Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Cell Infect Microbiol ; 14: 1347615, 2024.
Article in English | MEDLINE | ID: mdl-38465233

ABSTRACT

Picornaviruses, which are positive-stranded, non-enveloped RNA viruses, are known to infect people and animals with a broad spectrum of diseases. Among the nonstructural proteins in picornaviruses, 2C proteins are highly conserved and exhibit multiple structural domains, including amphipathic α-helices, an ATPase structural domain, and a zinc finger structural domain. This review offers a comprehensive overview of the functional structures of picornaviruses' 2C protein. We summarize the mechanisms by which the 2C protein enhances viral replication. 2C protein interacts with various host factors to form the replication complex, ultimately promoting viral replication. We review the mechanisms through which picornaviruses' 2C proteins interact with the NF-κB, RIG-I, MDA5, NOD2, and IFN pathways, contributing to the evasion of the antiviral innate immune response. Additionally, we provide an overview of broad-spectrum antiviral drugs for treating various enterovirus infections, such as guanidine hydrochloride, fluoxetine, and dibucaine derivatives. These drugs may exert their inhibitory effects on viral infections by targeting interactions with 2C proteins. The review underscores the need for further research to elucidate the precise mechanisms of action of 2C proteins and to identify additional host factors for potential therapeutic intervention. Overall, this review contributes to a deeper understanding of picornaviruses and offers insights into the antiviral strategies against these significant viral pathogens.


Subject(s)
Picornaviridae , Humans , Animals , NF-kappa B/metabolism , RNA , Virus Replication , Antiviral Agents/pharmacology , Structure-Activity Relationship
2.
Mol Med ; 30(1): 30, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38395749

ABSTRACT

BACKGROUND: Sepsis is a systemic inflammatory response which is frequently associated with acute lung injury (ALI). Activating transcription factor 3 (ATF3) promotes M2 polarization, however, the biological effects of ATF3 on macrophage polarization in sepsis remain undefined. METHODS: LPS-stimulated macrophages and a mouse model of cecal ligation and puncture (CLP)-induced sepsis were generated as in vitro and in vivo models, respectively. qRT-PCR and western blot were used to detect the expression of ATF3, ILF3, NEAT1 and other markers. The phenotypes of macrophages were monitored by flow cytometry, and cytokine secretion was measured by ELISA assay. The association between ILF3 and NEAT1 was validated by RIP and RNA pull-down assays. RNA stability assay was employed to assess NEAT1 stability. Bioinformatic analysis, luciferase reporter and ChIP assays were used to study the interaction between ATF3 and ILF3 promoter. Histological changes of lung tissues were assessed by H&E and IHC analysis. Apoptosis in lungs was monitored by TUNEL assay. RESULTS: ATF3 was downregulated, but ILF3 and NEAT1 were upregulated in PBMCs of septic patients, as well as in LPS-stimulated RAW264.7 cells. Overexpression of ATF3 or silencing of ILF3 promoted M2 polarization of RAW264.7 cells via regulating NEAT1. Mechanistically, ILF3 was required for the stabilization of NEAT1 through direct interaction, and ATF3 was a transcriptional repressor of ILF3. ATF3 facilitated M2 polarization in LPS-stimulated macrophages and CLP-induced septic lung injury via ILF3/NEAT1 axis. CONCLUSION: ATF3 triggers M2 macrophage polarization to protect against the inflammatory injury of sepsis through ILF3/NEAT1 axis.


Subject(s)
Activating Transcription Factor 3 , Macrophages , RNA, Long Noncoding , Sepsis , Animals , Humans , Mice , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Lipopolysaccharides , Macrophages/metabolism , Nuclear Factor 90 Proteins/genetics , Nuclear Factor 90 Proteins/metabolism , RAW 264.7 Cells , Sepsis/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
3.
J Infect Dis ; 229(2): 522-534, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-37647879

ABSTRACT

BACKGROUND: Patients with sepsis resulting in acute lung injury (ALI) usually have increased mortality. Ferroptosis is a vital regulator in sepsis-induced ALI. Exploring the association of ferroptosis and sepsis-induced ALI is crucial for the management of sepsis-induced ALI. METHODS: Whole blood was collected from sepsis patients. Mice were treated with cecal ligation and puncture (CLP) to model sepsis. Primary murine pulmonary microvascular endothelial cells were treated with lipopolysaccharide as a cell model. Ferroptosis was evaluated by analyzing levels of iron, malonaldehyde, glutathione, nonheme iron, ferroportin, ferritin, and GPX4. Hematoxylin and eosin and Masson's trichrome staining were applied to examine lung injury and collagen deposition. Cell apoptosis was analyzed by caspase-3 activity and TUNEL assays. Gene regulatory relationship was verified using RNA pull-down and immunoprecipitation assays. RESULTS: CircEXOC5 was highly expressed in sepsis patients and CLP-treated mice, in which knockdown alleviated CLP-induced pulmonary inflammation and injury, and ferroptosis. CircEXOC5 recruited IGF2BP2 to degrade ATF3 mRNA. The demethylase ALKBH5 was responsible for circEXOC5 upregulation through demethylation. CircEXOC5 silencing significantly improved sepsis-induced ALI and survival rate of mice by downregulating ATF3. CONCLUSIONS: ALKBH5-mediated upregulation of circEXOC5 exacerbates sepsis-induced ALI by facilitating ferroptosis through IGF2BP2 recruitment to degrade ATF3 mRNA.


Subject(s)
Acute Lung Injury , Ferroptosis , Sepsis , Humans , Mice , Animals , Endothelial Cells/metabolism , Acute Lung Injury/etiology , Lung/metabolism , Sepsis/metabolism , Iron/metabolism , RNA, Messenger/metabolism , Lipopolysaccharides , RNA-Binding Proteins/metabolism , Activating Transcription Factor 3/metabolism
4.
Appl Opt ; 62(30): 7910-7916, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-38038083

ABSTRACT

Deep learning has been attracting more and more attention in the phase unwrapping of fringe projection profilometry (FPP) in recent years. In order to improve the accuracy of the deep-learning-based unwrapped phase methods from a single fringe pattern, this paper proposes a single-input triple-output neural network structure with a physical prior. In the proposed network, a single-input triple-output network structure is developed to convert the input fringe pattern into three intermediate outputs: the wrapped phase, the fringe order, the coarse unwrapped phase, and the final output high-precision unwrapped phase from the three outputs. Moreover, a new, to the best of our knowledge, loss function is designed to improve the performance of the model using a physical prior about these three outputs in FPP. Numerous experiments demonstrated that the proposed network is able to improve the accuracy of the unwrapped phase, which can also be extended to other deep learning phase unwrapping models.

5.
Mol Cell Biochem ; 478(4): 743-754, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36074295

ABSTRACT

Acute lung injury (ALI) caused by sepsis is characterized by a destructive high inflammatory response in lungs, which is the ultimate cause of high mortality to patients diagnosed with sepsis. The objective of the present study is to explore the effect and related mechanisms of circEXOC5 on pyroptosis in septic ALI. Sepsis ALI mouse model was induced and established by CLP induction and sepsis MPVEC cell model by LPS. HE staining was used to detect lung tissue pathological changes. ELISA, flow cytometry, and Western blot were utilized to evaluate the release of inflammatory cytokines and cell pyroptosis, and RIP was applied to verify the binding relationship between EZH2 and circEXOC5 or Nrf2. Finally, the interaction between CircEXOC5 and EZH2, H3k27me3, and Nrf2 promoter regions was clarified using ChIP. CircEXOC5 levels were notably ascended in the lung tissues of septic ALI mice. And silencing circEXOC5 inhibited cell pyroptosis and the release of inflammatory cytokines in MPVEC stimulated by LPS. In addition, RIP and ChIP indicated that Nrf2 expression in MPVECs cells could be inhibited by circEXOC5 via recruiting EZH2. In addition, ML385 (a specific inhibitor of Nrf2) reversed the efficacy of Knockdown of circEXOC5 on the Inhibition of pyroptosis and inflammation of MPVEC cells stimulated by LPS. These results indicated that CircEXOC5 could promote cell pyroptosis through epigenetic inhibition of Nrf2 in septic ALI.


Subject(s)
Acute Lung Injury , Sepsis , Mice , Animals , Pyroptosis , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Lipopolysaccharides/toxicity , Lipopolysaccharides/metabolism , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Lung/pathology , Cytokines/metabolism , Sepsis/metabolism , Epigenesis, Genetic , Mice, Inbred C57BL
6.
Immunobiology ; 227(4): 152219, 2022 07.
Article in English | MEDLINE | ID: mdl-35709678

ABSTRACT

BACKGROUND: Sepsis causes severe acute lung injury (ALI). Circular RNA is involved in the regulation of sepsis-related ALI progression. The regulation mechanism of circEXOC5 in sepsis-induced ALI is still unclear. Whether circEXOC5 is involved in the regulation of ferroptosis remains to be explored. METHODS: We constructed a mouse model of sepsis through cecal ligation and puncture (CLP). LPS induced mouse lung microvascular endothelial cells (MPVECs) to construct a sepsis cell model. The expression of circEXOC5 in the sepsis model was detected by qPCR. The extent of lung injury in mice was analyzed by HE staining. The contents of GSH/GSSG, iron, MDA and 4HNE in mice lung tissues and cells were detected by the kit. And further the ROS content was detected in the cells. Finally, the binding relationship between circEXOC5 and PTBP1 was detected by RIP and RNA pulldown. RESULTS: Our results showed that the circEXOC5 expression was significantly increased in the in vivo and in vitro models of sepsis. And after inhibiting circEXOC5, it improved the lung injury of septic mice. It was confirmed in cell models that ROS levels and ferroptosis in cells were reduced after knocking down circEXOC5. In addition, the expressions of ACSL4 and Gpx4 proteins were regulated by the level of circEXOC5. Finally, we also found that circEXOC5 had a direct binding relationship with PTBP1. CONCLUSION: Our study found that the expression of cell ferroptosis and circEXOC5 increased in ALI induced by sepsis, and circEXOC5 aggravated ferroptosis in septic cells by regulating the PTBP1/ACSL4 axis.


Subject(s)
Acute Lung Injury , Ferroptosis , Sepsis , Acute Lung Injury/genetics , Animals , Coenzyme A Ligases/metabolism , Endothelial Cells/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Lung/metabolism , Mice , Mice, Inbred C57BL , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , RNA Stability , Reactive Oxygen Species/metabolism , Sepsis/complications
SELECTION OF CITATIONS
SEARCH DETAIL