Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Neurobiol ; 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38625620

RESUMO

Although naturally Streptococcus suis serotype 2 (SS2) causes meningitis resulting in death or sequela of neurological symptoms in pigs and humans, severely threatening public health in the world, it has been difficult to build up and confirm experimental meningitis mouse models with obvious neurological syndrome for about two decades, which strongly hampers the in-depth study on the control measures and mechanisms of SS2-induced meningitis. In this study, a typical meningitis mouse model of SS2 was successfully established, as confirmed by the behavioral indicators of balance beam test, suspension test, and gait analysis. With bacteria gathering in the brain, distinguishable unique features including meningeal thickening, vacuolization of the Nissl body, brain barrier damage, glial cell activation, and more infiltration of T cells, macrophages, and DCs are observed in SS2 meningitis mice with typical neurological signs. Some meningitis mice were also accompanied by identical nephritis, ophthalmia, and cochlearitis. Investigation of the metabolic features demonstrated the downregulated cholic acid and upregulated 2-hydroxyvaleric acid, tetrahydrocortisone, nicotinic acid, and lauric acid in blood serum of mice and piglets with meningitis. And feeding trials show that lauric acid can promote meningitis by promoting the infiltration of immune cells into brain. These findings demonstrated that infection of ICR (improved castle road) mice with SS2 was able to induce typical meningitis accompanied by immune cell infiltration and lauric acid upregulation. These data provide a basis for the deep study of SS2 meningitis.

2.
BMC Biol ; 22(1): 33, 2024 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-38331785

RESUMO

BACKGROUND: Ribosomal protein SA (RPSA) of human brain microvascular endothelial cells (HBMECs) can transfer from the cytosol to the cell surface and act as a receptor for some pathogens, including Streptococcus suis serotype 2 (SS2), a zoonotic pathogen causing meningitis in pigs and humans. We previously reported that SS2 virulence factor enolase (ENO) binds to RPSA on the cell surface of HBMECs and induces apoptosis. However, the mechanism that activates RPSA translocation to the cell surface and induces ENO-mediated HBMEC apoptosis is unclear. RESULTS: Here, we show that RPSA localization and condensation on the host cell surface depend on its internally disordered region (IDR). ENO binds to the IDR of RPSA and promotes its interaction with RPSA and vimentin (VIM), which is significantly suppressed after 1,6-Hexanediol (1,6-Hex, a widely used tool to disrupt phase separation) treatment, indicating that ENO incorporation and thus the concentration of RPSA/VIM complexes via co-condensation. Furthermore, increasing intracellular calcium ions (Ca2+) in response to SS2 infection further facilitates the liquid-like condensation of RPSA and aggravates ENO-induced HBMEC cell apoptosis. CONCLUSIONS: Together, our study provides a previously underappreciated molecular mechanism illuminating that ENO-induced RPSA condensation activates the migration of RPSA to the bacterial cell surface and stimulates SS2-infected HBMEC death and, potentially, disease progression. This study offers a fresh avenue for investigation into the mechanism by which other harmful bacteria infect hosts via cell surfaces' RPSA.


Assuntos
Infecções Estreptocócicas , Streptococcus suis , Humanos , Animais , Suínos , Células Endoteliais/metabolismo , Sorogrupo , Fosfopiruvato Hidratase/genética , Fosfopiruvato Hidratase/metabolismo , Encéfalo/metabolismo , Apoptose , Proteínas Ribossômicas/metabolismo , Infecções Estreptocócicas/metabolismo , Infecções Estreptocócicas/microbiologia
3.
Vet Microbiol ; 290: 109973, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38211361

RESUMO

Newcastle disease virus (NDV) is an RNA virus that can promote its own replication through the inhibition of cellular mitochondrial fusion. The proteins involved in mitochondrial fusion, namely mitofusin 1 (Mfn1) and optic atrophy 1 (OPA1) are associated with interferon-beta (IFN-ß) secretion during NDV infection. However, the precise mechanism by which NDV modulates the Mfn1-mediated or OPA1-mediated fusion of mitochondria, thereby impacting IFN-ß, remains elusive. This study revealed that the downregulation of the mitochondrial protein known as coiled-coil-helix-coiled-coil-helix domain containing 10 (CHCHD10) exerts a negative regulatory effect on OPA1 and Mfn1 in human lung adenocarcinoma (A549) cells during the late stage of NDV infection. This reduction in CHCHD10 expression impeded cellular mitochondrial fusion, subsequently leading to a decline in the activation of interferon regulatory factor 3 (IRF3) and nuclear factor kappa B (NF-κB), ultimately resulting in diminished secretion of IFN-ß. In contrast, the overexpression of CHCHD10 alleviated infection-induced detrimental effect in mitochondrial fusion, thereby impeding viral proliferation. In summary, NDV enhances its replication by inhibiting the CHCHD10 protein, which impedes mitochondrial fusion and suppresses IFN-ß production through the activation of IRF3 and NF-κB.


Assuntos
NF-kappa B , Vírus da Doença de Newcastle , Humanos , Animais , Vírus da Doença de Newcastle/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Dinâmica Mitocondrial , Interferon beta/genética , Interferon beta/metabolismo , Proliferação de Células , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo
4.
Vet Microbiol ; 290: 109986, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38244394

RESUMO

Newcastle disease (ND) is a disease that threatens the world's poultry industry, which is caused by virulent Newcastle disease virus (NDV). As its pathogenic mechanism remains not fully clear, the proteomics of NDV-infected cells were analyzed. The results revealed that coiled-coil-helix-coiled-coil-helix domain containing 10 (CHCHD10) protein displayed a significant decrease at the late stage of NDV infection. To investigate the function of CHCHD10 in NDV infection, its expression after NDV infection was detected both in vivo and in vitro. Besides, the tissue viral loads and pathological damage of C57BL/6 mice with CHCHD10 differently expressed were also investigated. The results showed that the CHCHD10 expression was significantly decreased both in vivo and in vitro at the late stage of NDV infection. The viral loads were significantly higher in CHCHD10 silenced C57BL/6 mice, along with more severe pathological damage and vice versa.


Assuntos
Doença de Newcastle , Doenças das Aves Domésticas , Doenças dos Roedores , Camundongos , Animais , Vírus da Doença de Newcastle/genética , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Camundongos Endogâmicos C57BL , Aves Domésticas , Galinhas
5.
Vet Microbiol ; 288: 109943, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38113574

RESUMO

O-acetyl-homoserine sulfhydrylase (OAHS) is a pyridoxal 5'-phosphate-dependent enzyme involved in microbial methionine biosynthesis, which catalyzes the conversion of o-acetyl-homoserine (OAH) to homocysteine. In our previous study, we found that OAHS of Streptococcus suis serotype 2 (SS2) can interact with the porcine blood-brain barrier (BBB) model, but whether OAHS regulates the penetration of BBB during SS2 infection is still unclear. To explore the role of OAHS in SS2 infection, OAHS-deficient SS2 mutant strain (SC19-ΔOAHS) and gene complemental strain (SC19-cΔOAHS) were constructed. Compared to the parent strain, with the loss of oahs, the chain length of SC19-ΔOAHS was shortened, the virulence was significantly reduced, the survival rate of mice infected with SC19-ΔOAHS was obviously increased accompanied by the relieved clinical symptoms. And the survival ability of SC19-ΔOAHS in whole blood was also remarkably decreased. Interestingly, the adhesion of SC19-ΔOAHS to endothelial cells was markedly increased, but the deficiency of OAHS significantly inhibited the strain penetrating BBB both in vivo and in vitro. Most of these phenomena can be reversed by the complemental strain (SC19-cΔOAHS). Further study showed that the deficiency of OAHS severely reduced SC19-induced endothelial cell apoptosis, tight junctions (TJs) protein impairment and the expression of SS2 virulence factor Enolase (Eno), involved in the destruction of BBB. Additionally, SC19-ΔOAHS immunized mice were able to resist SC19 or JZLQ022 infection. In conclusion, we confirmed that OAHS promoted the pathogenicity by enhancing host's BBB permeability and immune escape, and SC19- ΔOAHS is a potential live vaccine.


Assuntos
Infecções Estreptocócicas , Streptococcus suis , Doenças dos Suínos , Animais , Camundongos , Células Endoteliais , Homosserina/genética , Sorogrupo , Infecções Estreptocócicas/veterinária , Suínos , Doenças dos Suínos/metabolismo , Virulência
6.
Cells ; 12(5)2023 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-36899832

RESUMO

Actinobacillus pleuropneumoniae (A. pleuropneumoniae) causes porcine pleuropneumonia that seriously endangers pig's health. Adh, located in the head region of trimeric autotransporter adhesion of A. pleuropneumoniae, affects bacterial adhesion and pathogenicity. However, how Adh mediates A. pleuropneumoniae immune invasion is still unclear. Here, we established the A. pleuropneumoniae strain L20 or L20 ΔAdh-infected porcine alveolar macrophages (PAM) model, and applied protein overexpression, RNA interference, qRT-PCR, Western blot and immunoflourescence techniques to dissect the effects of Adh on PAM during A. pleuropneumoniae infection. We found that Adh could increase the A. pleuropneumoniae adhesion and intracellular survival in PAM. Gene chip analysis of piglet lungs further showed that Adh significantly induced cation transport regulatory-like protein 2 (CHAC2) expression, whose overexpression suppressed the phagocytic capacity of PAM. Furthermore, CHAC2 overexpression dramatically increased glutathione (GSH) expression, decreased reactive oxygen species (ROS), and promoted A. pleuropneumoniae survival in PAM, while the knockdown of CHAC2 reversed these phenomena. Meanwhile, CHAC2 silence activated the NOD1/NF-κB pathway, resulting in an increase in IL-1ß, IL-6, and TNF-α expression, whereas this effect was weakened by CHAC2 overexpression and addition of NOD1/NF-κB inhibitor ML130. Moreover, Adh enhanced the secretion of LPS of A. pleuropneumoniae, which regulated the expression of CHAC2 via TLR4. In conclusion, through a LPS-TLR4-CHAC2 pathway, Adh inhibits respiratory burst and inflammatory cytokines expression to promote A. pleuropneumoniae survival in PAM. This finding may provide a novel target for the prevention and treatment of A. pleuropneumoniae.


Assuntos
Actinobacillus pleuropneumoniae , Citocinas , Suínos , Animais , Citocinas/metabolismo , Macrófagos Alveolares/metabolismo , Actinobacillus pleuropneumoniae/genética , NF-kappa B/metabolismo , Explosão Respiratória , Lipopolissacarídeos/metabolismo , Receptor 4 Toll-Like/metabolismo
7.
Front Microbiol ; 14: 1329609, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38260894

RESUMO

Introduction: Klebsiella pneumoniae (K. pneumoniae) is an important opportunistic and zoonotic pathogen which is associated with many diseases in humans and animals. However, the pathogenicity of K. pneumoniae has been neglected and the prevalence of K. pneumoniae is poorly studied due to the lack of rapid and sensitive diagnosis techniques. Methods: In this study, we infected mice and pigs with K. pneumoniae strain from a human patient. An indirect ELISA was established using the KHE protein as the coating protein for the detection of K. pneumoniae specific antibody in clinical samples. A nested PCR method to detect nuclei acids of K. pneumoniae was also developed. Results: We showed that infection with K. pneumoniae strain from a human patient led to mild lung injury of pigs. For the ELISA, the optimal coating concentration of KHE protein was 10 µg/mL. The optimal dilutions of serum samples and secondary antibody were 1:100 and 1:2500, respectively. The analytical sensitivity was 1:800, with no cross-reaction between the coated antigen and porcine serum positive for antibodies against other bacteria. The intra-assay and inter-assay reproducibility coefficients of variation are less than 10%. Detection of 920 clinical porcine serum samples revealed a high K. pneumoniae infection rate by established indirect ELISA (27.28%) and nested PCR (19.13%). Moreover, correlation analysis demonstrated infection rate is positively correlated with gross population, Gross Domestic Product (GDP), and domestic tourists. Discussion: In conclusion, K. pneumoniae is highly prevalent among pigs in China. Our study highlights the role of K. pneumoniae in pig health, which provides a reference for the prevention and control of diseases associated with K. pneumoniae.

8.
Cells ; 11(13)2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35805155

RESUMO

Streptococcus suis serovar 2 (S. suis serovar 2) is a zoonotic pathogen that causes meningitis in pigs and humans, and is a serious threat to the swine industry and public health. Understanding the mechanism(s) by which S. suis serovar 2 penetrates the blood-brain barrier (BBB) is crucial to elucidating the pathogenesis of meningitis. In a previous study, we found that expression of the virulence factor enolase (Eno) by S. suis serovar 2 promotes the expression of host heat shock protein family D member 1 (HSPD1) in brain tissue, which leads to the apoptosis of porcine brain microvascular endothelial cells (PBMECs) and increased BBB permeability, which in turn promotes bacterial translocation across the BBB. However, the mechanism by which HSPD1 mediates Eno-induced apoptosis remains unclear. In this study, we demonstrate that Eno promotes the translocation of HSPD1 from mitochondria to the cytoplasm, where HSPD1 binds to ß-actin (ACTB), the translocated HSPD1, and its interaction with ACTB led to adverse changes in cell morphology and promoted the expression of apoptosis-related proteins, second mitochondria-derived activator of caspases (Smac), and cleaved caspase-3; inhibited the expression of X-linked inhibitor of apoptosis protein (XIAP); and finally promoted cell apoptosis. These results further elucidate the role of HSPD1 in the process of Eno-induced apoptosis and increased BBB permeability, increasing our understanding of the pathogenic mechanisms of meningitis, and providing a framework for novel therapeutic strategies.


Assuntos
Meningite , Streptococcus suis , Animais , Apoptose , Barreira Hematoencefálica , Chaperonina 60 , Citoplasma , Células Endoteliais , Mitocôndrias , Fosfopiruvato Hidratase , Sorogrupo , Suínos
9.
AMB Express ; 11(1): 175, 2021 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-34952961

RESUMO

Porcine infectious pleuropneumonia is characterized by a high-rate of carriage and mixed infection with other pathogens. The host immune response induced by Actinobacillus pleuropneumoniae (APP) is the basis for elucidating pathogenesis and controlling disease. However, there is currently no comprehensive and dynamic data characterising the host immune response. In this study, piglets were infected with APP and differentially expressed proteins of bronchoalveolar lavage fluid (BALF) and peripheral serum were identified by iTRAQ-LC-MS/MS, and differentially expressed genes of peripheral blood mononuclear cells (PBMC) by RNA-seq. The results of the integrated analysis of serum, BALF and PBMC showed significant metabolism and local immune responses in BALF, the general immune response in PBMC mainly involves cytokines, while that in serum mainly involves biosynthesis, phagosome, and complement and coagulation cascades. Furthermore, immune responses in PBMCs and serum were rapid and maintained compared to the lung where metabolism and cell adhesion activities were enriched. Some innate immunity pathways of the cellular response to ROS, neutrophil mediated immunity, granulocyte activation and leukocyte cell-cell adhesion were identified as central points, connecting multiple signaling pathways to form an integrated large network. At 24 h post-infection, 14 molecules were up regulated in BALF, 10 of which were shared with PBMC, but at 120 h, 20 down-regulated molecules were identified in BALF, 11 of them still up- regulated in PBMC. We conclude that, the immune response in the lung is different from that in blood, but there is a similarity in response in PBMC and serum.

10.
Infect Immun ; 89(6)2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-33685942

RESUMO

Porcine pleuropneumonia is a common infectious disease of pigs caused by Actinobacillus pleuropneumoniae Interferon gamma (IFN-γ) expression increases in the lung of pigs after A. pleuropneumoniae infection, but the role of IFN-γ during the infection is still obscure. In this study, an IFN-γ-/- mouse infection model was established, and bacterial load, levels of inflammatory cytokines, and types of neutrophils in the lungs were studied at different times post-A. pleuropneumoniae infection. We found that wild-type (WT) mice were more susceptible to A. pleuropneumoniae than IFN-γ-/- mice. At 6 h postinfection (hpi), the expression of interleukin 18 (IL-18) and IL-1ß in the lungs of IFN-γ-/- mice was significantly increased compared to WT mice. The bacterial load and levels of inflammatory cytokines (IL-1ß and IL-6) of IFN-γ-/- mice were significantly reduced at 12 hpi compared to WT mice. After an initial loss, the numbers of lung polymorphonuclear (PMN)-I cells dramatically increased in the lungs of IFN-γ-/- but not WT mice, whereas PMN-II cells continually decreased. Finally, in vivo administration of IL-18 significantly reduced clinical scores and bacterial load in the lungs of A. pleuropneumoniae-infected mice. This study identifies IFN-γ as a target for regulating the inflammatory response in the lung and provides a basis for understanding the course of clinical bacterial pneumonia and for the formulation of treatment protocols.


Assuntos
Infecções por Actinobacillus/imunologia , Infecções por Actinobacillus/metabolismo , Actinobacillus pleuropneumoniae/imunologia , Interações Hospedeiro-Patógeno , Interleucina-18/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Infecções por Actinobacillus/microbiologia , Infecções por Actinobacillus/patologia , Animais , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Pulmão/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Knockout , Infiltração de Neutrófilos , Neutrófilos/patologia
11.
Vet Res ; 52(1): 30, 2021 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-33618766

RESUMO

Host proteins interacting with pathogens are receiving more attention as potential therapeutic targets in molecular medicine. Streptococcus suis serotype 2 (SS2) is an important cause of meningitis in both humans and pigs worldwide. SS2 Enolase (Eno) has previously been identified as a virulence factor with a role in altering blood brain barrier (BBB) integrity, but the host cell membrane receptor of Eno and The mechanism(s) involved are unclear. This study identified that SS2 Eno binds to 40S ribosomal protein SA (RPSA) on the surface of porcine brain microvascular endothelial cells leading to activation of intracellular p38/ERK-eIF4E signalling, which promotes intracellular expression of HSPD1 (heat-shock protein family D member 1), and initiation of host-cell apoptosis, and increased BBB permeability facilitating bacterial invasion. This study reveals novel functions for the host-interactional molecules RPSA and HSPD1 in BBB integrity, and provides insight for new therapeutic strategies in meningitis.


Assuntos
Barreira Hematoencefálica , Células Endoteliais/metabolismo , Fosfopiruvato Hidratase/metabolismo , Proteínas Ribossômicas/metabolismo , Infecções Estreptocócicas/veterinária , Streptococcus suis/metabolismo , Animais , Apoptose , Técnicas de Cocultura , Células Endoteliais/microbiologia , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Camundongos , Ligação Proteica , Sorogrupo , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Streptococcus suis/patogenicidade , Suínos , Doenças dos Suínos/microbiologia , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Vet Microbiol ; 254: 108981, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33445055

RESUMO

Streptococcus suis serotype 2 (SS2) is an important zoonotic pathogen that causes meningitis. The ubiquitously expressed 40S ribosome protein SA (RPSA) is a multifunctional protein involved in the pathogenesis of multiple pathogens, especially those causing meningitis. However, the role of RPSA in SS2-induced meningitis is not clear. In this study, immunofluorescence staining revealed that SS2 infection promoted the intracellular transfer of RPSA to the surface of human cerebral microvascular endothelial cells (HCMECs). Moreover, SS2 infection promoted the accumulation of caveolin 1 (CAV1) and the formation of membrane bulges where RPSA enveloped CAV1 on the cell surface. SS2 infection also caused dynamic changes in the localization of RPSA and CAV1 on the cell surface which could be eliminated by disruption of caveolae/rafts by addition of methyl-ß-cyclodextrin (MßCD). Co-immunoprecipitation analysis demonstrated that α-enolase (ENO), a key virulence factor of SS2, interacted with RPSA, and promoted the interaction between RPSA and CAV1. Immunofluorescence staining, western blotting and flow cytometry analyses showed that damaged caveolae/rafts significantly enhanced ENO adhesion to HCMECs, promoted the "destruction" of RPSA by ENO, and enhanced the toxic effect of ENO on HCMECs. Importantly, these effects could be relieved upon the addition of cholesterol. We conclude that caveolae/rafts weaken the toxic effect of SS2 ENO on RPSA-mediated events in HCMECs. Our study has led to better understanding of the roles of RPSA and caveolae/rafts upon SS2 infection, and a new pathological role for RPSA in infection.


Assuntos
Cavéolas/metabolismo , Caveolina 1/metabolismo , Células Endoteliais/microbiologia , Fosfopiruvato Hidratase/metabolismo , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Streptococcus suis/patogenicidade , Animais , Linhagem Celular , Imunofluorescência , Células HEK293 , Humanos , Fosfopiruvato Hidratase/genética , Subunidades Ribossômicas Menores de Eucariotos/genética , Sorogrupo , Streptococcus suis/classificação , Streptococcus suis/enzimologia , Fatores de Virulência
13.
Microb Pathog ; 152: 104609, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33217534

RESUMO

AIMS: 40S ribosomal protein SA (RPSA), a component of the small ribosomal subunit, is a high-affinity receptor of laminin that is widely expressed in cells and involves in many biological processes. However, it hasn't been reported which tissues and cells may be targeted by RPSA-mediated pathogen regulation. Therefore, in this study, a gram-positive bacterium Streptococcus suis Type 2 (SS2), gram-negative bacterium Actinobacillus pleuropneumoniae (A.pleuropneumoniae), and porcine circovirus Type 2 (PCV2) were used to infect ICR mice. METHODS AND RESULTS: The effects of infection with the three pathogens on expression levels of RPSA in mouse tissues and peripheral blood immune cells were analysed by immunohistochemistry and flow cytometry. The results suggested that the pathological changes in mice infected with SS2 were mainly manifested as congestion and inflammatory infiltration in the meninges, lungs, hearts and livers. The mice infected with A.pleuropneumoniae or PCV2 showed lung lesions and mild hepatocyte degeneration, respectively. In uninfected mice, RPSA protein was expressed to various degrees in all tissues except the spleen. After SS2 infection for 3 d, the expression of RPSA in the liver and brain increased, while decreased significantly in the heart and duodenum. These results were corroborated on examining the correlation between RPSA expression and the process of SS2 infection, except that there was no significant difference between the expression levels in the heart at 1 d and 3 d. After A.pleuropneumoniae and PCV2 infection for 3 d, the expression of RPSA decreased in the heart, and brain, respectively. Additionally, under physiological conditions, RPSA expression in CD4+ T cells, CD8+ T cells, neutrophils, and macrophages in the peripheral blood of mice was higher than that in B cells and NK cells. After SS2 infection for 3 d, RPSA expression increased significantly in CD4+ T cells and CD8+ T cells but decreased significantly in macrophages. The expression of RPSA after A.pleuropneumoniae and PCV2 infection were similar, and RPSA expression decreased only in macrophages. CONCLUSIONS: The results revealed that RPSA showed different expression levels in tissues and immune cells due to different pathogens causing disease courses, suggesting different target tissues and target cells in RPSA-mediated pathogenesis after infection, which supports the systematic study of the pathogenesis of RPSA in infectious diseases.


Assuntos
Infecções por Circoviridae , Circovirus , Streptococcus suis , Doenças dos Suínos , Animais , Linfócitos T CD8-Positivos , Camundongos , Camundongos Endogâmicos ICR , Proteínas Ribossômicas , Suínos
14.
Vet Microbiol ; 247: 108755, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32686648

RESUMO

Excessive cytokine production is an important component of the acute respiratory distress syndrome and multiple organ failure. Pneumonia can lead to an overexpression of cytokines, although comparatively little is known about the relevance and differences in cytokines between blood and lung. In this study, piglets were experimentally infected intranasally with Actinobacillus pleuropneumoniae (APP), and transcriptomes of lung tissue and peripheral blood mononuclear cells determined. In addition, the levels of 30 cytokines in broncheoalveolar lavage fluid (BALF) and sera were determined by ELISA. Post infection, there was an early increase in lung monocytes, and a later rise in inflammatory cytokines in BALF. Blood lymphocytes increased early in infection and there was a rise in inflammatory cytokines in the peripheral blood of infected piglets. Genes involved in cytokine production, leukocyte migration and differentiation, lymphocyte activation, and cytokine-mediated signaling pathways in the transcriptomes of lung tissue were significantly down-regulated early in infection. At this early phase of APP infection (0-6 h), the cytokines IL-1ß, MCP-1, and IL-5 in sera increased rapidly and significantly, while many cytokines in BALF decreased. At 48 h post-infection, cytokines in sera were no longer significantly increased, although some were up-regulated in BALF, and there was aggravated pathological damage in the lungs at this time. The data indicate there are substantial differences between immune cells and cytokines in the lung and peripheral blood of APP infected piglets at equivalent time points. The results increase our understanding of pig-APP host interactive biology, and will be important in formulating future therapeutic and preventative strategies to prevent disease caused by APP.


Assuntos
Infecções por Actinobacillus/sangue , Infecções por Actinobacillus/veterinária , Imunidade , Pulmão/microbiologia , Sistema Respiratório/microbiologia , Actinobacillus pleuropneumoniae/imunologia , Animais , Líquido da Lavagem Broncoalveolar/microbiologia , Quimiocinas/imunologia , Citocinas/imunologia , Leucócitos Mononucleares/imunologia , Pulmão/patologia , Linfócitos/imunologia , Organismos Livres de Patógenos Específicos , Suínos , Transcriptoma
15.
J Infect Dev Ctries ; 14(5): 502-510, 2020 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-32525837

RESUMO

INTRODUCTION: Streptococcus suis serotype 2 (SS2) is an important zoonotic pathogen that causes meningitis in China. This study's aim was comparative analysis of serum proteomics from meningitis and non-meningitis piglets. METHODOLOGY: SS2 meningitis and non-meningitis piglet models were established. The serum samples were collected and analyzed by label-free LC-MS/MS proteomics technology. Differentially expressed proteins (DEPs) from serum were screened out by comparing the meningitis group and non-meningitis group to the healthy group (M/C; N/C), respectively. And then, globally and comparative analysis of DEPs in "M/C" and "N/C" in serum were performed using bioinformatics method. Finally, we comparatively analyzed the serum and cerebrospinal fluid proteomics in piglets that lived with meningitis. RESULTS: We obtained 316 and 191 DEPs from "M/C" and "N/C" which classification visualizations were established. 157 DEPs were common in both groups and 159 DEPs were unique to the "M/C". These DEPs and the signaling pathways which they participated in were visualized. Moreover, some DEPs which participated in multiple pathways were discovered and the interaction between 159 DEPs was also mapped. 39 common DEPs were also screened out in serum and cerebrospinal fluid during meningitis, and signaling pathways associated with these DEPs were further visualized. CONCLUSIONS: DEPs in "M/C" and "N/C" were comparatively analyzed and the similarities and differences of these DEPS which were involved in signal pathways were summarized. Moreover, several important molecules were screened out.


Assuntos
Meningites Bacterianas/sangue , Meningites Bacterianas/veterinária , Proteômica , Infecções Estreptocócicas/sangue , Infecções Estreptocócicas/veterinária , Fatores Etários , Animais , China , Biologia Computacional , Meningites Bacterianas/líquido cefalorraquidiano , Sorogrupo , Infecções Estreptocócicas/líquido cefalorraquidiano , Streptococcus suis/classificação , Suínos
16.
Front Immunol ; 11: 585399, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33603733

RESUMO

Streptococcus suis serotype 2 (SS2), an important zoonotic pathogen that causes septicemia, arthritis, and irreversible meningitis in pigs and humans, can be transmitted to humans from pigs. S. suis causes huge economic losses to the swine industry and poses a serious threat to public health. Previously, we found that the brain tissues of mice with SS2-induced meningitis showed disrupted structural integrity and significantly enhanced polymorphonuclear neutrophil (PMN) infiltration. We showed that the brain tissues of SS2-infected mice had increased ribosomal protein SA (RPSA)-positive PMN counts. However, the inflammatory responses of RPSA+ PMNs to SS2 and their effects on the blood-brain barrier (BBB) remain unclear. Therefore, in studying the pathogenesis of SS2-induced meningitis, it is essential that we explore the functions of RPSA+ PMNs and their effects on the BBB. Herein, using flow cytometry and immunofluorescence microscopy analyses, we found that RPSA expression enhances PMN-induced phagocytosis and PMN-induced formation of neutrophil extracellular traps (NETs), which facilitate further elimination of bacteria. PMN surface expression of RPSA also alleviates local inflammation and tissue injuries by inhibiting secretion of the pro-inflammatory cytokines, TNF-α and IL-6. Moreover, the single-cell BBB model showed that RPSA disrupts BBB integrity by downregulating expression of tight junction-associated membrane proteins on PMNs. Taken together, our data suggest that PMN-surface expression of RPSA is a double-edged sword. RPSA+ PMN owns a stronger ability of bacterial cleaning and weakens inflammatory cytokines release which are useful to anti-infection, but does hurt BBB. Partly, RPSA+ PMN may be extremely useful to control the infection as a therapeutic cellular population, following novel insights into the special PMN population.


Assuntos
Armadilhas Extracelulares/imunologia , Meningite Pneumocócica/imunologia , Neutrófilos/imunologia , Fagocitose/imunologia , Receptores de Laminina/imunologia , Proteínas Ribossômicas/imunologia , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Citocinas/imunologia , Meningite Pneumocócica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Receptores de Laminina/metabolismo , Proteínas Ribossômicas/metabolismo , Streptococcus suis/imunologia
17.
Antonie Van Leeuwenhoek ; 111(12): 2371-2384, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30008077

RESUMO

Actinobacillus pleuropneumoniae is the cause of porcine pleuropneumonia, for which the mortality rate is high. Host peripheral blood is a body site for the immune clearance of pathogens mediated by release of inflammatory factors. However, "out of control" inflammatory factor release can contribute to host death. To further understand the changes in the transcription level of immune-related effectors, samples of peripheral blood mononuclear cells (PBMCs) collected from piglets at different stages of infection (0, 24 and 120 h) were sequenced on an Illumina HiSeq™ 4000 platform. We found 3818 differentially expressed genes (DEGs) in the 24 h-infection group compared to the 0 h-infection group (Pb24-Vs-Pb0). DEGs mainly involved in the Gene ontology and KEGG pathways that included nucleic acid metabolism regulation, cell growth, cell differentiation, and organ morphological maintenance were not significantly enriched (P > 0.05). However, DEGs associated with protein kinase activity, receptor activation, metabolism, local adhesion and immune inflammatory responses were significantly enriched in Pb120-Vs-Pb24 (P < 0.05), as were those related to the T cell receptor signalling pathway, with most being down-regulated compared to the preceding stage (Pb24-Vs-Pb0). In PBMCs there were some changes in glucose metabolism, local adhesion and the immune inflammatory response (Pb120-Vs-Pb0). In addition, up-regulated DEGs, such as IL8, IL1ß, and CCL2, and were significantly enriched in immune-inflammatory related pathways compared to the uninfected stage, although they began to decline after 24 h.


Assuntos
Infecções por Actinobacillus/veterinária , Actinobacillus pleuropneumoniae/fisiologia , Leucócitos Mononucleares/imunologia , Pleuropneumonia/veterinária , Doenças dos Suínos/genética , Infecções por Actinobacillus/genética , Infecções por Actinobacillus/imunologia , Infecções por Actinobacillus/microbiologia , Animais , Feminino , Perfilação da Expressão Gênica , Leucócitos Mononucleares/microbiologia , Masculino , Pleuropneumonia/genética , Pleuropneumonia/imunologia , Pleuropneumonia/microbiologia , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/microbiologia
18.
Artigo em Inglês | MEDLINE | ID: mdl-29479521

RESUMO

Streptococcus suis serotype 2 (SS2) is a zoonotic pathogen that can cause meningitis both in pigs and in human beings. However, the pathogenesis of central nervous system (CNS) infection caused by SS2 have not yet been elucidated. To find the key molecules in cerebrospinal fluid (CSF) needed for the pathogenesis, a SS2 meningoencephalitic pig model and a SS2 non-meningoencephalitic pig model were established in this study. CSF was collected from infected piglets, and protein profiling was performed with label-free proteomics technology. A total of 813 differential proteins, including 52 up-regulated proteins and 761 down-regulated proteins, were found in the CSF of meningoencephalitic pigs compared with both non-meningoencephalitic pigs and healthy pigs. These 813 differential proteins were clustered into three main categories, namely, cellular component, biological process, and molecular function by gene ontology (GO) analysis. The most enriched subclasses of differential proteins in each category were exosome (44.3%), energy pathway (25.0%) and catalytic activity (11.3%), respectively. The most enriched subclasses of upregulated proteins were extracellular (62.1%), protein metabolism (34.5%) and cysteine-type peptidase activity (6.9%), and of downregulated proteins were exosomes (45.0%), energy pathway (24.0%) and catalytic activity (9.4%). Then, the differential proteins were further investigated by using the KEGG database and were found to participate in 16 KEGGs. The most enriched KEGG was citrate cycle (56.6%), and some of these differential proteins are associated with brain diseases such as Huntington's disease (18.6%), Parkinson's disease (23.8%) and Alzheimer's disease (17.6%). Sixteen of the 813 differential proteins, chosen randomly as examples, were further confirmed by enzyme-linked immunosorbent assay (ELISA) to support the proteomic data. To our knowledge, this is the first study to analyze the differential protein profiling of CSF between SS2 meningoencephalitic piglets and non-meningoencephalitic piglets by employing proteomic technology. The discovery and bioinformatics analysis of these differential proteins provides reference data not only for research on pathogenesis of SS2 CNS infection but also for diagnosis and drug therapy research.


Assuntos
Meningoencefalite/veterinária , Proteoma , Proteômica , Infecções Estreptocócicas/veterinária , Streptococcus suis/fisiologia , Doenças dos Suínos/líquido cefalorraquidiano , Doenças dos Suínos/microbiologia , Animais , Estudos de Casos e Controles , Cromatografia Líquida , Biologia Computacional/métodos , Ontologia Genética , Anotação de Sequência Molecular , Proteômica/métodos , Streptococcus suis/classificação , Suínos , Doenças dos Suínos/diagnóstico , Espectrometria de Massas em Tandem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...