoptosis. Consequently, betaine has beneficial actions in several human diseases, such as obesity, diabetes, cancer, and Alzheimer's disease.


Asunto(s)
Antiinflamatorios/farmacología , Betaína/farmacología , Inflamación/etiología , Inflamación/metabolismo , Animales , Antiinflamatorios/química , Antiinflamatorios/uso terapéutico , Betaína/química , Betaína/uso terapéutico , Biomarcadores , Estrés del Retículo Endoplásmico/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Humanos , Inflamación/tratamiento farmacológico , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
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.
Cell Death Dis ; 8(3): e2655, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28252650

RESUMEN

T-cell-mediated immune responses aim to protect mammals against cancers and infections, and are also involved in the pathogenesis of various inflammatory or autoimmune diseases. Cellular uptake and the utilization of nutrients is closely related to the T-cell fate decision and function. Research in this area has yielded surprising findings in the importance of amino-acid transporters for T-cell development, homeostasis, activation, differentiation and memory. In this review, we present current information on amino-acid transporters, such as LAT1 (l-leucine transporter), ASCT2 (l-glutamine transporter) and GAT-1 (γ-aminobutyric acid transporter-1), which are critically important for mediating peripheral naive T-cell homeostasis, activation and differentiation, especially for Th1 and Th17 cells, and even memory T cells. Mechanically, the influence of amino-acid transporters on T-cell fate decision may largely depend on the mechanistic target of rapamycin complex 1 (mTORC1) signaling. These discoveries remarkably demonstrate the role of amino-acid transporters in T-cell fate determination, and strongly indicate that manipulation of the amino-acid transporter-mTORC1 axis could ameliorate many inflammatory or autoimmune diseases associated with T-cell-based immune responses.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Aminoácidos/metabolismo , Diferenciación Celular/fisiología , Activación de Linfocitos/fisiología , Linfocitos T/metabolismo , Linfocitos T/fisiología , Animales , Homeostasis/fisiología , Humanos
19.
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
20.
Amino Acids ; 45(4): 947-55, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23884693

RESUMEN

This study was conducted to determine the effects of graded doses of L-glutamine supplementation on the replication and distribution of Pasteurella multocida, and the expression of its major virulence factors in mouse model. Mice were randomly assigned to the basal diet supplemented with 0, 0.5, 1.0 or 2.0 % glutamine. Pasteurella multocida burden was detected in the heart, liver, spleen, lung and kidney after 12 h of P. multocida infection. The expression of major virulence factors, toll-like receptors (TLRs), proinflammatory cytokines (interleukin-1 beta, interleukin-6, and tumor necrosis factor alpha) and anti-oxidative factors (GPX1 and CuZnSOD) was analyzed in the lung and spleen. Dietary 0.5 % glutamine supplementation has little significant effect on these parameters, compared to those with basal diet. However, results showed that a high dose of glutamine supplementation increased the P. multocida burden (P < 0.001) and the expression of its major virulence factors (P < 0.05) as compared to those with a lower dose of supplementation. In the lung, high dose of glutamine supplementation inhibited the proinflammatory responses (P < 0.05) and TLRs signaling (P < 0.05). In the spleen, the effect of glutamine supplementation on different components in TLR signaling depends on glutamine concentration, and high dose of glutamine supplementation activated the proinflammatory response. In conclusion, glutamine supplementation increased P. multocida burden and the expression of its major virulence factors, while affecting the functions of the lung and spleen.


Asunto(s)
Suplementos Dietéticos , Regulación de la Expresión Génica/efectos de los fármacos , Glutamina/administración & dosificación , Glutamina/farmacología , Infecciones por Pasteurella/microbiología , Pasteurella/genética , Factores de Virulencia/genética , Animales , Antioxidantes/metabolismo , Citocinas/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Inflamación/inmunología , Inflamación/microbiología , Ratones , Ratones Endogámicos , Especificidad de Órganos , Receptores Toll-Like/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA