Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.652
Filtrer
1.
Cell Death Dis ; 15(7): 477, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961079

RÉSUMÉ

Mitochondrial dysfunction can elicit multiple inflammatory pathways, especially when apoptotic caspases are inhibited. Such an inflammatory program is negatively regulated by the autophagic disposal of permeabilized mitochondria. Recent data demonstrate that the ubiquitination of mitochondrial proteins is essential for NEMO-driven NF-kB activation downstream of mitochondrial permeabilization.


Sujet(s)
Mitochondries , Facteur de transcription NF-kappa B , Transduction du signal , Animaux , Humains , Apoptose , Autophagie , I-kappa B Kinase/métabolisme , Mitochondries/métabolisme , Protéines mitochondriales/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Ubiquitination
2.
Drug Des Devel Ther ; 18: 2693-2712, 2024.
Article de Anglais | MEDLINE | ID: mdl-38974121

RÉSUMÉ

Background: Chronic kidney disease (CKD) is a significant worldwide health concern that leads to high mortality rates. The bioactive substance costunolide (CTD) has demonstrated several pharmacological effects and holds promise as a CKD treatment. This study aims to investigate the impact of CTD on CKD and delve into its mechanisms of action. Methods: Unilateral ureteral obstruction (UUO) methods and renal fibrosis mice models were created. Various concentrations of CTD were injected into UUO mice models to investigate the therapeutic effects of CTD on renal fibrosis of mice. Then, renal morphology, pathological changes, and the expression of genes related to fibrosis, inflammation and ferroptosis were analysed. RNA sequencing was utilized to identify the main biological processes and pathways involved in renal injury. Finally, both overexpression and inhibition of IKKß were studied to examine their respective effects on fibrosis and inflammation in both in vitro and in vivo models. Results: CTD treatment was found to significantly alleviate fibrosis, inflammation and ferroptosis in UUO-induced renal fibrosis mice models. The results of RNA sequencing suggested that the IKKß acted as key regulatory factor in renal injury and the expression of IKKß was increased in vitro and in vivo renal fibrosis model. Functionally, down-regulated IKKß expression inhibits ferroptosis, inflammatory cytokine production and collagen deposition. Conversely, IKKß overexpression exacerbates progressive renal fibrosis. Mechanistically, CTD alleviated renal fibrosis and inflammation by inhibiting the expression of IKKß and attenuating IKKß/NF-κB pathway. Conclusion: This study demonstrates that CTD could mitigate renal fibrosis, ferroptosis and inflammation in CKD by modulating the IKKß/NF-κB pathway, which indicates targeting IKKß has an enormous potential for treating CKD.


Sujet(s)
I-kappa B Kinase , Souris de lignée C57BL , Facteur de transcription NF-kappa B , Insuffisance rénale chronique , Sesquiterpènes , Animaux , I-kappa B Kinase/métabolisme , I-kappa B Kinase/antagonistes et inhibiteurs , Souris , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Insuffisance rénale chronique/traitement médicamenteux , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/anatomopathologie , Sesquiterpènes/pharmacologie , Mâle , Modèles animaux de maladie humaine , Fibrose/traitement médicamenteux , Humains , Obstruction urétérale/traitement médicamenteux , Obstruction urétérale/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Inflammation/traitement médicamenteux , Inflammation/métabolisme
3.
Gut Microbes ; 16(1): 2374608, 2024.
Article de Anglais | MEDLINE | ID: mdl-38972055

RÉSUMÉ

With the increasing of aging population and the consumption of high-fat diets (HFD), the incidence of Alzheimer's disease (AD) has skyrocketed. Natural antioxidants show promising potential in the prevention of AD, as oxidative stress and neuroinflammation are two hallmarks of AD pathogenesis. Here, we showed that quinic acid (QA), a polyphenol derived from millet, significantly decreased HFD-induced brain oxidative stress and neuroinflammation and the levels of Aß and p-Tau. Examination of gut microbiota suggested the improvement of the composition of gut microbiota in HFD mice after QA treatment. Metabolomic analysis showed significant increase of gut microbial tryptophan metabolites indole-3-acetic acid (IAA) and kynurenic acid (KYNA) by QA. In addition, IAA and KYNA showed negative correlation with pro-inflammatory factors and AD indicators. Further experiments on HFD mice proved that IAA and KYNA could reproduce the effects of QA that suppress brain oxidative stress and inflammation and decrease the levels of of Aß and p-Tau. Transcriptomics analysis of brain after IAA administration revealed the inhibition of DR3/IKK/NF-κB signaling pathway by IAA. In conclusion, this study demonstrated that QA could counteract HFD-induced brain oxidative stress and neuroinflammation by regulating inflammatory DR3/IKK/NF-κB signaling pathway via gut microbial tryptophan metabolites.


Sujet(s)
Encéphale , Alimentation riche en graisse , Microbiome gastro-intestinal , Souris de lignée C57BL , Facteur de transcription NF-kappa B , Stress oxydatif , Acide quinique , Transduction du signal , Tryptophane , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Tryptophane/métabolisme , Alimentation riche en graisse/effets indésirables , Souris , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Stress oxydatif/effets des médicaments et des substances chimiques , Acide quinique/analogues et dérivés , Acide quinique/pharmacologie , Acide quinique/métabolisme , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/prévention et contrôle , I-kappa B Kinase/métabolisme , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/prévention et contrôle , Acides indolacétiques/métabolisme , Acide kynurénique/métabolisme , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Inflammation/prévention et contrôle
4.
Immunohorizons ; 8(7): 478-491, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-39007717

RÉSUMÉ

IκB kinase (IKK)α controls noncanonical NF-κB signaling required for lymphoid organ development. We showed previously that lymph node formation is ablated in IkkαLyve-1 mice constitutively lacking IKKα in lymphatic endothelial cells (LECs). We now reveal that loss of IKKα in LECs leads to the formation of BALT in the lung. Tertiary lymphoid structures appear only in the lungs of IkkαLyve-1 mice and are not present in any other tissues, and these highly organized BALT structures form after birth and in the absence of inflammation. Additionally, we show that IkkαLyve-1 mice challenged with influenza A virus (IAV) exhibit markedly improved survival and reduced weight loss compared with littermate controls. Importantly, we determine that the improved morbidity and mortality of IkkαLyve-1 mice is independent of viral load and rate of clearance because both mice control and clear IAV infection similarly. Instead, we show that IFN-γ levels are decreased, and infiltration of CD8 T cells and monocytes into IkkαLyve-1 lungs is reduced. We conclude that ablating IKKα in LECs promotes BALT formation and reduces the susceptibility of IkkαLyve-1 mice to IAV infection through a decrease in proinflammatory stimuli.


Sujet(s)
Homéostasie , I-kappa B Kinase , Virus de la grippe A , Poumon , Infections à Orthomyxoviridae , Animaux , I-kappa B Kinase/métabolisme , I-kappa B Kinase/génétique , Souris , Poumon/immunologie , Poumon/virologie , Poumon/anatomopathologie , Infections à Orthomyxoviridae/immunologie , Virus de la grippe A/immunologie , Cellules endothéliales/immunologie , Cellules endothéliales/métabolisme , Lymphocytes T CD8+/immunologie , Souris de lignée C57BL , Souris knockout , Transduction du signal/immunologie , Interféron gamma/métabolisme
5.
J Transl Med ; 22(1): 642, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982514

RÉSUMÉ

BACKGROUND: Oncogenic mutations in the RAS gene are associated with uncontrolled cell growth, a hallmark feature contributing to tumorigenesis. While diverse therapeutic strategies have been diligently applied to treat RAS-mutant cancers, successful targeting of the RAS gene remains a persistent challenge in the field of cancer therapy. In our study, we discover a promising avenue for addressing this challenge. METHODS: In this study, we tested the viability of several cell lines carrying oncogenic NRAS, KRAS, and HRAS mutations upon treatment with IkappaBalpha (IκBα) inhibitor BAY 11-7082. We performed both cell culture-based viability assay and in vivo subcutaneous xenograft-based assay to confirm the growth inhibitory effect of BAY 11-7082. We also performed large RNA sequencing analysis to identify differentially regulated genes and pathways in the context of oncogenic NRAS, KRAS, and HRAS mutations upon treatment with BAY 11-7082. RESULTS: We demonstrate that oncogenic NRAS, KRAS, and HRAS activate the expression of IκBα kinase. BAY 11-7082, an inhibitor of IκBα kinase, attenuates the growth of NRAS, KRAS, and HRAS mutant cancer cells in cell culture and in mouse model. Mechanistically, BAY 11-7082 inhibitor treatment leads to suppression of the PI3K-AKT signaling pathway and activation of apoptosis in all RAS mutant cell lines. Additionally, we find that BAY 11-7082 treatment results in the downregulation of different biological pathways depending upon the type of RAS protein that may also contribute to tumor growth inhibition. CONCLUSION: Our study identifies BAY 11-7082 to be an efficacious inhibitor for treating RAS oncogene (HRAS, KRAS, and NRAS) mutant cancer cells. This finding provides new therapeutic opportunity for effective treatment of RAS-mutant cancers.


Sujet(s)
Antinéoplasiques , Nitriles , Sulfones , Humains , Nitriles/pharmacologie , Sulfones/pharmacologie , Animaux , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Tumeurs/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , I-kappa B Kinase/métabolisme , I-kappa B Kinase/antagonistes et inhibiteurs , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Mutation/génétique , Souris , Protéines proto-oncogènes c-akt/métabolisme , Inhibiteur alpha de NF-KappaB/métabolisme , Protéines G ras/métabolisme , Protéines G ras/antagonistes et inhibiteurs
6.
Viruses ; 16(6)2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38932241

RÉSUMÉ

African swine fever (ASF) is an acute, hemorrhagic, highly contagious disease in pigs caused by African swine fever virus (ASFV). Our previous study identified that the ASFV MGF300-2R protein functions as a virulence factor and found that MGF300-2R degrades IKKß via selective autophagy. However, the E3 ubiquitin ligase responsible for IKKß ubiquitination during autophagic degradation still remains unknown. In order to solve this problem, we first pulled down 328 proteins interacting with MGF300-2R through immunoprecipitation-mass spectrometry. Next, we analyzed and confirmed the interaction between the E3 ubiquitin ligase TRIM21 and MGF300-2R and demonstrated the catalytic role of TRIM21 in IKKß ubiquitination. Finally, we indicated that the degradation of IKKß by MGF300-2R was dependent on TRIM21. In summary, our results indicate TRIM21 is the E3 ubiquitin ligase involved in the degradation of IKKß by MGF300-2R, thereby augmenting our understanding of the functions of MGF300-2R and offering insights into the rational design of live attenuated vaccines and antiviral strategies against ASF.


Sujet(s)
Virus de la peste porcine africaine , I-kappa B Kinase , Ribonucléoprotéines , Ubiquitin-protein ligases , Ubiquitination , Protéines virales , Animaux , Virus de la peste porcine africaine/métabolisme , Virus de la peste porcine africaine/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Suidae , I-kappa B Kinase/métabolisme , Ribonucléoprotéines/métabolisme , Ribonucléoprotéines/génétique , Protéines virales/métabolisme , Protéines virales/génétique , Peste porcine africaine/virologie , Peste porcine africaine/métabolisme , Humains , Cellules HEK293 , Interactions hôte-pathogène , Facteurs de virulence/métabolisme , Autophagie , Liaison aux protéines
7.
Biochem J ; 481(14): 959-980, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38941070

RÉSUMÉ

While IκB-kinase-ε (IKKε) induces immunomodulatory genes following viral stimuli, its up-regulation by inflammatory cytokines remains under-explored. Since airway epithelial cells respond to airborne insults and potentiate inflammation, IKKε expression was characterized in pulmonary epithelial cell lines (A549, BEAS-2B) and primary human bronchial epithelial cells grown as submersion or differentiated air-liquid interface cultures. IKKε expression was up-regulated by the pro-inflammatory cytokines, interleukin-1ß (IL-1ß) and tumour necrosis factor-α (TNFα). Thus, mechanistic interrogations in A549 cells were used to demonstrate the NF-κB dependence of cytokine-induced IKKε. Furthermore, chromatin immunoprecipitation in A549 and BEAS-2B cells revealed robust recruitment of the NF-κB subunit, p65, to one 5' and two intronic regions within the IKKε locus (IKBKE). In addition, IL-1ß and TNFα induced strong RNA polymerase 2 recruitment to the 5' region, the first intron, and the transcription start site. Stable transfection of the p65-binding regions into A549 cells revealed IL-1ß- and TNFα-inducible reporter activity that required NF-κB, but was not repressed by glucocorticoid. While critical NF-κB motifs were identified in the 5' and downstream intronic regions, the first intronic region did not contain functional NF-κB motifs. Thus, IL-1ß- and TNFα-induced IKKε expression involves three NF-κB-binding regions, containing multiple functional NF-κB motifs, and potentially other mechanisms of p65 binding through non-classical NF-κB binding motifs. By enhancing IKKε expression, IL-1ß may prime, or potentiate, responses to alternative stimuli, as modelled by IKKε phosphorylation induced by phorbol 12-myristate 13-acetate. However, since IKKε expression was only partially repressed by glucocorticoid, IKKε-dependent responses could contribute to glucocorticoid-resistant disease.


Sujet(s)
Cellules épithéliales , I-kappa B Kinase , Humains , I-kappa B Kinase/métabolisme , I-kappa B Kinase/génétique , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules A549 , Facteur de transcription RelA/métabolisme , Facteur de transcription RelA/génétique , Interleukine-1 bêta/pharmacologie , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/génétique , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/génétique , Facteur de nécrose tumorale alpha/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/génétique , Poumon/métabolisme , Poumon/cytologie , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/cytologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques
8.
Molecules ; 29(11)2024 Jun 02.
Article de Anglais | MEDLINE | ID: mdl-38893493

RÉSUMÉ

GSK-3ß, IKK-ß, and ROCK-1 kinases are implicated in the pathomechanism of Alzheimer's disease due to their involvement in the misfolding and accumulation of amyloid ß (Aß) and tau proteins, as well as inflammatory processes. Among these kinases, GSK-3ß plays the most crucial role. In this study, we present compound 62, a novel, remarkably potent, competitive GSK-3ß inhibitor (IC50 = 8 nM, Ki = 2 nM) that also exhibits additional ROCK-1 inhibitory activity (IC50 = 2.3 µM) and demonstrates anti-inflammatory and neuroprotective properties. Compound 62 effectively suppresses the production of nitric oxide (NO) and pro-inflammatory cytokines in the lipopolysaccharide-induced model of inflammation in the microglial BV-2 cell line. Furthermore, it shows neuroprotective effects in an okadaic-acid-induced tau hyperphosphorylation cell model of neurodegeneration. The compound also demonstrates the potential for further development, characterized by its chemical and metabolic stability in mouse microsomes and fair solubility.


Sujet(s)
Maladie d'Alzheimer , Glycogen synthase kinase 3 beta , I-kappa B Kinase , Thiazoles , rho-Associated Kinases , Protéines tau , Protéines tau/métabolisme , Glycogen synthase kinase 3 beta/antagonistes et inhibiteurs , Glycogen synthase kinase 3 beta/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Animaux , Thiazoles/pharmacologie , Thiazoles/composition chimique , Humains , rho-Associated Kinases/antagonistes et inhibiteurs , rho-Associated Kinases/métabolisme , Souris , I-kappa B Kinase/métabolisme , I-kappa B Kinase/antagonistes et inhibiteurs , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Lignée cellulaire , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/composition chimique , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Monoxyde d'azote/métabolisme , Lipopolysaccharides , Agrégats de protéines/effets des médicaments et des substances chimiques , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/métabolisme
9.
Mol Cell ; 84(13): 2436-2454.e10, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38925114

RÉSUMÉ

Signal transduction proteins containing a pLxIS motif induce interferon (IFN) responses central to antiviral immunity. Apart from their established roles in activating the IFN regulator factor (IRF) transcription factors, the existence of additional pathways and functions associated with the pLxIS motif is unknown. Using a synthetic biology-based platform, we identified two orphan pLxIS-containing proteins that stimulate IFN responses independent of all known pattern-recognition receptor pathways. We further uncovered a diversity of pLxIS signaling mechanisms, where the pLxIS motif represents one component of a multi-motif signaling entity, which has variable functions in activating IRF3, the TRAF6 ubiquitin ligase, IκB kinases, mitogen-activated protein kinases, and metabolic activities. The most diverse pLxIS signaling mechanisms were associated with the highest antiviral activities in human cells. The flexibility of domains that regulate IFN signaling may explain their prevalence in nature.


Sujet(s)
Facteur-3 de régulation d'interféron , Interférons , Transduction du signal , Facteur-6 associé aux récepteurs de TNF , Humains , Interférons/métabolisme , Cellules HEK293 , Facteur-3 de régulation d'interféron/métabolisme , Facteur-3 de régulation d'interféron/génétique , Facteur-6 associé aux récepteurs de TNF/métabolisme , Facteur-6 associé aux récepteurs de TNF/génétique , I-kappa B Kinase/métabolisme , I-kappa B Kinase/génétique , Domaines protéiques , Animaux , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Motifs d'acides aminés , Mitogen-Activated Protein Kinases/métabolisme
10.
Nat Immunol ; 25(7): 1193-1206, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38834865

RÉSUMÉ

Immune cells experience large cell shape changes during environmental patrolling because of the physical constraints that they encounter while migrating through tissues. These cells can adapt to such deformation events using dedicated shape-sensing pathways. However, how shape sensing affects immune cell function is mostly unknown. Here, we identify a shape-sensing mechanism that increases the expression of the chemokine receptor CCR7 and guides dendritic cell migration from peripheral tissues to lymph nodes at steady state. This mechanism relies on the lipid metabolism enzyme cPLA2, requires nuclear envelope tensioning and is finely tuned by the ARP2/3 actin nucleation complex. We also show that this shape-sensing axis reprograms dendritic cell transcription by activating an IKKß-NF-κB-dependent pathway known to control their tolerogenic potential. These results indicate that cell shape changes experienced by immune cells can define their migratory behavior and immunoregulatory properties and reveal a contribution of the physical properties of tissues to adaptive immunity.


Sujet(s)
Mouvement cellulaire , Cellules dendritiques , Homéostasie , Noeuds lymphatiques , Souris de lignée C57BL , Récepteurs CCR7 , Animaux , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/cytologie , Récepteurs CCR7/métabolisme , Souris , Mouvement cellulaire/immunologie , Forme de la cellule , Facteur de transcription NF-kappa B/métabolisme , Souris knockout , Transduction du signal/immunologie , I-kappa B Kinase/métabolisme , Complexe Arp-2-3/métabolisme
11.
Math Biosci Eng ; 21(4): 5164-5180, 2024 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-38872531

RÉSUMÉ

B-cell acute lymphoblastic leukemia (B-ALL) is a malignant blood disorder, particularly detrimental to children and adolescents, with recurrent or unresponsive cases contributing significantly to cancer-associated fatalities. IKBKE, associated with innate immunity, tumor promotion, and drug resistance, remains poorly understood in the context of B-ALL. Thus, this research aimed to explore the impact of the IKBKE inhibitor MCCK1 on B-ALL cells. The study encompassed diverse experiments, including clinical samples, in vitro and in vivo investigations. Quantitative real-time fluorescence PCR and protein blotting revealed heightened IKBKE mRNA and protein expression in B-ALL patients. Subsequent in vitro experiments with B-ALL cell lines demonstrated that MCCK1 treatment resulted in reduced cell viability and survival rates, with flow cytometry indicating cell cycle arrest. In vivo experiments using B-ALL mouse tumor models substantiated MCCK1's efficacy in impeding tumor proliferation. These findings collectively suggest that IKBKE, found to be elevated in B-ALL patients, may serve as a promising drug target, with MCCK1 demonstrating potential for inducing apoptosis in B-ALL cells both in vitro and in vivo.


Sujet(s)
Apoptose , Prolifération cellulaire , I-kappa B Kinase , Animaux , Humains , Souris , I-kappa B Kinase/antagonistes et inhibiteurs , I-kappa B Kinase/métabolisme , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Survie cellulaire/effets des médicaments et des substances chimiques , Mâle , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie , Enfant , Adolescent , Tests d'activité antitumorale sur modèle de xénogreffe , Inhibiteurs de protéines kinases/pharmacologie
12.
J Biol Chem ; 300(6): 107384, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38762177

RÉSUMÉ

Antimicrobial resistance poses a serious threat to human health worldwide and its incidence continues to increase owing to the overuse of antibiotics and other factors. Macrolide antibiotics such as erythromycin (EM) have immunomodulatory effects in addition to their antibacterial activity. Long-term, low-dose administration of macrolides has shown clinical benefits in treating non-infectious inflammatory respiratory diseases. However, this practice may also increase the emergence of drug-resistant bacteria. In this study, we synthesized a series of EM derivatives, and screened them for two criteria: (i) lack of antibacterial activity and (ii) ability to suppress tumor necrosis factor-α (TNF-α) production in THP-1 cells stimulated with lipopolysaccharide. Among the 37 synthesized derivatives, we identified a novel 12-membered ring macrolide EM982 that lacked antibacterial activity against Staphylococcus aureus and suppressed the production of TNF-α and other cytokines. The effects of EM982 on Toll-like receptor 4 (TLR4) signaling were analyzed using a reporter assay and Western blotting. The reporter assay showed that EM982 suppressed the activation of transcription factors, NF-κB and/or activator protein 1 (AP-1), in HEK293 cells expressing human TLR4. Western blotting showed that EM982 inhibited the phosphorylation of both IκB kinase (IKK) ß and IκBα, which function upstream of NF-κB, whereas it did not affect the phosphorylation of p38 mitogen-activated protein kinase, extracellular signal-regulated kinase, and c-Jun N-terminal kinase, which act upstream of AP-1. These results suggest that EM982 suppresses cytokine production by inhibiting phosphorylation of IKKß and IκBα, resulting in the inactivation of NF-κB.


Sujet(s)
Cytokines , I-kappa B Kinase , Inhibiteur alpha de NF-KappaB , Humains , I-kappa B Kinase/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Inhibiteur alpha de NF-KappaB/métabolisme , Cytokines/métabolisme , Érythromycine/pharmacologie , Érythromycine/composition chimique , Cellules THP-1 , Facteur de nécrose tumorale alpha/métabolisme , Antibactériens/pharmacologie , Antibactériens/composition chimique , Macrolides/pharmacologie , Macrolides/composition chimique , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Staphylococcus aureus/effets des médicaments et des substances chimiques , Récepteur de type Toll-4/métabolisme
14.
J Virol ; 98(6): e0026824, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38775480

RÉSUMÉ

Enteroviruses are the causative agents associated with several human and animal diseases, posing a significant threat to human and animal health. As one of the host immune defense strategies, innate immunity plays a crucial role in defending against invading pathogens, where the host utilizes a variety of mechanisms to inhibit or eliminate the pathogen. Here, we report a new strategy for the host to repress enterovirus replication by the 78 kDa glucose-regulated protein (GRP78), also known as heat shock protein family A member 5 (HSPA5). The GRP78 recognizes the EV-encoded RNA-dependent RNA polymerases (RdRPs) 3D protein and interacts with the nuclear factor kappa B kinase complex (CHUK) and subunit beta gene (IKBKB) to facilitate the phosphorylation and nuclear translocation of NF-κB, which induces the production of inflammatory factors and leads to a broad inhibition of enterovirus replication. These findings demonstrate a new role of GRP78 in regulating host innate immunity in response to viral infection and provide new insights into the mechanism underlying enterovirus replication and NF-κB activation.IMPORTANCEGRP78 is known as a molecular chaperone for protein folding and plays a critical role in maintaining protein folding and participating in cell proliferation, cell survival, apoptosis, and metabolism. However, the functions of GRP78 to participate in enterovirus genome replication and innate immune responses are rarely documented. In this study, we explored the functions of the EV-3D-interacting protein GRP78 and found that GRP78 inhibits enterovirus replication by activating NF-κB through binding to EV-F 3D and interacting with the NF-κB signaling molecules CHUK/IKBKB. This is the first report that GRP78 interacts with CHUK/IKBKB to activate the NF-κB signaling pathway, which leads to the expression of the proinflammatory cytokines and inhibition of enterovirus replication. These results demonstrate a unique mechanism of virus replication regulation by GRP78 and provide insights into the prevention and treatment of viral infections.


Sujet(s)
Chaperonne BiP du réticulum endoplasmique , I-kappa B Kinase , Facteur de transcription NF-kappa B , Protéines virales , Réplication virale , Animaux , Humains , Chlorocebus aethiops , Chaperonne BiP du réticulum endoplasmique/métabolisme , Enterovirus/croissance et développement , Enterovirus/immunologie , Enterovirus/métabolisme , Enterovirus/physiologie , Infections à entérovirus/virologie , Infections à entérovirus/métabolisme , Infections à entérovirus/immunologie , Protéines du choc thermique/métabolisme , Cellules HEK293 , Interactions hôte-pathogène/immunologie , I-kappa B Kinase/métabolisme , Immunité innée , Médiateurs de l'inflammation/immunologie , Médiateurs de l'inflammation/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Phosphorylation , Liaison aux protéines , RNA replicase/métabolisme , Transduction du signal , Cellules Vero , Protéines virales/métabolisme
15.
Nat Commun ; 15(1): 4096, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750019

RÉSUMÉ

The presence of heterogeneity in responses to oncolytic virotherapy poses a barrier to clinical effectiveness, as resistance to this treatment can occur through the inhibition of viral spread within the tumor, potentially leading to treatment failures. Here we show that 4-octyl itaconate (4-OI), a chemical derivative of the Krebs cycle-derived metabolite itaconate, enhances oncolytic virotherapy with VSVΔ51 in various models including human and murine resistant cancer cell lines, three-dimensional (3D) patient-derived colon tumoroids and organotypic brain tumor slices. Furthermore, 4-OI in combination with VSVΔ51 improves therapeutic outcomes in a resistant murine colon tumor model. Mechanistically, we find that 4-OI suppresses antiviral immunity in cancer cells through the modification of cysteine residues in MAVS and IKKß independently of the NRF2/KEAP1 axis. We propose that the combination of a metabolite-derived drug with an oncolytic virus agent can greatly improve anticancer therapeutic outcomes by direct interference with the type I IFN and NF-κB-mediated antiviral responses.


Sujet(s)
Thérapie virale de cancers , Virus oncolytiques , Succinates , Animaux , Humains , Thérapie virale de cancers/méthodes , Succinates/pharmacologie , Souris , Lignée cellulaire tumorale , Interféron de type I/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Tumeurs du côlon/thérapie , Tumeurs du côlon/immunologie , Tumeurs du côlon/traitement médicamenteux , Antiviraux/pharmacologie , Facteur de transcription NF-kappa B/métabolisme , I-kappa B Kinase/métabolisme , Protéine-1 de type kelch associée à ECH/métabolisme , Inflammation/traitement médicamenteux , Femelle , Virus de la stomatite vésiculeuse de type Indiana/physiologie , Virus de la stomatite vésiculeuse de type Indiana/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques
16.
Cell Biol Int ; 48(8): 1138-1147, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38769645

RÉSUMÉ

Asthma is an inflammatory disease. Airway epithelial cell pyroptosis and cytokine secretion promote asthma progression. Tripartite motif 47 (TRIM47) belongs to the E3 ubiquitin ligase family and is associated with apoptosis and inflammation in a range of diseases. However, the role of TRIM47 in asthma has not been explored. In this study, the human bronchial epithelial cell line BEAS-2B was treated with house dust mite (HDM) and TRIM47 expression was detected by RT-qPCR and Western blot. After transfection with TRIM47 interfering and overexpressing plasmids, the synthesis and secretion of cytokines, as well as pyroptosis-related indicators, were examined. Nuclear factor kappa-B (NF-κB) pathway proteins and nod-like receptor protein 3 (NLRP3) inflammasome were measured to explore the mechanism of TRIM47 action. In addition, the effect of TRIM47 on the level of NF-κB essential modulator (NEMO) ubiquitination was detected by an immunoprecipitation assay. The results showed that TRIM47 was upregulated in HDM-induced BEAS-2B cells and that TRIM47 mediated HDM-induced BEAS-2B cell pyroptosis and cytokine secretion. Mechanistically, TRIM47 promoted the K63-linked ubiquitination of NEMO and facilitated NF-κB/NLRP3 pathway activation. In conclusion, TRIM47 may promote cytokine secretion mediating inflammation and pyroptosis in bronchial epithelial cells by activating the NF-κB/NLRP3 pathway. Therefore, TRIM47 may be a potential therapeutic target for HDM-induced asthma.


Sujet(s)
Bronches , Cellules épithéliales , Facteur de transcription NF-kappa B , Protéine-3 de la famille des NLR contenant un domaine pyrine , Pyroptose , Transduction du signal , Ubiquitination , Humains , Facteur de transcription NF-kappa B/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Cellules épithéliales/métabolisme , Bronches/métabolisme , Bronches/anatomopathologie , Animaux , Lignée cellulaire , I-kappa B Kinase/métabolisme , Pyroglyphidae , Asthme/métabolisme , Asthme/anatomopathologie , Inflammasomes/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Cytokines/métabolisme , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique
17.
Front Immunol ; 15: 1375168, 2024.
Article de Anglais | MEDLINE | ID: mdl-38690287

RÉSUMÉ

Human T-cell leukemia virus type 1 (HTLV-1) is the etiological agent of adult T-cell leukemia/lymphoma. The HTLV-1 Tax constitutively activates nuclear factor-κB (NF-κB) to promote the survival and transformation of HTLV-1-infected T cells. Despite extensive study of Tax, how Tax interacts with host factors to regulate NF-κB activation and HTLV-1-driven cell proliferation is not entirely clear. Here, we showed that overexpression of Poly (rC)-binding protein 1 (PCBP1) promoted Tax-mediated IκB kinase (IKK)-NF-κB signaling activation, whereas knockdown of PCBP1 attenuated Tax-dependent IKK-NF-κB activation. However, Tax activation of HTLV-1 long terminal repeat was unaffected by PCBP1. Furthermore, depletion of PCBP1 led to apoptosis and reduced proliferation of HTLV-1-transformed cells. Mechanistically, PCBP1 interacted and co-localized with Tax in the cytoplasm, and PCBP1 KH3 domain was indispensable for the interaction between PCBP1 and Tax. Moreover, PCBP1 facilitated the assembly of Tax/IKK complex. Collectively, our results demonstrated that PCBP1 may exert an essential effect in Tax/IKK complex combination and subsequent NF-κB activation, which provides a novel insight into the pathogenetic mechanisms of HTLV-1.


Sujet(s)
Protéines de liaison à l'ADN , Produits du gène tax , Ribonucléoprotéines nucléaires hétérogènes , Virus T-lymphotrope humain de type 1 , Facteur de transcription NF-kappa B , Protéines de liaison à l'ARN , Humains , Produits du gène tax/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Virus T-lymphotrope humain de type 1/physiologie , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Transduction du signal , Cellules HEK293 , Liaison aux protéines , Prolifération cellulaire , Infections à HTLV-I/métabolisme , Infections à HTLV-I/virologie , Apoptose , I-kappa B Kinase/métabolisme , Interactions hôte-pathogène
18.
Cells ; 13(9)2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38727309

RÉSUMÉ

The activation of endothelial cells is crucial for immune defense mechanisms but also plays a role in the development of atherosclerosis. We have previously shown that inflammatory stimulation of endothelial cells on top of elevated lipoprotein/cholesterol levels accelerates atherogenesis. The aim of the current study was to investigate how chronic endothelial inflammation changes the aortic transcriptome of mice at normal lipoprotein levels and to compare this to the inflammatory response of isolated endothelial cells in vitro. We applied a mouse model expressing constitutive active IκB kinase 2 (caIKK2)-the key activator of the inflammatory NF-κB pathway-specifically in arterial endothelial cells and analyzed transcriptomic changes in whole aortas, followed by pathway and network analyses. We found an upregulation of cell death and mitochondrial beta-oxidation pathways with a predicted increase in endothelial apoptosis and necrosis and a simultaneous reduction in protein synthesis genes. The highest upregulated gene was ACE2, the SARS-CoV-2 receptor, which is also an important regulator of blood pressure. Analysis of isolated human arterial and venous endothelial cells supported these findings and also revealed a reduction in DNA replication, as well as repair mechanisms, in line with the notion that chronic inflammation contributes to endothelial dysfunction.


Sujet(s)
Cholestérol , Cellules endothéliales , Inflammation , Animaux , Humains , Cellules endothéliales/métabolisme , Souris , Inflammation/anatomopathologie , Inflammation/métabolisme , Cholestérol/métabolisme , Lipoprotéines/métabolisme , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Artères/métabolisme , Artères/anatomopathologie , Transcriptome/génétique , Aorte/métabolisme , Aorte/anatomopathologie , Souris de lignée C57BL , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , I-kappa B Kinase/métabolisme , Mâle , Facteur de transcription NF-kappa B/métabolisme
19.
Immunity ; 57(5): 929-932, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38749393

RÉSUMÉ

The ubiquitin-binding endoribonuclease N4BP1 is a critical immunosuppressor, but the mechanism by which it acts to constrain TLR-induced inflammatory cytokine production has remained unclear. In this issue of Immunity, Gitlin et al. find that N4BP1 works in concert with the non-canonical IκB kinase (IKK) to limit activity of the IKK complex.


Sujet(s)
I-kappa B Kinase , Humains , I-kappa B Kinase/métabolisme , Animaux , Endoribonucleases/métabolisme , Transduction du signal/immunologie , Cytokines/métabolisme
20.
Immunity ; 57(5): 973-986.e7, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38697117

RÉSUMÉ

The ubiquitin-binding endoribonuclease N4BP1 potently suppresses cytokine production by Toll-like receptors (TLRs) that signal through the adaptor MyD88 but is inactivated via caspase-8-mediated cleavage downstream of death receptors, TLR3, or TLR4. Here, we examined the mechanism whereby N4BP1 limits inflammatory responses. In macrophages, deletion of N4BP1 prolonged activation of inflammatory gene transcription at late time points after TRIF-independent TLR activation. Optimal suppression of inflammatory cytokines by N4BP1 depended on its ability to bind polyubiquitin chains, as macrophages and mice-bearing inactivating mutations in a ubiquitin-binding motif in N4BP1 displayed increased TLR-induced cytokine production. Deletion of the noncanonical IκB kinases (ncIKKs), Tbk1 and Ikke, or their adaptor Tank phenocopied N4bp1 deficiency and enhanced macrophage responses to TLR1/2, TLR7, or TLR9 stimulation. Mechanistically, N4BP1 acted in concert with the ncIKKs to limit the duration of canonical IκB kinase (IKKα/ß) signaling. Thus, N4BP1 and the ncIKKs serve as an important checkpoint against over-exuberant innate immune responses.


Sujet(s)
Endoribonucleases , I-kappa B Kinase , Inflammation , Macrophages , Souris knockout , Protein-Serine-Threonine Kinases , Transduction du signal , Récepteurs de type Toll , Animaux , Souris , Inflammation/immunologie , Inflammation/métabolisme , Récepteurs de type Toll/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , I-kappa B Kinase/métabolisme , I-kappa B Kinase/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Endoribonucleases/métabolisme , Endoribonucleases/génétique , Ubiquitine/métabolisme , Cytokines/métabolisme , Souris de lignée C57BL , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...