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.
Front Immunol ; 13: 927215, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36148229

RESUMO

Neutrophil extracellular traps (NETs) are produced by neutrophil activation and usually have both anti-infective and pro-damage effects. Streptococcus uberis (S. uberis), one of the common causative organisms of mastitis, can lead to the production of NETs. Taurine, a free amino acid abundant in the organism, has been shown to have immunomodulatory effects. In this study, we investigated the molecular mechanisms of S. uberis-induced NETs formation and the regulatory role of taurine. The results showed that NETs had a disruptive effect on mammary epithelial cells and barriers, but do not significantly inhibit the proliferation of S. uberis. S. uberis induced NADPH oxidase-dependent NETs. TLR2-mediated activation of the MAPK signaling pathway was involved in this process. Taurine could inhibit the activation of MAPK signaling pathway and NADPH oxidase by modulating the activity of TAK1, thereby inhibiting the production of ROS and NETs. The effects of taurine on NADPH oxidase and NETs in S. uberis infection were also demonstrated in vivo. These results suggest that taurine can protect mammary epithelial cells and barriers from damage by reducing S. uberis-induced NETs. These data provide new insights and strategies for the prevention and control of mastitis.


Assuntos
Armadilhas Extracelulares , Mastite , Aminoácidos , Armadilhas Extracelulares/metabolismo , Feminino , Humanos , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Streptococcus , Taurina/farmacologia , Receptor 2 Toll-Like/metabolismo
2.
Food Funct ; 13(4): 1774-1784, 2022 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-35112684

RESUMO

Antibiotic-resistant strains of Streptococcus uberis (S. uberis) frequently cause clinical mastitis in dairy cows resulting in enormous economic losses. The regulation of immunometabolism is a promising strategy for controlling this bacterial infection. To investigate whether taurine alleviates S. uberis infection by the regulation of host glycolysis via HIF1α, the murine mammary epithelial cell line (EpH4-Ev) and C57BL/6J mice were challenged with S. uberis. Our data indicate that HIF1α-driven glycolysis promotes inflammation and damage in response to the S. uberis challenge. The activation of HIF1α is dependent on mTOR-mediated ROS production. These results were confirmed in vivo. Taurine, an intracellular metabolite present in most animal tissues, has been shown to effectively modulate HIF1α-triggered metabolic reprogramming and contributes to a reduction of inflammation, which reduces mammary tissue damage and prevents mammary gland dysfunction in S. uberis-induced mastitis. These data provide a novel putative prophylactic and therapeutic strategy for amelioration of dairy cow mastitis and bacterial inflammation.


Assuntos
Glicólise/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Infecções Estreptocócicas/metabolismo , Taurina/farmacologia , Animais , Linhagem Celular , Feminino , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Streptococcus/efeitos dos fármacos
3.
Int Immunopharmacol ; 101(Pt B): 108371, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34789427

RESUMO

Streptococcus uberis (S. uberis) is an important causative agent of mastitis, leading to significant economic losses to dairy industry. This research used a mouse mastitis model to investigate the protective effects of taurine on mammary inflammatory response and blood-milk barrier integrity in S. uberis challenge. The results showed that taurine attenuated S. uberis-induced mammary histopathological changes, especially neutrophil infiltration. The S. uberis-induced expression of pro-inflammatory mediators were decreased significantly by taurine. Further, we demonstrated that taurine limited the S. uberis-induced inflammatory responses via inhibiting the activation of NF-κB and MAPK signaling pathways. Inflammation usually disrupts the mammary barrier system. The recovery of claudin-3 and occludin expressions indicated that attenuation of inflammatory response by taurine can protect the integrity of blood-milk barrier in S. uberis infection. Taken together, our results reveal that the development of taurine as an effective prevention and control strategy for S. uberis-induced mastitis.


Assuntos
Inflamação/prevenção & controle , Mastite/veterinária , Leite , Infecções Estreptocócicas/tratamento farmacológico , Streptococcus , Taurina/farmacologia , Animais , Feminino , Mastite/tratamento farmacológico , Mastite/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Organismos Livres de Patógenos Específicos , Infecções Estreptocócicas/microbiologia
4.
Vet Microbiol ; 261: 109207, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34419774

RESUMO

Mammary gland-derived Escherichia coli (E. coli) is an important pathogen causing dairy cow mastitis. YdiV, with EAL-like domains, inhibits flagellum biogenesis and motility and affects c-di-GMP (eubacterial signaling molecule) concentration changes in bacteria. However, the pathophysiological role of ydiV in host-pathogen cross-talk still needs to be elucidated. In this study, firstly constructed the ydiV mutant (NJ17ΔydiV) and ydiV complementary (cNJ17ΔydiV) E. coli strains to infect mouse mammary epithelial cells (EpH4-Ev) and macrophages (RAW264.7), as well as mouse mammary glands, respectively. Then biological characteristics, adaptor molecules in related signaling pathways, proinflammatory cytokines and the extent of host cell damage was evaluated. Compared with E. coli NJ17 infected mice, the bacterial load in the mammary gland of NJ17ΔydiV was significantly lower and the extent of the damage was alleviated. Notably, the deletion of ydiV significantly aggravated cell damage in RAW264.7 cells and compared with the wild-type strain, NJ17ΔydiV significantly activated the STING/TBK1/IRF3 pathway in macrophages. In EpH4-Ev cells, although STING did not sense E. coli NJ17 invasion, IRF3 was activated by the NJ17ΔydiV strain. Taken together, ydiV deletion significantly affects a variety of biological characteristics and induces severe cell damage, while the STING/TBK1/IRF3 pathway actively participated in pathogen elimination in the host. This study highlights a new role for ydiV in E. coli infection and provides a foundation for further studies to better understand host-bacteria interactions and potential prophylactic strategies for infectious diseases.


Assuntos
Proteínas de Transporte/metabolismo , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Escherichia coli/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Evasão da Resposta Imune/genética , Animais , Carga Bacteriana , Proteínas de Transporte/genética , Linhagem Celular , Células Epiteliais/imunologia , Células Epiteliais/virologia , Proteínas de Escherichia coli/genética , Feminino , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Fator Regulador 3 de Interferon/imunologia , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/virologia , Proteínas de Membrana/imunologia , Camundongos , Mutação , Proteínas Serina-Treonina Quinases/imunologia , Células RAW 264.7
5.
Front Immunol ; 12: 631113, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33777017

RESUMO

Streptococcus uberis infection can cause serious inflammation and damage to mammary epithelial cells and tissues that can be significantly alleviated by taurine. Autophagy plays an important role in regulating immunity and clearing invasive pathogens and may be regulated by taurine. However, the relationships between taurine, autophagy, and S. uberis infection remain unclear. Herein, we demonstrate that taurine augments PTEN activity and inhibits Akt/mTOR signaling, which decreases phosphorylation of ULK1 and ATG13 by mTOR and activates autophagy. Activating autophagy accelerates the degradation of intracellular S. uberis, reduces intracellular bacterial load, inhibits over-activation of the NF-κB pathway, and alleviates the inflammation and damage caused by S. uberis infection. This study increases our understanding of the mechanism through which taurine regulates autophagy and is the first to demonstrate the role of autophagy in S. uberis infected MAC-T cells. Our study also provides a theoretical basis for employing nutritional elements (taurine) to regulate innate immunity and control S. uberis infection. It also provides theoretical support for the development of prophylactic strategies for this important pathogen.


Assuntos
Autofagia/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/microbiologia , Inflamação/microbiologia , Inflamação/prevenção & controle , Streptococcus/patogenicidade , Taurina/farmacologia , Animais , Bovinos , Linhagem Celular , Contagem de Colônia Microbiana , Inflamação/imunologia , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/efeitos dos fármacos , Mastite Bovina/microbiologia , Transdução de Sinais/efeitos dos fármacos , Streptococcus/imunologia
6.
Cells ; 9(2)2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-32098158

RESUMO

Mastitis caused by Streptococcus uberis (S. uberis) is a common and difficult-to-cure clinical disease in dairy cows. In this study, the role of Toll-like receptors (TLRs) and TLR-mediated signaling pathways in mastitis caused by S. uberis was investigated using mouse models and mammary epithelial cells (MECs). We used S. uberis to infect mammary glands of wild type, TLR2-/- and TLR4-/- mice and quantified the adaptor molecules in TLR signaling pathways, proinflammatory cytokines, tissue damage, and bacterial count. When compared with TLR4 deficiency, TLR2 deficiency induced more severe pathological changes through myeloid differentiation primary response 88 (MyD88)-mediated signaling pathways during S. uberis infection. In MECs, TLR2 detected S. uberis infection and induced mitochondrial reactive oxygen species (mROS) to assist host in controlling the secretion of inflammatory factors and the elimination of intracellular S. uberis. Our results demonstrated that TLR2-mediated mROS has a significant effect on S. uberis-induced host defense responses in mammary glands as well as in MECs.


Assuntos
Mastite/metabolismo , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Infecções Estreptocócicas/metabolismo , Streptococcus/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Feminino , Masculino , Glândulas Mamárias Animais/microbiologia , Glândulas Mamárias Animais/patologia , Mastite/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , Organismos Livres de Patógenos Específicos , Infecções Estreptocócicas/microbiologia , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
7.
Vet Res ; 50(1): 109, 2019 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-31831050

RESUMO

The function of Autoinducer-2 (AI-2) which acts as the signal molecule of LuxS-mediated quorum sensing, is regulated through the lsr operon (which includes eight genes: lsrK, lsrR, lsrA, lsrC, lsrD, lsrB, lsrF, and lsrG). However, the functions of the lsr operon remain unclear in avian pathogenic Escherichia coli (APEC), which causes severe respiratory and systemic diseases in poultry. In this study, the presence of the lsr operon in 60 APEC clinical strains (serotypes O1, O2, and O78) was investigated and found to be correlated with serotype and has the highest detection rate in O78. The AI-2 binding capacity of recombinant protein LsrB of APEC (APEC-LsrB) was verified and was found to bind to AI-2 in vitro. In addition, the lsr operon was mutated in an APEC strain (APEC94Δlsr(Cm)) and the mutant was found to be defective in motility and AI-2 uptake. Furthermore, deletion of the lsr operon attenuated the virulence of APEC, with the LD50 of APEC94Δlsr(Cm) decreasing 294-fold compared with wild-type strain APEC94. The bacterial load in the blood, liver, spleen, and kidneys of ducks infected with APEC94Δlsr(Cm) decreased significantly (p < 0.0001). The results of transcriptional analysis showed that 62 genes were up-regulated and 415 genes were down-regulated in APEC94Δlsr(Cm) compared with the wild-type strain and some of the down-regulated genes were associated with the virulence of APEC. In conclusion, our study suggests that lsr operon plays a role in the pathogenesis of APEC.


Assuntos
Proteínas de Transporte/metabolismo , Infecções por Escherichia coli/veterinária , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Homosserina/análogos & derivados , Lactonas/metabolismo , Doenças das Aves Domésticas/microbiologia , Percepção de Quorum , Animais , Biofilmes , Proteínas de Transporte/genética , China/epidemiologia , Escherichia coli/genética , Escherichia coli/patogenicidade , Infecções por Escherichia coli/epidemiologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Homosserina/genética , Homosserina/metabolismo , Aves Domésticas , Doenças das Aves Domésticas/epidemiologia , Sorogrupo
8.
AMB Express ; 9(1): 185, 2019 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-31728678

RESUMO

Escherichia coli, Pasteurella multocida, Proteus mirabilis, Pseudomonas aeruginosa, Salmonella spp. and Staphylococcus aureus are six bacterial pathogens of avian. However, these pathogens may cause many similar pathological changes, resulting in clinical isolates that are difficult to quickly and simultaneously detect and identify. Here, a multiplex polymerase chain reaction (m-PCR) assay is reported to rapidly identify targets genes (phoA, KMT1, ureR, toxA, invA, and nuc) of these six pathogens in clinical samples. Six pairs of specific primers were designed. The optimal reaction conditions, specificity, and sensitivity of the m-PCR assay were investigated. The results showed that betaine remarkably improved amplification of the target genes. Specific test results showed that all six pathogens were detected by the proposed m-PCR protocol without cross-amplification with viruses or parasites. Sensitivity test results showed that the m-PCR system could amplify the six target genes from bacterial genomes or cultures with template amounts of 500 pg or 2.8-8.6 × 103 colony forming units, respectively. Furthermore, the six bacterial pathogens isolated from the infected tissue samples were successfully identified. The proposed m-PCR assay is a useful tool to monitor and diagnose bacterial infection in birds with high specificity, sensitivity and throughput.

9.
Front Immunol ; 10: 1825, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31447841

RESUMO

Taurine may alleviate the inflammatory injury induced by Streptococcus uberis (S. uberis) infection by regulating intracellular Ca2+ levels. However, the underlying mechanisms remain unclear. Infection leads to subversion of phosphoinositides (PIs) which are closely related to Ca2+ signaling. In order to investigate whether taurine regulates inflammation by means of PIs/ Ca2+ systems, competitive inhibitors of taurine (ß-alanine) siTauT, siPAT1, siPLC, siCaN, siPKC, and inhibitors of PLC (U73122), PKC (RO31-8220), and CaN (FK 506) were used. The results indicate that taurine transfers the extracellular nutrient signal for intercellular innate immunity to phosphoinositides without a need to enter the cytoplasm while regulating intracellular Ca2+ levels during inflammation. Both the Ca2+-PKCα-NF-κB, and Ca2+-CaM-CaN-NFAT signaling pathways of S. uberis infection and the regulatory roles of taurine follow activation of PIs/Ca2+ systems. These data increase our understanding on the mechanisms of multifunctional nutrient, taurine attenuated inflammatory responses caused by S. uberis infection, and provide theoretical support for the prevention of this disease.


Assuntos
Sinalização do Cálcio/fisiologia , Mastite Bovina/metabolismo , Fosfatidilinositóis/metabolismo , Infecções Estreptocócicas/metabolismo , Taurina/metabolismo , Animais , Bovinos , Células Cultivadas , Feminino , Inflamação/imunologia , Inflamação/metabolismo , Mastite Bovina/imunologia , Fosfatidilinositóis/imunologia , Infecções Estreptocócicas/imunologia , Streptococcus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA