Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Vet Microbiol ; 288: 109954, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38104440

RESUMO

Clinically, Porcine circovirus type 2 (PCV2) often causes disease through coinfection with other bacterial pathogens, including Glaesserella parasuis (G. parasuis), which causes high morbidity and mortality. However, the mechanism of PCV2 and G. parasuis serotype 4 (GPS4) co-infection is still not fully understood. In this study, swine tracheal epithelial cells (STEC) were used as a barrier model, and our results showed that PCV2 infection increased the adhesion of GPS4 to STEC, while decreasing the levels of ZO-1, Occludin and increasing tracheal epithelial permeability, and ultimately facilitated GPS4 translocation. Snail1 is a transcriptional repressor, and has been known to induce epithelial-to-mesenchymal transition (EMT) during development or in cancer metastasis. Importantly, we found that Snail1, as a transcriptional repressor, was crucial in destroying the tracheal epithelial barrier induced by PCV2, GPS4, PCV2 and GPS4 coinfection. For the first time, we found that PCV2, GPS4, PCV2 and GPS4 coinfection cross-activates TGF-ß and p38/MAPK signaling pathways to upregulate the expression of Snail1, down-regulate the levels of ZO-1 and Occludin, and thus disrupt the integrity of tracheal epithelial barrier then promoting GPS4 translocation. Finally, PCV2 and GPS4 co-infection also can activate TGF-ß and p38/MAPK signaling pathways in vivo and upregulate Snail1, ultimately down-regulating the expression of ZO-1 and Occludin. Our study elucidates how PCV2 infection promotes GPS4 to breach the tracheal epithelial barrier and aggravate clinical manifestations.


Assuntos
Infecções por Circoviridae , Circovirus , Coinfecção , Doenças dos Suínos , Suínos , Animais , Circovirus/fisiologia , Coinfecção/microbiologia , Coinfecção/veterinária , Ocludina , Sorogrupo , Junções Intercelulares/patologia , Fator de Crescimento Transformador beta , Epitélio/patologia , Infecções por Circoviridae/veterinária
2.
Vet Res ; 54(1): 7, 2023 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-36717839

RESUMO

Streptococcus suis serotype 2 (SS2) frequently colonizes the swine upper respiratory tract and can cause Streptococcal disease in swine with clinical manifestations of pneumonia, meningitis, and septicemia. Previously, we have shown that vimentin, a kind of intermediate filament protein, is involved in the penetration of SS2 through the tracheal epithelial barrier. The initiation of invasive disease is closely related to SS2-induced excessive local inflammation; however, the role of vimentin in airway epithelial inflammation remains unclear. Here, we show that vimentin deficient mice exhibit attenuated lung injury, diminished production of proinflammatory cytokines interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and the IL-8 homolog, keratinocyte-derived chemokine (KC), and substantially reduced neutrophils in the lungs following intranasal infection with SS2. We also found that swine tracheal epithelial cells (STEC) without vimentin show decreased transcription of IL-6, TNF-α, and IL-8. SS2 infection caused reassembly of vimentin in STEC, and pharmacological disruption of vimentin filaments prevented the transcription of those proinflammatory cytokines. Furthermore, deficiency of vimentin failed to increase the transcription of nucleotide oligomerization domain protein 2 (NOD2), which is known to interact with vimentin, and the phosphorylation of NF-κB protein p65. This study provides insights into how vimentin promotes excessive airway inflammation, thereby exacerbating airway injury and SS2-induced systemic infection.


Assuntos
Infecções Estreptocócicas , Streptococcus suis , Doenças dos Suínos , Animais , Camundongos , Citocinas/genética , Epitélio/patologia , Inflamação/veterinária , Interleucina-6 , Interleucina-8 , Filamentos Intermediários/patologia , Infiltração de Neutrófilos , Sorogrupo , Infecções Estreptocócicas/veterinária , Infecções Estreptocócicas/patologia , Suínos , Traqueia/patologia , Fator de Necrose Tumoral alfa , Vimentina/genética
3.
Vet Res ; 52(1): 135, 2021 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-34674760

RESUMO

Glaesserella parasuis (G. parasuis) is a commensal bacterium in the upper respiratory tract of pigs that can also cause the swine Glässer disease, which induces an intensive inflammatory response and results in significant economic losses to the swine industry worldwide. G. parasuis can cause disease through infection of the respiratory tract, resulting in systemic infection, but the mechanism is largely unknown. Recently we showed that Glaesserella parasuis serotype 4 (GPS4) increased swine tracheal epithelial barrier permeability, resulting in easier bacterial translocation. Tight junction proteins (TJ) play a crucial role in maintaining the integrity and impermeability of the epithelial barrier. GPS4 decreased the expression of the TJ ZO-1 and occludin in swine tracheal epithelial cells (STEC). Furthermore, the proinflammatory cytokines IL-6, IL-8 and TNF-α were significantly upregulated in GPS4-infected STEC, and both the MAPK and NF-κB signaling pathways were activated and contributed to the expression of TNF-α. We demonstrate that the production of proinflammatory cytokines, especially TNF-α, during GPS4 infection was involved in barrier dysfunction. Additionally, animal challenge experiments confirmed that GPS4 infection downregulated TJ in the lungs of piglets and induced a severe inflammatory response. In general, G. parasuis infection downregulated the expression of TJ and induced massive secretion of proinflammatory cytokines, resulting in epithelial barrier disruption and favoring bacterial infection. This study allowed us to better understand the mechanism by which G. parasuis crosses the respiratory tract of pigs.


Assuntos
Translocação Bacteriana , Haemophilus parasuis/fisiologia , Infecções por Pasteurellaceae/veterinária , Transdução de Sinais , Doenças dos Suínos/microbiologia , Animais , Células Epiteliais , Infecções por Haemophilus/microbiologia , Infecções por Haemophilus/fisiopatologia , Infecções por Haemophilus/veterinária , Haemophilus parasuis/genética , Infecções por Pasteurellaceae/microbiologia , Infecções por Pasteurellaceae/fisiopatologia , Sorogrupo , Sus scrofa , Suínos , Doenças dos Suínos/fisiopatologia
4.
Vet Microbiol ; 244: 108684, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32402351

RESUMO

Porcine epidemic diarrhea virus (PEDV) has caused, and continues to cause, severe economic losses to the swine industry worldwide. The pathogenic mechanism and immune regulatory interactions between PEDV and the host remain largely unknown. In this study, the interaction between autophagy and PEDV replication in intestinal porcine epithelial (IPEC-J2) cells was investigated. The effects of the structural and nonstructural proteins of PEDV on the autophagy process and the autophagy-related signaling pathways were also examined. The results shown that PEDV replication increased the autophagy flux in IPEC-J2 cells, and that autophagy was beneficial to PEDV replication, which may be one of the reasons for the rapid damage to intestinal epithelial cells and the enhanced virulence of PEDV in both newborn piglets and finishing pigs. When autophagy was pharmacologically induced by rapamycin, PEDV replication increased from 8.5 × 105 TCID50/mL to 8.8 × 106 TCID50/mL in IPEC-J2 cells. When autophagy was pharmacologically suppressed by hydroxychloroquine, PEDV replication decreased from 8.5 × 105 TCID50/mL to 7.9 × 104 TCID50/mL. To identify which PEDV proteins were the key inducers of autophagy, all 4 structural proteins and 17 nonstructural proteins of PEDV were eukaryotic expressed. It was found that the nonstructural protein 6 (nsp6) and ORF3 of PEDV were able to induce significant autophagy in IPEC-J2 cells, but the other proteins were unable to induce autophagy. It was indicated that nsp6-induced autophagy mainly occurred via the PI3K/Akt/mTOR signaling pathway. The results accelerate the understanding of the biology and pathogenesis of PEDV infection and provide new insights into the development of effective therapeutic strategies.


Assuntos
Autofagia , Vírus da Diarreia Epidêmica Suína/patogenicidade , Transdução de Sinais , Proteínas não Estruturais Virais/genética , Replicação Viral , Animais , Linhagem Celular , Células Epiteliais/patologia , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Intestinos/citologia , Intestinos/virologia , Proteína Oncogênica v-akt/genética , Proteína Oncogênica v-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Vírus da Diarreia Epidêmica Suína/fisiologia , Suínos , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Proteínas não Estruturais Virais/metabolismo
5.
Vet Res ; 51(1): 31, 2020 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-32106883

RESUMO

Porcine circovirus type 2 (PCV2) is considered as the primary pathogen of porcine circovirus-associated disease (PCVAD), which results in significant economic losses worldwide. Clinically, PCV2 often causes disease through coinfection with other bacterial pathogens, including Streptococcus suis (S. suis), and especially the highly prevalent S. suis serotype 2 (SS2). The present study determined that continuous PCV2 infection in piglets down-regulates tight junction proteins (TJ) ZO-1 and occludin in the lungs. Swine tracheal epithelial cells (STEC) were used to explore the mechanisms and consequences of disruption of TJ, and an in vitro tracheal epithelial barrier model was established. Our results show that PCV2 infection in STEC decreases the expression levels of ZO-1 and occludin and increases the permeability of the tracheal epithelial barrier, resulting in easier translocation of SS2. Moreover, Western blot analysis indicates that PCV2 infection activates the JNK/MAPK pathway. The disruption of TJ in SETC and increased permeability of the epithelial barrier induced by PCV2 could be alleviated by inhibition of JNK phosphorylation, which indicates that the JNK/MAPK pathway regulates the expression of ZO-1 and occludin during PCV2 infection. This study allows us to better understand the mechanisms of PCV2 coinfection with bacterial pathogens and provides new insight into controlling the occurrence of PCVAD.


Assuntos
Infecções por Circoviridae/veterinária , Circovirus/fisiologia , Coinfecção/veterinária , Transdução de Sinais , Infecções Estreptocócicas/veterinária , Streptococcus suis/fisiologia , Doenças dos Suínos/microbiologia , Animais , Linhagem Celular , Infecções por Circoviridae/virologia , Coinfecção/microbiologia , Coinfecção/virologia , Células Epiteliais/microbiologia , Células Epiteliais/virologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Infecções Estreptocócicas/microbiologia , Suínos , Doenças dos Suínos/virologia , Junções Íntimas , Traqueia/microbiologia , Traqueia/virologia
6.
J Proteome Res ; 16(6): 2113-2120, 2017 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-28506058

RESUMO

Porcine epidemic diarrhea (PED) re-emerged in China in late 2010 and has now become widespread. Accumulated evidence indicates that this large-scale outbreak of diarrhea was caused by variants of the highly virulent porcine epidemic diarrhea virus (PEDV). A pandemic PEDV YC2014 strain (YC2014) was isolated from clinical samples. An iTRAQ-based comparative quantitative proteomic study of IPEC-J2 cells infected with YC2014 and a classical CV777 strain (CV777) was performed to determine the differences between pandemic and classical PEDV strain infection. Totals of 353 and 299 differentially expressed proteins were identified upon YC2014 and CV777 infection, respectively. The canonical pathways and functional networks involved in both PEDV infections were analyzed. The results indicated that the PEDV suppressed protein synthesis of IPEC-J2 cells through down-regulation of the PI3K-AKT/mTOR signaling pathways. Infection with YC2014 could activate the JAK-STAT signaling pathway and the NF-κB pathway more intensively than CV777. YC2014 could activate NF-κB pathway more intensively than CV777. On the basis of differentially expressed proteins, we propose that PEDV might disrupt apoptosis and may elicit stronger inflammatory cascades as well. This study might contribute to an understanding of the pathogenesis of PEDV infection and aid in the development of effective preventive and control vaccines.


Assuntos
Vírus da Diarreia Epidêmica Suína/patogenicidade , Proteínas/análise , Proteômica/métodos , Transdução de Sinais , Animais , Apoptose , Linhagem Celular , Chlorocebus aethiops , Infecções por Coronavirus/etiologia , Infecções por Coronavirus/metabolismo , Regulação para Baixo , Inflamação/metabolismo , Janus Quinases/metabolismo , NF-kappa B/metabolismo , Proteína Oncogênica v-akt/metabolismo , Pandemias , Fosfatidilinositol 3-Quinases/metabolismo , Vírus da Diarreia Epidêmica Suína/metabolismo , Fatores de Transcrição STAT/metabolismo , Suínos , Serina-Treonina Quinases TOR/metabolismo
7.
Artigo em Inglês | MEDLINE | ID: mdl-28373968

RESUMO

Invasive infections caused by Streptococcus suis serotype 2 (SS2) has emerged as a clinical problem in recent years. Neutrophil extracellular traps (NETs) are an important mechanism for the trapping and killing of pathogens that are resistant to phagocytosis. Biofilm formation can protect bacteria from being killed by phagocytes. Until now, there have only been a few studies that focused on the interactions between bacterial biofilms and NETs. SS2 in both a biofilm state and a planktonic cell state were incubated with phagocytes and NETs, and bacterial survival was assessed. DNase I and cytochalasin B were used to degrade NET DNA or suppress phagocytosis, respectively. Extracellular DNA was stained with impermeable fluorescent dye to quantify NET formation. Biofilm formation increased up to 6-fold in the presence of neutrophils, and biofilms were identified in murine tissue. Both planktonic and biofilm cells induced neutrophils chemotaxis to the infection site, with neutrophils increasing by 85.1 and 73.8%, respectively. The bacteria in biofilms were not phagocytized. The bactericidal efficacy of NETs on the biofilms and planktonic cells were equal; however, the biofilm extracellular matrix can inhibit NET release. Although biofilms inhibit NETs release, NETs appear to be an important mechanism to eliminate SS2 biofilms. This knowledge advances the understanding of biofilms and may aid in the development of treatments for persistent infections with a biofilm component.


Assuntos
Biofilmes/crescimento & desenvolvimento , Armadilhas Extracelulares/metabolismo , Interações Hospedeiro-Patógeno , Neutrófilos/imunologia , Sorogrupo , Streptococcus suis/imunologia , Streptococcus suis/fisiologia , Animais , Células Cultivadas , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Viabilidade Microbiana , Neutrófilos/microbiologia , Fagocitose
8.
Sci Rep ; 7: 43990, 2017 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-28272485

RESUMO

To characterize neutralizing mimotopes, phages were selected from a 12-mer phage display library using three anti-porcine reproductive and respiratory syndrome virus (PRRSV) neutralizing monoclonal antibodies: (1) A1; (2) A2; and (3) A7. Of these, A2 and A7 recognize the mimotope, P2, which contains the SRHDHIH motif, which has conserved consensus sequences from amino acid positions 156 to 161 in the N-terminal ectodomain of GP3. The artificial multi-epitope gene, mp2, was designed by combining three repeats of the mimotope P2. The resulting sequence was inserted into the swinepox virus (SPV) genome to construct a recombinant swinepox virus (rSPV-mp2). The rSPV-mp2 was able to stably express the multi-epitope peptide, mP2, in vitro. The rSPV-mp2 immunized pigs exhibited a significantly shorter fever duration compared with the wtSPV treated group (P < 0.05). There was an enhanced humoral and cellular immune response, decreased number of PRRSV genomic copies, and a significant reduction in the gross lung pathology (P < 0.05) was observed following PRRSV infection in rSPV-mp2-immunized animals. The results suggest that the recombinant rSPV-mp2 provided pigs with significant protection against PRRSV infection.


Assuntos
Epitopos/genética , Peptídeos/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/metabolismo , Suipoxvirus/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Temperatura Corporal , Epitopos/química , Epitopos/imunologia , Interferon gama/sangue , Interleucina-4/sangue , Pulmão/patologia , Biblioteca de Peptídeos , Peptídeos/química , Peptídeos/imunologia , Síndrome Respiratória e Reprodutiva Suína/metabolismo , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , RNA Viral/sangue , Suipoxvirus/genética , Suipoxvirus/metabolismo , Suínos , Vacinas Virais/genética , Vacinas Virais/imunologia , Vacinas Virais/metabolismo
9.
Res Microbiol ; 167(5): 403-12, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27068185

RESUMO

The arginine deiminase system (ADS) is a secondary metabolic system found in many different bacterial pathogens and it is often associated with virulence. Here, a systematic study of ADS functions in Streptococcus equi subsp. zooepidemicus (SEZ) was performed. Transcriptional levels of ADS operon genes were observed to be significantly increased when SEZ was grown under acidic conditions. We constructed arcA and arcD deletion mutants (SEZ ΔarcA and SEZ ΔarcD, respectively) and found that SEZ ΔarcA was unable to metabolize arginine and synthesize ammonia; however, arcD deletion resulted in an initial decrease in arginine consumption and ammonia production, followed by recovery to the levels of wild-type SEZ after 24 h of cultivation. Cell extracts of SEZ ΔarcA showed no arginine deiminase (AD) activity, whereas no difference in AD activity between SEZ ΔarcD and wild-type SEZ was observed. SEZ survival tests demonstrated a significant decrease in survival for SEZ ΔarcA, when compared with wild-type SEZ, under acidic conditions and in epithelial cells. These findings indicate that ADS in SEZ contributes to environmental adaptability via ammonia synthesis to reduce pH stress.


Assuntos
Adaptação Fisiológica , Hidrolases/metabolismo , Streptococcus equi/enzimologia , Streptococcus equi/fisiologia , Amônia/metabolismo , Arginina/metabolismo , Linhagem Celular , Células Epiteliais/microbiologia , Deleção de Genes , Perfilação da Expressão Gênica , Humanos , Concentração de Íons de Hidrogênio , Hidrolases/genética , Viabilidade Microbiana , Streptococcus equi/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA