Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 142(Pt B): 113171, 2024 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-39312862

RESUMO

Recently studies showed that cow mastitis seriously affected the economic benefit of dairy industry and pathogen infection including S. aureus is the main cause of mastitis. However, there is still a lack of safe and effective treatment for S. aureus-induced mastitis due to its complex pathogenesis. Endogenous retroviruses (ERVs) have long been symbiotic with mammals, and most ERVs still have the ability to produces complementary DNA (cDNA) by reverse transcription, whose induction by commensal or pathogens can regulate host immunity and inflammatory responses through the cGAS-STING pathway. However, whether and how ERVs participate in the pathogenesis of S. aureus-induced mastitis still unclear. In this study, we found that S. aureus treatment increased the levels of ERVs and IFN-ß. Inhibition the transcription of ERVs by emtricitabine alleviated S. aureus-induced mammary injury, reduced mammary bacterial burden, and inhibited the production of mammary proinflammatory factors including TNF-α, IL-1ß and MPO activity. Moreover, inhibition of ERVs restored the function of blood-milk barrier caused by S. aureus. Next, we showed that S. aureus infection activated mammary cGAS-STING signaling pathway, which was mediated by ERVs, as evidenced by emtricitabine inhibited S. aureus-induced activation of the cGAS-STING pathway. Interestingly, inhibition of cGAS-STING by Ru.521 and H151 respectively, significantly alleviated S. aureus-induced mammary injury and inflammatory responses, which was associated with the inhibition of NF-κB and NLRP3 signaling pathways. In conclusion, our study revealed that ERVs regulate the development of S. aureus-induced mastitis in mice through NF-κB- and NLRP3-mediated inflammatory responses via the activation of cGAS-STING pathway, suggesting that targeting ERVs-cGAS-STING axis may be a potential approach for the treatment of S. aureus-induced mastitis.

2.
Int Immunopharmacol ; 140: 112818, 2024 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-39083924

RESUMO

Cell death caused by severe Staphylococcus aureus (S. aureus) infection is a fatal threat to humans and animals. However, whether ferroptosis, an iron-dependent form of cell death, is involved in S. aureus-induced cell death and its role in S. aureus-induced diseases are unclear. Using a mouse mastitis model and mammary epithelial cells (MMECs), we investigated the role of ferroptosis in the pathogenesis of S. aureus infection. The results revealed that S. aureus-induced ferroptosis in vivo and in vitro as demonstrated by dose-dependent increases in cell death; the level of malondialdehyde (MDA), the final product of lipid peroxidation; and dose-dependent decrease the production of the antioxidant glutathione (GSH). Treatment with typical inhibitors of ferroptosis, including ferrostatin-1 (Fer-1) and deferiprone (DFO), significantly inhibited S. aureus-induced death in MMECs. Mechanistically, treatment with S. aureus activated the protein kinase RNA-like ER kinase (PERK)-eukaryotic initiation factor 2, α subunit (eIF2α)-activating transcription factor 4 (ATF4)-C/EBP homologous protein (CHOP) pathway, which subsequently upregulated autophagy and promoted S. aureus-induced ferroptosis. The activation of autophagy degraded ferritin, resulting in iron dysregulation and ferroptosis. In addition, we found that excessive reactive oxygen species (ROS) production induced ferroptosis and activated endoplasmic reticulum (ER) stress, manifesting as elevated p-PERK-p-eIF2α-ATF4-CHOP pathway protein levels. Collectively, our findings indicate that ferroptosis is involved in S. aureus-induced mastitis via ER stress-mediated autophagy activation, implying a potential strategy for the prevention of S. aureus-associated diseases by targeting ferroptosis. In conclusion, the ROS-ER stress-autophagy axis is involved in regulating S. aureus-induced ferroptosis in MMECs. These findings not only provide a new potential mechanism for mastitis induced by S. aureus but also provide a basis for the treatment of other ferroptotic-related diseases.


Assuntos
Autofagia , Estresse do Retículo Endoplasmático , Ferroptose , Mastite , Infecções Estafilocócicas , Staphylococcus aureus , Animais , Ferroptose/efeitos dos fármacos , Mastite/microbiologia , Mastite/metabolismo , Mastite/patologia , Feminino , Staphylococcus aureus/fisiologia , Infecções Estafilocócicas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Células Epiteliais/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Fator 4 Ativador da Transcrição/genética , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Animais/microbiologia , Fator de Transcrição CHOP/metabolismo , Humanos , Transdução de Sinais , Espécies Reativas de Oxigênio/metabolismo , Células Cultivadas
3.
PLoS Pathog ; 19(11): e1011764, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37948460

RESUMO

Subacute ruminal acidosis (SARA) has been demonstrated to promote the development of mastitis, one of the most serious diseases in dairy farming worldwide, but the underlying mechanism is unclear. Using untargeted metabolomics, we found hexadecanamide (HEX) was significantly reduced in rumen fluid and milk from cows with SARA-associated mastitis. Herein, we aimed to assess the protective role of HEX in Staphylococcus aureus (S. aureus)- and SARA-induced mastitis and the underlying mechanism. We showed that HEX ameliorated S. aureus-induced mastitis in mice, which was related to the suppression of mammary inflammatory responses and repair of the blood-milk barrier. In vitro, HEX depressed S. aureus-induced activation of the NF-κB pathway and improved barrier integrity in mouse mammary epithelial cells (MMECs). In detail, HEX activated PPARα, which upregulated SIRT1 and subsequently inhibited NF-κB activation and inflammatory responses. In addition, ruminal microbiota transplantation from SARA cows (S-RMT) caused mastitis and aggravated S. aureus-induced mastitis, while these changes were reversed by HEX. Our findings indicate that HEX effectively attenuates S. aureus- and SARA-induced mastitis by limiting inflammation and repairing barrier integrity, ultimately highlighting the important role of host or microbiota metabolism in the pathogenesis of mastitis and providing a potential strategy for mastitis prevention.


Assuntos
Mastite , Staphylococcus aureus , Humanos , Feminino , Animais , Camundongos , Bovinos , Staphylococcus aureus/metabolismo , NF-kappa B/metabolismo , Leite , Mastite/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA