Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Brief Bioinform ; 25(4)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38975892

RESUMO

Understanding the biological functions and processes of genes, particularly those not yet characterized, is crucial for advancing molecular biology and identifying therapeutic targets. The hypothesis guiding this study is that the 3D proximity of genes correlates with their functional interactions and relevance in prokaryotes. We introduced 3D-GeneNet, an innovative software tool that utilizes high-throughput sequencing data from chromosome conformation capture techniques and integrates topological metrics to construct gene association networks. Through a series of comparative analyses focused on spatial versus linear distances, we explored various dimensions such as topological structure, functional enrichment levels, distribution patterns of linear distances among gene pairs, and the area under the receiver operating characteristic curve by utilizing model organism Escherichia coli K-12. Furthermore, 3D-GeneNet was shown to maintain good accuracy compared to multiple algorithms (neighbourhood, co-occurrence, coexpression, and fusion) across multiple bacteria, including E. coli, Brucella abortus, and Vibrio cholerae. In addition, the accuracy of 3D-GeneNet's prediction of long-distance gene interactions was identified by bacterial two-hybrid assays on E. coli K-12 MG1655, where 3D-GeneNet not only increased the accuracy of linear genomic distance tripled but also achieved 60% accuracy by running alone. Finally, it can be concluded that the applicability of 3D-GeneNet will extend to various bacterial forms, including Gram-negative, Gram-positive, single-, and multi-chromosomal bacteria through Hi-C sequencing and analysis. Such findings highlight the broad applicability and significant promise of this method in the realm of gene association network. 3D-GeneNet is freely accessible at https://github.com/gaoyuanccc/3D-GeneNet.


Assuntos
Redes Reguladoras de Genes , Software , Algoritmos , Biologia Computacional/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo
2.
Domest Anim Endocrinol ; 88: 106851, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38733944

RESUMO

Colostrum and milk offer a complete diet and vital immune protection for newborn mammals with developing immune systems. High immunoglobulin levels in colostrum serve as the primary antibody source for newborn piglets and calves. Subsequent milk feeding support continued local antibody protection against enteric pathogens, as well as maturation of the developing immune system and provide nutrients for newborn growth. Mammals have evolved hormonal strategies that modulate the levels of immunoglobulins in colostrum and milk to facilitate effective lactational immunity. In addition, hormones regulate the gut-mammary gland-secretory immunoglobulin A (sIgA) axis in pregnant mammals, controlling the levels of sIgA in milk, which serves as the primary source of IgA for piglets and helps them resist pathogens such as PEDV and TGEV. In the present study, we review the existing studies on the interactions between hormones and the gut-mammary-sIgA axis/lactogenic immunity in mammals and explore the potential mechanisms of hormonal regulation that have not been studied in detail, to draw attention to the role of hormones in influencing the immune response of pregnant and lactating mammals and their offspring, and highlight the effect of hormones in regulating sIgA-mediated anti-infection processes in colostrum and milk. Discussion of the relationship between hormones and lactogenic immunity may lead to a better way of improving lactogenic immunity by determining a better injection time and developing new vaccines.


Assuntos
Colostro , Hormônios , Lactação , Animais , Suínos/imunologia , Bovinos/imunologia , Bovinos/fisiologia , Feminino , Lactação/fisiologia , Colostro/imunologia , Colostro/química , Hormônios/fisiologia , Gravidez , Leite/química , Imunoglobulina A Secretora
3.
Vet Microbiol ; 294: 110106, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38776767

RESUMO

Glaesserella parasuis (G. parasuis) is the causative agent of porcine Glässer's disease, resulting in high mortality rates in pigs due to excessive inflammation-induced tissue damage. Previous studies investigating the protective effects of G. parasuis vaccination indicated a possible role of ApoA1 in reflecting disease progression following G. parasuis infection. However, the mechanisms of ApoA1 expression and its role in these infections are not well understood. In this investigation, newborn porcine tracheal (NPTr) epithelial cells infected with G. parasuis were used to elucidate the molecular mechanism and role of ApoA1. The study revealed that the AMPK pathway activation inhibited ApoA1 expression in NPTr cells infected with G. parasuis for the first time. Furthermore, Egr1 was identified as a core transcription factor regulating ApoA1 expression using a CRISPR/Cas9-based system. Importantly, it was discovered that APOA1 protein significantly reduced apoptosis, pyroptosis, necroptosis, and inflammatory factors induced by G. parasuis in vivo. These findings not only enhance our understanding of ApoA1 in response to bacterial infections but also highlight its potential in mitigating tissue damage caused by G. parasuis infection.


Assuntos
Proteínas Quinases Ativadas por AMP , Apolipoproteína A-I , Proteína 1 de Resposta de Crescimento Precoce , Haemophilus parasuis , Transdução de Sinais , Doenças dos Suínos , Animais , Suínos , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Haemophilus parasuis/genética , Doenças dos Suínos/microbiologia , Doenças dos Suínos/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Infecções por Haemophilus/veterinária , Infecções por Haemophilus/microbiologia , Células Epiteliais/microbiologia , Regulação da Expressão Gênica , Traqueia/microbiologia , Traqueia/metabolismo , Apoptose , Animais Recém-Nascidos
4.
Virus Evol ; 10(1): veae005, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38361823

RESUMO

Understanding phylogenetic relationships among species is essential for many biological studies, which call for an accurate phylogenetic tree to understand major evolutionary transitions. The phylogenetic analyses present a major challenge in estimation accuracy and computational efficiency, especially recently facing a wave of severe emerging infectious disease outbreaks. Here, we introduced a novel, efficient framework called Bases-dependent Rapid Phylogenetic Clustering (Bd-RPC) for new sample placement for viruses. In this study, a brand-new recoding method called Frequency Vector Recoding was implemented to approximate the phylogenetic distance, and the Phylogenetic Simulated Annealing Search algorithm was developed to match the recoded distance matrix with the phylogenetic tree. Meanwhile, the indel (insertion/deletion) was heuristically introduced to foreign sequence recognition for the first time. Here, we compared the Bd-RPC with the recent placement software (PAGAN2, EPA-ng, TreeBeST) and evaluated it in Alphacoronavirus, Alphaherpesvirinae, and Betacoronavirus by using Split and Robinson-Foulds distances. The comparisons showed that Bd-RPC maintained the highest precision with great efficiency, demonstrating good performance in new sample placement on all three virus genera. Finally, a user-friendly website (http://www.bd-rpc.xyz) is available for users to classify new samples instantly and facilitate exploration of the phylogenetic research in viruses, and the Bd-RPC is available on GitHub (http://github.com/Bin-Ma/bd-rpc).

5.
Microbiol Spectr ; 11(6): e0164423, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37874148

RESUMO

Tsukamurella species have been clinically regarded as rare but emerging opportunistic pathogens causing various infections in humans. Tsukamurella pneumonia has often been misdiagnosed as pulmonary tuberculosis due to its clinical presentation resembling tuberculosis-like syndromes. Tsukamurella species have also been confused in the laboratory with other phylogenetic bacteria, such as Gordonia. This study aimed to investigate the clinical, microbiological, and molecular characteristics; species distribution; and antimicrobial susceptibility of Tsukamurella species. Immunodeficiency and chronic pulmonary disease appeared to be risk factors for Tsukamurella pneumonia, and the presence of bronchiectasis and pulmonary nodules on imaging was highly correlated with this infection. The study confirmed that groEL (heat shock protein 60) and secA (the secretion ATPase) genes are reliable for identifying Tsukamurella species. Additionally, the ssrA (stable small RNA) gene showed promise as a tool for discriminating between different Tsukamurella species with the shortest sequence length. In terms of antimicrobial susceptibility, quinolones, trimethoprim/sulfamethoxazole, amikacin, minocycline, linezolid, and tigecycline demonstrated potent in vitro activity against Tsukamurella isolates in our study. The study also proposed a resistance mechanism involving a substitution (S91R) within the quinolone-resistance-determining region of the gyrA gene, which confers resistance to levofloxacin and ciprofloxacin. Furthermore, we found that disk diffusion testing is not suitable for testing the susceptibilities of Tsukamurella isolates to ciprofloxacin, imipenem, and minocycline. In conclusion, our systematic investigation may contribute to a better understanding of this rare pathogen. Tsukamurella species are rare but emerging human pathogens that share remarkable similarities with other mycolic acid-containing genera of the order Actinomycetales, especially Mycobacterium tuberculosis. Consequently, misdiagnosis and therapeutic failures can occur in clinical settings. Despite the significance of accurate identification, antimicrobial susceptibility, and understanding the resistance mechanism of this important genus, our knowledge in these areas remains fragmentary and incomplete. In this study, we aimed to address these gaps by investigating promising identification methods, the antimicrobial susceptibility patterns, and a novel quinolone resistance mechanism in Tsukamurella species, utilizing a collection of clinical isolates. The findings of our study will contribute to improve diagnosis and successful management of infections caused by Tsukamurella species, as well as establishing well-defined performance and interpretive criteria for antimicrobial susceptibility testing.


Assuntos
Actinomycetales , Anti-Infecciosos , Pneumonia , Quinolonas , Humanos , Minociclina , Filogenia , Testes de Sensibilidade Microbiana , Actinomycetales/genética , Ciprofloxacina , Hospitais de Ensino , China , Antibacterianos/farmacologia
6.
Microbiol Spectr ; 11(1): e0387122, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36602356

RESUMO

Identification of microbial functional association networks allows interpretation of biological phenomena and a greater understanding of the molecular basis of pathogenicity and also underpins the formulation of control measures. Here, we describe PPNet, a tool that uses genome information and analysis of phylogenetic profiles with binary similarity and distance measures to derive large-scale bacterial gene association networks of a single species. As an exemplar, we have derived a functional association network in the pig pathogen Streptococcus suis using 81 binary similarity and dissimilarity measures which demonstrates excellent performance based on the area under the receiver operating characteristic (AUROC), the area under the precision-recall (AUPR), and a derived overall scoring method. Selected network associations were validated experimentally by using bacterial two-hybrid experiments. We conclude that PPNet, a publicly available (https://github.com/liyangjie/PPNet), can be used to construct microbial association networks from easily acquired genome-scale data. IMPORTANCE This study developed PPNet, the first tool that can be used to infer large-scale bacterial functional association networks of a single species. PPNet includes a method for assigning the uniqueness of a bacterial strain using the average nucleotide identity and the average nucleotide coverage. PPNet collected 81 binary similarity and distance measures for phylogenetic profiling and then evaluated and divided them into four groups. PPNet can effectively capture gene networks that are functionally related to phenotype from publicly prokaryotic genomes, as well as provide valuable results for downstream analysis and experiment testing.


Assuntos
Genes Bacterianos , Células Procarióticas , Animais , Suínos , Filogenia , Bactérias/genética , Redes Reguladoras de Genes
7.
Virulence ; 14(1): 2171636, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-36694280

RESUMO

Haemophilus parasuis (H. parasuis) causes exudative inflammation, implying endothelial dysfunction during pathogen infection. However, so far, the molecular mechanism of endothelial dysfunction caused by H. parasuis has not been clarified. By using the transwell-based cell co-culture system, we demonstrate that knocking out resistin in porcine alveolar macrophages (PAMs) dramatically attenuated endothelial monolayer damage caused by H. parasuis. The resistin secreted by PAMs inhibited the expression of the tight junction proteins claudin-5 and occludin rather than the adherens junction protein VE-cadherin in co-cultured porcine aortic endothelial cells (PAECs). Furthermore, we demonstrate that resistin regulated claudin-5 and occludin expression and monolayer PAEC permeability in an LKB1/AMPK/mTOR pathway-dependent manner. Additionally, we reveal that the outer membrane lipoprotein gene lppA in H. parasuis induced resistin expression in PAMs, as deleting lppA reduced resistin expression in H. parasuis-infected PAMs, causing a significant change in LKB1/AMPK/mTOR pathway activity in co-cultured PAECs, thereby restoring tight junction protein levels and endothelial monolayer permeability. Thus, we postulate that the H. parasuis lppA gene enhances resistin production in PAMs, disrupting tight junctions in PAECs and causing endothelial barrier dysfunction. These findings elucidate the pathogenic mechanism of exudative inflammation caused by H. parasuis for the first time and provide a more profound angle of acute exudative inflammation caused by bacteria.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis , Suínos , Animais , Macrófagos Alveolares/microbiologia , Haemophilus parasuis/genética , Células Endoteliais , Resistina/genética , Resistina/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Claudina-5/metabolismo , Ocludina/metabolismo , Infecções por Haemophilus/veterinária , Infecções por Haemophilus/microbiologia , Inflamação , Serina-Treonina Quinases TOR/metabolismo
8.
Front Cell Infect Microbiol ; 12: 952377, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36034702

RESUMO

Glaesserella parasuis is well-known for causing Glässer's disease, which costs the worldwide swine industry millions of dollars each year. It has been reported the symptom of pleural thickening during Glässer's disease but this symptom has received little attention. And there is no research on the elements which promote pleural thickening. In this study, pleural thickening was discovered to be associated with increased collagen fibers and elastic fibers. Furthermore, collagen-I and elastin were found to be up-regulated and concentrated in the pleura at the mRNA and protein levels following infection. To summarize, our findings add to the theoretical understanding of Glässer's disease and provide strong support for further research into the pathogenic mechanism of Glaesserella parasuis and the program's target treatment.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis , Doenças dos Suínos , Animais , Colágeno , Elastina , Suínos
9.
Dev Comp Immunol ; 128: 104327, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34863954

RESUMO

Haemophilus parasuis is a widespread bacterial pathogen causing acute systemic inflammation and leading to the sudden death of piglets. Resistin, a multifunctional peptide hormone previously demonstrated to influence the inflammation in porcine, was extremely increased in H. parasuis-infected tissues. However, the mechanism of resistin expression regulation in porcine, especially during pathogen infection, remains unclear. In the present study, we explored for the first time the transcription factor and signaling pathway mediating the expression of pig resistin during H. parasuis stimulation. We found that H. parasuis induced the expression of pig resistin in a time- and dose-dependent manner via the transcription factor Ets2 in porcine alveolar macrophages during H. parasuis stimulation. Moreover, the expression of Ets2 was mediated by the activation of the p38 MAPK pathway induced by H. parasuis, thus promoting resistin production. These results revealed a novel view of the molecular mechanism of pig resistin production during acute inflammation induced by pathogenic bacteria.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis , Doenças dos Suínos , Animais , Infecções por Haemophilus/metabolismo , Haemophilus parasuis/fisiologia , Macrófagos Alveolares/metabolismo , Resistina/metabolismo , Suínos , Doenças dos Suínos/metabolismo , Doenças dos Suínos/microbiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Cytokine ; 136: 155287, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32950027

RESUMO

Haemophilus parasuis induces severe acute systemic infection in pigs, characterized by fibrinous polyserositis, polyarthritis and meningitis. Our previous study demonstrated that H. parasuis induced the activation of p38 mitogen-activated protein kinase (MAPK) pathway, increasing the expression of proinflammatory genes and mediating H. parasuis-induced inflammation. Moreover, Wnt/ß-catenin signaling activation induced by H. parasuis disrupts the adherens junction between epithelial cells and initiates the epithelial-mesenchymal transition (EMT). In the present study, p38 MAPK was found to be involved in the accumulation of nuclear location of ß-catenin during H. parasuis infection in PK-15 and NPTr cells, via modulating the expression of dickkofp-1 (DKK-1), a negative regulator of Wnt/ß-catenin signaling. We generated DKK-1 knockout cell lines by CRISPR/Cas9-mediated genome editing in PK-15 and NPTr cells, and found that knockout of DKK-1 led to the dysfunction of p38 MAPK in regulating Wnt/ß-catenin signaling activity in H. parasuis-infected cells. Furthermore, p38 MAPK activity was independent of the activation of Wnt/ß-catenin signaling during H. parasuis infection. This is the first study to explore the crosstalk between p38 MAPK and Wnt/ß-catenin signaling during H. parasuis infection. It provides a more comprehensive view of intracellular signaling pathways during pathogenic bacteria-induced acute inflammation.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Doenças dos Suínos , Suínos/imunologia , Via de Sinalização Wnt/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Animais , Linhagem Celular , Infecções por Haemophilus/imunologia , Infecções por Haemophilus/microbiologia , Infecções por Haemophilus/veterinária , Suínos/microbiologia , Doenças dos Suínos/imunologia , Doenças dos Suínos/microbiologia
11.
Dev Comp Immunol ; 105: 103588, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31887319

RESUMO

Haemophilus parasuis infection causes typical acute systemic inflammation in pigs, is characterized by fibrinous polyserositis inflammation, and results in great economic losses to the swine industry worldwide. However, the molecular details of how the host modulates the acute inflammatory response induced by H. parasuis are largely unknown. In previous studies, we found that H. parasuis high-virulence strain SH0165 infection induced the activation of both Wnt/ß-catenin and NF-κB signaling in PK-15 and NPTr cells. In this study, we found that the activation of NF-κB, a central hub in inflammatory signaling, was impeded by the Wnt/ß-catenin pathway during H. parasuis infection. In contrast, blocking NF-κB activity had no effect on the Wnt/ß-catenin pathway during H. parasuis infection. Furthermore, we found that the inhibitory effect of ß-catenin on NF-κB activity was mediated by its target gene, pig cyclooxygenase-2 (COX-2). Therefore, we demonstrated that H. parasuis infection activates the canonical Wnt/ß-catenin signaling pathway, which leads to decreased NF-κB activity, reducing the acute inflammatory response in pigs. Additionally, the data provide a possible perspective for understanding the anti-inflammatory role of Wnt/ß-catenin in pigs during bacterial infection.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Células Epiteliais/metabolismo , Infecções por Haemophilus/metabolismo , Haemophilus parasuis/fisiologia , Inflamação/imunologia , Rim/citologia , Suínos/imunologia , Animais , Linhagem Celular , Ciclo-Oxigenase 2/genética , Células Epiteliais/imunologia , Infecções por Haemophilus/imunologia , Haemophilus parasuis/patogenicidade , NF-kappa B/metabolismo , Receptor Cross-Talk , Suínos/microbiologia , Virulência , Via de Sinalização Wnt , beta Catenina/metabolismo
12.
Artigo em Inglês | MEDLINE | ID: mdl-31106159

RESUMO

Bacterial infections activate autophagy and autophagy restricts pathogens such as Haemophilus parasuis through specific mechanisms. Autophagy is associated with the pathogenesis of H. parasuis. However, the mechanisms have not been clarified. Here, we monitored autophagy processes using confocal microscopy, western blot, and transmission electron microscopy (TEM) and found that H. parasuis SH0165 (high-virulent strain) but not HN0001 (non-virulent strain) infection enhanced autophagy flux. The AMPK/mTOR autophagy pathway was required for autophagy initiation and ATG5, Beclin-1, ATG7, and ATG16L1 emerged as important components in the generation of the autophagosome during H. parasuis infection. Moreover, autophagy induced by H. parasuis SH0165 turned to fight against invaded bacteria and inhibit inflammation. Then we further demonstrated that autophagy blocked the production of the cytokines IL-8, CCL4, and CCL5 induced by SH0165 infection through the inhibition of NF-κB, p38, and JNK MAPK signaling pathway. Therefore, our findings suggest that autophagy may act as a cellular defense mechanism in response to H. parasuis and provide a new way that autophagy protects the host against H. parasuis infection.


Assuntos
Autofagia , Mecanismos de Defesa , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Infecções por Haemophilus/veterinária , Haemophilus parasuis/imunologia , Inflamação/imunologia , Animais , Linhagem Celular , Citocinas/metabolismo , Infecções por Haemophilus/imunologia , Modelos Teóricos , Transdução de Sinais , Suínos
13.
Cytokine ; 111: 325-333, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30269029

RESUMO

TRAF family member-associated NF-κB activator (TANK)-binding kinase 1 (TBK1) serves as hub molecule at the crossroad of multiple signaling pathways of type I interferon (IFN) induction. The importance of TBK1 in innate immunity has been demonstrated in mammalian, however the characterization and function of TBK1 in avian remains largely unknown. In this study, we cloned duck TBK1 (duTBK1) from duck embryo fibroblasts (DEFs) for the first time, which encoded 729 amino acids and had a high amino acid identity with goose and cormorant TBK1s. The duTBK1 showed a diffuse cytoplasmic localization in DEFs and was extensively expressed in all tested tissues. Overexpression of duTBK1 induced IFN-ß production through the activation of IRF1 and NF-κB in DEFs. The N-terminal kinase domain and the ubiquitin-like domain in middle of duTBK1 played pivotal roles in IFN-ß induction as well as in IRF1 and NF-κB activation. Furthermore, knockdown of duTBK1 by small interfering RNA significantly decreased poly(I:C)- or Sendai virus (SeV)-induced IFN-ß expression. In addition, duTBK1 expression dramatically reduced the replication of both duck reovirus (DRV) and duck Tembusu virus (DTMUV) in DEFs. These results suggested that the duTBK1 played a pivotal role in mediating duck antiviral innate immunity.


Assuntos
Proteínas Aviárias/imunologia , Patos/imunologia , Interferon beta/imunologia , Sequência de Aminoácidos , Aminoácidos/imunologia , Animais , Linhagem Celular , Patos/virologia , Fibroblastos/imunologia , Fibroblastos/virologia , Flavivirus/imunologia , Gansos/imunologia , Gansos/virologia , Imunidade Inata/imunologia , Fator Regulador 1 de Interferon/imunologia , NF-kappa B/imunologia , Poli I-C/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Reoviridae/imunologia , Alinhamento de Sequência , Transdução de Sinais/imunologia
14.
Artigo em Inglês | MEDLINE | ID: mdl-30258822

RESUMO

In this study, animal experimentation verified that the canonical Wnt/ß-catenin signaling pathway was activated under a reduced activity of p-ß-catenin (Ser33/37/Thr41) and an increased accumulation of ß-catenin in the lungs and kidneys of pigs infected with a highly virulent strain of H. parasuis. In PK-15 and NPTr cells, it was also confirmed that infection with a high-virulence strain of H. parasuis induced cytoplasmic accumulation and nuclear translocation of ß-catenin. H. parasuis infection caused a sharp degradation of E-cadherin and an increase of the epithelial cell monolayer permeability, as well as a broken interaction between ß-catenin and E-cadherin dependent on Wnt/ß-catenin signaling pathway. Moreover, Wnt/ß-catenin signaling pathway also contributed to the initiation of epithelial-mesenchymal transition (EMT) during high-virulence strain of H. parasuis infection with expression changes of epithelial/mesenchymal markers, increased migratory capabilities as well as the morphologically spindle-like switch in PK-15 and NPTr cells. Therefore, we originally speculated that H. parasuis infection activates the canonical Wnt/ß-catenin signaling pathway leading to a disruption of the epithelial barrier, altering cell structure and increasing cell migration, which results in severe acute systemic infection characterized by fibrinous polyserositis during H. parasuis infection.


Assuntos
Junções Aderentes/patologia , Infecções por Haemophilus/patologia , Haemophilus parasuis/crescimento & desenvolvimento , Transdução de Sinais , Doenças dos Suínos/patologia , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Linhagem Celular , Células Epiteliais/patologia , Interações Hospedeiro-Patógeno , Rim/patologia , Pulmão/patologia , Suínos
15.
Dev Comp Immunol ; 79: 158-165, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29097236

RESUMO

Haemophilus parasuis, an important swine pathogen, was recently proven able to invade into endothelial or epithelial cell in vitro. NOD1/2 are specialized NLRs that participate in the recognition of pathogens able to invade intracellularly and therefore, we assessed that the contribution of NOD1/2 to inflammation responses during H. parasuis infection. We observed that H. parasuis infection enhanced NOD2 expression and RIP2 phosphorylation in porcine kidney 15 cells. Our results also showed that knock down of NOD1/2 or RIP2 expression respectively significantly decreased H. parasuis-induced NF-κB activity, while the phosphorylation level of p38, JNK or ERK was not changed. Moreover, real-time PCR result showed that NOD1, NOD2 or RIP2 was involved in the expression of CCL4, CCL5 and IL-8. Inhibition of NOD1 and NOD2 significantly reduced CCL5 promoter activity, even in a more effective way compared with inhibition of TLR.


Assuntos
Células Endoteliais/imunologia , Células Epiteliais/imunologia , Infecções por Haemophilus/imunologia , Haemophilus parasuis/imunologia , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Suínos/imunologia , Animais , Linhagem Celular , Quimiocina CCL4/metabolismo , Quimiocina CCL5/metabolismo , Células Endoteliais/microbiologia , Células Epiteliais/microbiologia , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD1/genética , Proteína Adaptadora de Sinalização NOD2/genética , RNA Interferente Pequeno/genética , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/genética , Transdução de Sinais
16.
Dev Comp Immunol ; 76: 255-261, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28666650

RESUMO

Human LSm14A is a key component of processing body (P-body) assembly that mediates interferon-ß (IFN-ß) production by sensing viral RNA or DNA. To the best of our knowledge, we are the first to report duck LSm14A (duLSm14A) cloning from duck embryo fibroblasts (DEFs). Full-length duLSm14A encoded 461 amino acids and was highly homologous with chicken and swan goose sequences. More interestingly, the duLSm14A mRNA was extensively expressed in all the studied tissues. In DEFs, duLSm14A was localized in the cytoplasm as P-body-like dots. Expression of duLSm14A induced IFN-ß through the activation of interferon regulatory factor-1 and nuclear factor-κB in DEFs. Furthermore, knockdown of duLSm14A by small interfering RNA notably decreased poly(I:C)- or duck reovirus-induced IFN-ß production. The present study results indicate that the duLSm14A is an essential sensor that mediates duck innate immunity against viral infections.


Assuntos
Proteínas Aviárias/metabolismo , Doenças das Aves/imunologia , Patos/imunologia , Fibroblastos/fisiologia , Receptores de Reconhecimento de Padrão/metabolismo , Infecções por Reoviridae/imunologia , Reoviridae/imunologia , Animais , Proteínas Aviárias/genética , Células Cultivadas , Clonagem Molecular , Humanos , Imunidade Inata , Fator Regulador 1 de Interferon/metabolismo , Interferon beta/metabolismo , NF-kappa B/metabolismo , Filogenia , Receptores de Reconhecimento de Padrão/genética , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo
19.
Mol Immunol ; 67(2 Pt B): 661-6, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26198698

RESUMO

RANTES is a member of the CC chemokine involved in inflammation and immune response during pathogen infection. In our previous study, Haemophilus parasuis (H. parasuis), which is responsible for the great economic losses in the pig industry worldwide, has been shown to enhance RANTES expression in PK-15 cells. However, the mechanisms behind this biological phenomenon have remained unclear. In this study, we showed that H. parasuis infection significantly upregulated RANTES gene transcription in a time- and dose-dependent manner. Promoter analysis by site-directed mutagenesis indicated that the nuclear factor NF-κB binding site was the most important cis-regulatory element controlling H. parasuis-induced RANTES transcription. Inhibition of NF-κB and JNK activity also significantly reduced H. parasuis-induced RANTES production. In addition, TLRs signaling pathway was found to be involved in H. parasuis induced-RANTES expression. These results represent an important molecular mechanism whereby H. parasuis induced RANTES in the inflammatory response.


Assuntos
Quimiocina CCL5/metabolismo , Infecções por Haemophilus/microbiologia , Haemophilus parasuis/fisiologia , Animais , Linhagem Celular , Quimiocina CCL5/genética , Infecções por Haemophilus/genética , Sistema de Sinalização das MAP Quinases , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sus scrofa , Receptores Toll-Like/metabolismo , Transcrição Gênica
20.
Mol Immunol ; 65(2): 360-6, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25733389

RESUMO

Glässer's disease in pigs caused by Haemophilus parasuis is characterized by a severe membrane inflammation. In our previous study, we have identified activation of the transcription factor NF-κB after H. parasuis infection of porcine epithelial cells. In this study, we found that H. parasuis infection also contributed to the activation of p38/JNK MAPK pathway predominantly linked to inflammation, but not the ERK MAPK pathway associated with growth, differentiation and development. Inhibition of NF-κB, p38 and JNK but not ERK activity significantly reduced IL-8 and CCL4 expression by H. parasuis. We also found TLR1, TLR2, TLR4 and TLR6 were required for NF-κB, p38 and JNK MAPK activation. Furthermore, MyD88 and TRIF signaling cascades were essential for H. parasuis-induced NF-κB activation. These results provided new insights into the molecular pathways underlying the inflammatory response induced by H. parasuis.


Assuntos
Infecções por Haemophilus/imunologia , Infecções por Haemophilus/veterinária , Haemophilus parasuis/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , NF-kappa B/imunologia , Doenças dos Suínos/imunologia , Receptores Toll-Like/imunologia , Animais , Linhagem Celular , Quimiocina CCL4/imunologia , Infecções por Haemophilus/patologia , Interleucina-8/imunologia , MAP Quinase Quinase 4/imunologia , Suínos , Doenças dos Suínos/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA