Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.523
Filtrer
1.
Mol Med Rep ; 30(3)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38963029

RÉSUMÉ

Viral infections in the respiratory tract are common, and, in recent years, severe acute respiratory syndrome coronavirus 2 outbreaks have highlighted the effect of viral infections on antiviral innate immune and inflammatory reactions. Specific treatments for numerous viral respiratory infections have not yet been established and they are mainly treated symptomatically. Therefore, understanding the details of the innate immune system underlying the airway epithelium is crucial for the development of new therapies. The present study aimed to investigate the function and expression of interferon (IFN)­stimulated gene (ISG)60 in non­cancerous bronchial epithelial BEAS­2B cells exposed to a Toll­like receptor 3 agonist. BEAS­2B cells were treated with a synthetic TLR3 ligand, polyinosinic­polycytidylic acid (poly IC). The mRNA and protein expression levels of ISG60 were analyzed using reverse transcription­quantitative PCR and western blotting, respectively. The levels of C­X­C motif chemokine ligand 10 (CXCL10) were examined using an enzyme­linked immunosorbent assay, and the effects of knockdown of IFN­ß, ISG60 and ISG56 were examined using specific small interfering RNAs. Notably, ISG60 expression was increased in proportion to poly IC concentration, and recombinant human IFN­ß also induced ISG60 expression. By contrast, knockdown of IFN­ß and ISG56 decreased ISG60 expression, and ISG60 knockdown reduced CXCL10 and ISG56 expression. These findings suggested that ISG60 is partly implicated in CXCL10 expression and that ISG60 may serve a role in the innate immune response of bronchial epithelial cells. The present study highlights ISG60 as a potential target for new therapeutic strategies against viral infections in the airway.


Sujet(s)
Bronches , Chimiokine CXCL10 , Cellules épithéliales , Poly I-C , Transduction du signal , Récepteur de type Toll-3 , Humains , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/génétique , Chimiokine CXCL10/métabolisme , Chimiokine CXCL10/génétique , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Bronches/cytologie , Bronches/métabolisme , Poly I-C/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Lignée cellulaire , Immunité innée , Interféron bêta/métabolisme , Interféron bêta/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines de liaison à l'ARN , Protéines adaptatrices de la transduction du signal , Protéines régulatrices de l'apoptose
2.
Exp Biol Med (Maywood) ; 249: 10122, 2024.
Article de Anglais | MEDLINE | ID: mdl-38881847

RÉSUMÉ

Rheumatoid fibroblast-like synoviocytes (RFLS) have an important role in the inflammatory pathogenesis of rheumatoid arthritis (RA). Toll-like receptor 3 (TLR3) is upregulated in RFLS; its activation leads to the production of interferon-ß (IFN-ß), a type I IFN. IFN-stimulated gene 56 (ISG56) is induced by IFN and is involved in innate immune responses; however, its role in RA remains unknown. Therefore, the purpose of this study was to investigate the role of TLR3-induced ISG56 in human RFLS. RFLS were treated with polyinosinic-polycytidylic acid (poly I:C), which served as a TLR3 ligand. ISG56, melanoma differentiation-associated gene 5 (MDA5), and C-X-C motif chemokine ligand 10 (CXCL10) expression were measured using quantitative reverse transcription-polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assay. Using immunohistochemistry, we found that ISG56 was expressed in synovial tissues of patients with RA and osteoarthritis. Under poly I:C treatment, ISG56 was upregulated in RFLS. In addition, we found that the type I IFN-neutralizing antibody mixture suppressed ISG56 expression. ISG56 knockdown decreased CXCL10 expression and MDA5 knockdown decreased ISG56 expression. In addition, we found that ISG56 was strongly expressed in the synovial cells of patients with RA. TLR3 signaling induced ISG56 expression in RFLS and type I IFN was involved in ISG56 expression. ISG56 was also found to be associated with CXCL10 expression, suggesting that ISG56 may be involved in TLR3/type I IFN/CXCL10 axis, and play a role in RA synovial inflammation.


Sujet(s)
Polyarthrite rhumatoïde , Chimiokine CXCL10 , Poly I-C , Transduction du signal , Cellules synoviales , Récepteur de type Toll-3 , Humains , Récepteur de type Toll-3/métabolisme , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Poly I-C/pharmacologie , Cellules synoviales/métabolisme , Chimiokine CXCL10/métabolisme , Hélicase IFIH1 inductrice de l'interféron/métabolisme , Hélicase IFIH1 inductrice de l'interféron/génétique , Cellules cultivées , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Protéines adaptatrices du transport vésiculaire/métabolisme , Protéines adaptatrices du transport vésiculaire/génétique , Protéines de liaison à l'ARN , Protéines adaptatrices de la transduction du signal , Protéines régulatrices de l'apoptose
3.
Biol Pharm Bull ; 47(5): 946-954, 2024.
Article de Anglais | MEDLINE | ID: mdl-38735732

RÉSUMÉ

There is accumulating evidence that selective serotonin reuptake inhibitors (SSRIs), clinically used as antidepressants, have a beneficial effect on inflammatory diseases such as coronavirus disease 2019 (COVID-19). We previously compared the inhibitory effects of five U.S. Food and Drug Administration (FDA)-approved SSRIs on the production of an inflammatory cytokine, interleukin-6 (IL-6), and concluded that fluoxetine (FLX) showed the most potent anti-inflammatory activity. Here, we investigated the structure-activity relationship of FLX for anti-inflammatory activity towards J774.1 murine macrophages. FLX suppressed IL-6 production induced by the TLR3 agonist polyinosinic-polycytidylic acid (poly(I : C)) with an IC50 of 4.76 µM. A derivative of FLX containing chlorine instead of the methylamino group lacked activity, suggesting that the methylamino group is important for the anti-inflammatory activity. FLX derivatives bearing an N-propyl or N-(pyridin-3-yl)methyl group in place of the N-methyl group exhibited almost the same activity as FLX. Other derivatives showed weaker activity, and the N-phenyl and N-(4-trifluoromethyl)benzyl derivatives were inactive. The chlorine-containing derivative also lacked inhibitory activity against TLR9- or TLR4-mediated IL-6 production. These derivatives showed similar structure-activity relationships for TLR3- and TLR9-mediated inflammatory responses. However, the activities of all amino group-containing derivatives against the TLR4-mediated inflammatory response were equal to or higher than the activity of FLX. These results indicate that the substituent at the nitrogen atom in FLX strongly influences the anti-inflammatory effect.


Sujet(s)
Anti-inflammatoires , Fluoxétine , Interleukine-6 , Relation structure-activité , Animaux , Fluoxétine/pharmacologie , Souris , Interleukine-6/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/composition chimique , Lignée cellulaire , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Cytokines/métabolisme , Récepteur de type Toll-3/métabolisme , Poly I-C/pharmacologie , Inbiteurs sélectifs de la recapture de la sérotonine/pharmacologie , Inbiteurs sélectifs de la recapture de la sérotonine/composition chimique , Inflammation/traitement médicamenteux
4.
Nat Commun ; 15(1): 3969, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38730242

RÉSUMÉ

Encephalitis is a rare and potentially fatal manifestation of herpes simplex type 1 infection. Following genome-wide genetic analyses, we identified a previously uncharacterized and very rare heterozygous variant in the E3 ubiquitin ligase WWP2, in a 14-month-old girl with herpes simplex encephalitis. The p.R841H variant (NM_007014.4:c.2522G > A) impaired TLR3 mediated signaling in inducible pluripotent stem cells-derived neural precursor cells and neurons; cells bearing this mutation were also more susceptible to HSV-1 infection compared to control cells. The p.R841H variant increased TRIF ubiquitination in vitro. Antiviral immunity was rescued following the correction of p.R841H by CRISPR-Cas9 technology. Moreover, the introduction of p.R841H in wild type cells reduced such immunity, suggesting that this mutation is linked to the observed phenotypes.


Sujet(s)
Encéphalite à herpès simplex , Herpèsvirus humain de type 1 , Mutation , Ubiquitin-protein ligases , Humains , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Femelle , Encéphalite à herpès simplex/génétique , Nourrisson , Herpèsvirus humain de type 1/génétique , Cellules souches pluripotentes induites/métabolisme , Récepteur de type Toll-3/génétique , Récepteur de type Toll-3/métabolisme , Ubiquitination , Neurones/métabolisme , Cellules souches neurales/métabolisme , Cellules souches neurales/virologie , Systèmes CRISPR-Cas
5.
Nat Commun ; 15(1): 4099, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38816352

RÉSUMÉ

Chronic inflammation is a major cause of cancer worldwide. Interleukin 33 (IL-33) is a critical initiator of cancer-prone chronic inflammation; however, its induction mechanism by environmental causes of chronic inflammation is unknown. Herein, we demonstrate that Toll-like receptor (TLR)3/4-TBK1-IRF3 pathway activation links environmental insults to IL-33 induction in the skin and pancreas inflammation. An FDA-approved drug library screen identifies pitavastatin to effectively suppress IL-33 expression by blocking TBK1 membrane recruitment/activation through the mevalonate pathway inhibition. Accordingly, pitavastatin prevents chronic pancreatitis and its cancer sequela in an IL-33-dependent manner. The IRF3-IL-33 axis is highly active in chronic pancreatitis and its associated pancreatic cancer in humans. Interestingly, pitavastatin use correlates with a significantly reduced risk of chronic pancreatitis and pancreatic cancer in patients. Our findings demonstrate that blocking the TBK1-IRF3-IL-33 signaling axis suppresses cancer-prone chronic inflammation. Statins present a safe and effective prophylactic strategy to prevent chronic inflammation and its cancer sequela.


Sujet(s)
Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase , Facteur-3 de régulation d'interféron , Interleukine-33 , Tumeurs du pancréas , Protein-Serine-Threonine Kinases , Quinoléines , Transduction du signal , Interleukine-33/métabolisme , Animaux , Facteur-3 de régulation d'interféron/métabolisme , Humains , Tumeurs du pancréas/prévention et contrôle , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/usage thérapeutique , Souris , Protein-Serine-Threonine Kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Quinoléines/pharmacologie , Quinoléines/usage thérapeutique , Inflammation/prévention et contrôle , Inflammation/métabolisme , Pancréatite chronique/prévention et contrôle , Pancréatite chronique/métabolisme , Récepteur de type Toll-3/métabolisme , Souris de lignée C57BL , Récepteur de type Toll-4/métabolisme , Acide mévalonique/métabolisme , Mâle , Femelle , Souris knockout
6.
Int Immunopharmacol ; 134: 112182, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38703568

RÉSUMÉ

Seipin plays a crucial role in lipid metabolism and is involved in neurological disorders. However, the function and mechanism of action of seipin in acute ischemic stroke have not yet been elucidated. Here, we aimed to investigate the effect of seipin on neuroinflammation induced by oxygen-glucose deprivation/reoxygenation (OGD/R) and further explore the molecular mechanism by functional experiments. Our results revealed a significant decrease in seipin mRNA levels, accompanied by enhanced expression of TNF-α in patients with AIS, and a significant negative correlation between seipin and TNF-α was observed. Additionally, there was a negative correlation between seipin levels and the National Institutes of Health Stroke Scale (NIHSS) score. Furthermore, seipin levels were also decreased in middle cerebral artery occlusion/reperfusion (MCAO/R) mice and OGD/R-treated BV2 cells. RNA sequencing analysis showed that seipin knockdown altered the Toll-like receptor 3 (TLR3) signaling pathway. It was further confirmed in vitro that seipin knockdown caused significantly increased secretion of inflammatory factors including TNF-α, interleukin (IL)-1ß, and interferon (IFN)-ß. Meanwhile, seipin knockdown activated the Tlr3 signal pathway while this effect could be reversed by Tlr3 inhibitor in OGD/R treated BV2 cells. Furthermore, neuroinflammation induced by OGD/R was significantly reduced by seipin overexpression. Overall, our study demonstrate that seipin deficiency aggravates neuroinflammation by activating the TLR3/TRAF3/NF-κB signaling pathway after OGD/R stimuli, and suggest that seipin may be a potential therapeutic target for AIS.


Sujet(s)
Glucose , Facteur de transcription NF-kappa B , Maladies neuro-inflammatoires , Oxygène , Transduction du signal , Facteur-3 associé aux récepteurs de TNF , Récepteur de type Toll-3 , Sujet âgé , Animaux , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , Lignée cellulaire , Modèles animaux de maladie humaine , Glucose/métabolisme , Sous-unités gamma des protéines G/métabolisme , Sous-unités gamma des protéines G/génétique , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/immunologie , Accident vasculaire cérébral ischémique/métabolisme , Accident vasculaire cérébral ischémique/immunologie , Souris de lignée C57BL , Maladies neuro-inflammatoires/immunologie , Maladies neuro-inflammatoires/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Oxygène/métabolisme , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/immunologie , Facteur-3 associé aux récepteurs de TNF/métabolisme , Facteur-3 associé aux récepteurs de TNF/génétique , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/génétique
7.
Molecules ; 29(9)2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38731436

RÉSUMÉ

In our research, we explored a natural substance called Oxymatrine, found in a traditional Chinese medicinal plant, to fight against a common bird flu virus known as H9N2. This virus not only affects birds but can also pose a threat to human health. We focused on how this natural compound can help in stopping the virus from spreading in cells that line the lungs of birds and potentially humans. Our findings show that Oxymatrine can both directly block the virus and boost the body's immune response against it. This dual-action mechanism is particularly interesting because it indicates that Oxymatrine might be a useful tool in developing new ways to prevent and treat this type of bird flu. Understanding how Oxymatrine works against the H9N2 virus could lead to safer and more natural ways to combat viral infections in animals and humans, contributing to the health and well-being of society. The H9N2 Avian Influenza Virus (AIV) is a persistent health threat because of its rapid mutation rate and the limited efficacy of vaccines, underscoring the urgent need for innovative therapies. This study investigated the H9N2 AIV antiviral properties of Oxymatrine (OMT), a compound derived from traditional Chinese medicine, particularly focusing on its interaction with pulmonary microvascular endothelial cells (PMVECs). Employing an array of in vitro assays, including 50% tissue culture infectious dose, Cell Counting Kit-8, reverse transcription-quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, and Western blot, we systematically elucidated the multifaceted effects of OMT. OMT dose-dependently inhibited critical antiviral proteins (PKR and Mx1) and modulated the expression of type I interferons and key cytokines (IFN-α, IFN-ß, IL-6, and TNF-α), thereby affecting TLR3 signaling and its downstream elements (NF-κB and IRF-3). OMT's antiviral efficacy extended beyond TLR3-mediated responses, suggesting its potential as a versatile antiviral agent. This study not only contributes to the growing body of research on the use of natural compounds as antiviral agents but also underscores the importance of further investigating the broader application of OMT for combating viral infections.


Sujet(s)
Antiviraux , Sous-type H9N2 du virus de la grippe A , Grippe chez les oiseaux , , Transduction du signal , Récepteur de type Toll-3 , Animaux , Chiens , Humains , Antiviraux/pharmacologie , Sous-type H9N2 du virus de la grippe A/effets des médicaments et des substances chimiques , Grippe chez les oiseaux/traitement médicamenteux , Grippe chez les oiseaux/immunologie , Cellules rénales canines Madin-Darby , Transduction du signal/effets des médicaments et des substances chimiques , Récepteur de type Toll-3/métabolisme
8.
EMBO Mol Med ; 16(5): 1193-1219, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38671318

RÉSUMÉ

Radiotherapy (RT) has been reported to induce abscopal effect in advanced hepatocellular carcinoma (HCC), but such phenomenon was only observed in sporadic cases. Here, we demonstrated that subcutaneous administration of Toll-like receptor 3 (TLR3) agonist poly(I:C) could strengthen the abscopal effect during RT through activating tumor cell ferroptosis signals in bilateral HCC subcutaneous tumor mouse models, which could be significantly abolished by TLR3 knock-out or ferroptosis inhibitor ferrostatin-1. Moreover, poly(I:C) could promote the presentation of tumor neoantigens by dendritic cells to enhance the recruitment of activated CD8+ T cells into distant tumor tissues for inducing tumor cell ferroptosis during RT treatment. Finally, the safety and feasibility of combining poly(I:C) with RT for treating advanced HCC patients were further verified in a prospective clinical trial. Thus, enhancing TLR3 signaling activation during RT could provide a novel strategy for strengthening abscopal effect to improve the clinical benefits of advanced HCC patients.


Sujet(s)
Carcinome hépatocellulaire , Ferroptose , Tumeurs du foie , Poly I-C , Récepteur de type Toll-3 , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/agonistes , Animaux , Carcinome hépatocellulaire/radiothérapie , Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/radiothérapie , Tumeurs du foie/anatomopathologie , Humains , Souris , Poly I-C/pharmacologie , Mâle , Femelle , Lignée cellulaire tumorale , Souris de lignée C57BL , Modèles animaux de maladie humaine , Souris knockout , Adulte d'âge moyen
9.
Biochem Biophys Res Commun ; 712-713: 149915, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38663038

RÉSUMÉ

Viral infections pose a significant threat to public health, and the production of interferons represents one of the most critical antiviral innate immune responses of the host. Consequently, the screening and identification of compounds or reagents that induce interferon production are of paramount importance. This study commenced with the cultivation of host bacterium 15,597, followed by the infection of Escherichia coli with the MS2 bacteriophage. Utilizing the J2 capture technique, a class of dsRNA mixtures (MS2+15,597) was isolated from the E. coli infected with the MS2 bacteriophage. Subsequent investigations were conducted on the immunostimulatory activity of the MS2+15,597 mixture. The results indicated that the dsRNA mixtures (MS2+15,597) extracted from E. coli infected with the MS2 bacteriophage possess the capability to activate innate immunity, thereby inducing the production of interferon-ß. These dsRNA mixtures can activate the RIG-I and TLR3 pattern recognition receptors, stimulating the expression of interferon stimulatory factors 3/7, which in turn triggers the NF-κB signaling pathway, culminating in the cellular production of interferon-ß to achieve antiviral effects. This study offers novel insights and strategies for the development of broad-spectrum antiviral drugs, potentially providing new modalities for future antiviral therapies.


Sujet(s)
Escherichia coli , Levivirus , ARN double brin , Escherichia coli/virologie , Escherichia coli/génétique , Escherichia coli/métabolisme , ARN double brin/métabolisme , Humains , Levivirus/génétique , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/génétique , Immunité innée , Interféron bêta/métabolisme , Interféron bêta/génétique , Facteur de transcription NF-kappa B/métabolisme , Protéine-58 à domaine DEAD/métabolisme , Protéine-58 à domaine DEAD/génétique , Transduction du signal , Facteur-7 de régulation d'interféron/métabolisme , Facteur-7 de régulation d'interféron/génétique , Récepteurs immunologiques , Facteur-3 de régulation d'interféron/métabolisme , Facteur-3 de régulation d'interféron/génétique
10.
Fish Shellfish Immunol ; 149: 109581, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38670412

RÉSUMÉ

Deubiquitinating enzyme A (DUBA), a member of the ovarian tumor (OTU) subfamily of deubiquitinases (DUBs), is recognized for its negative regulatory role in type I interferon (IFN) expression downstream of Toll-like receptor 3 (TLR3). However, its involvement in the TLR3 signaling pathway in fish remains largely unexplored. In this study, we investigated the regulatory role of DUBA (OmDUBA) in the TLR3 response in rainbow trout (Oncorhynchus mykiss). OmDUBA features a conserved OTU domain, and its expression increased in RTH-149 cells following stimulation with the TLR3 agonist poly(I:C). Gain- and loss-of-function experiments demonstrated that OmDUBA attenuated the activation of TANK-binding kinase 1 (TBK1), resulting in a subsequent reduction in type I IFN expression and IFN-stimulated response element (ISRE) activation in poly(I:C)-stimulated cells. OmDUBA interacted with TRAF3, a crucial mediator in TLR3-mediated type I IFN production. Under poly(I:C) stimulation, there was an augmentation in the K63-linked polyubiquitination of TRAF3, a process significantly inhibited upon OmDUBA overexpression. These findings suggest that OmDUBA may function similarly to its mammalian counterparts in downregulating the poly(I:C)-induced type I IFN response in rainbow trout by removing the K63-linked ubiquitin chain on TRAF3. Our study provides novel insights into the role of fish DUBA in antiviral immunity.


Sujet(s)
Protéines de poisson , Interféron de type I , Oncorhynchus mykiss , Poly I-C , Transduction du signal , Facteur-3 associé aux récepteurs de TNF , Animaux , Oncorhynchus mykiss/immunologie , Facteur-3 associé aux récepteurs de TNF/génétique , Facteur-3 associé aux récepteurs de TNF/métabolisme , Facteur-3 associé aux récepteurs de TNF/immunologie , Interféron de type I/immunologie , Interféron de type I/génétique , Interféron de type I/métabolisme , Protéines de poisson/génétique , Protéines de poisson/immunologie , Protéines de poisson/métabolisme , Transduction du signal/immunologie , Poly I-C/pharmacologie , Immunité innée , Régulation de l'expression des gènes/immunologie , Ubiquitination , Récepteur de type Toll-3/génétique , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/immunologie
11.
Mol Biol Rep ; 51(1): 550, 2024 Apr 20.
Article de Anglais | MEDLINE | ID: mdl-38642183

RÉSUMÉ

BACKGROUND: The therapeutic efficacy of intra-articular mesenchymal stem cells (MSCs) injection for patients with osteoarthritis (OA) currently exhibits inconsistency, and the underlying mechanism remains elusive. It has been postulated that the immunomodulatory properties and paracrine activity of MSCs might be influenced by the inflammatory micro-environment within osteoarthritic joints, potentially contributing to this observed inconsistency. METHODS: Adipose-derived MSCs (ADSCs) were isolated from SD rats and pre-treated with Toll-like receptor 3 (TLR3) agonist Poly I:C or Toll-like receptor 4 (TLR4) agonist LPS. The pre-treated ADSCs were then co-cultured with IL-1ß-induced osteoarthritic chondrocytes using a Transwell system to analyze the paracrine effect of ADSCs on reversing the osteoarthritic phenotype of chondrocytes. RESULTS: RT-PCR and Western blot analysis revealed that Poly I:C and LPS pre-treatments up-regulated the expression of IL-10 and IL-6 in ADSCs, respectively. Furthermore, only Poly I:C-preconditioned ADSCs significantly promoted proliferation while inhibiting apoptosis in IL-1ß-treated chondrocytes. Additionally, Poly I:C-preconditioned ADSCs downregulated MMP13 expression while upregulating aggrecan and collagen II expression levels in IL-1ß-treated chondrocytes. CONCLUSIONS: TLR3 activation polarizes ADSCs into an immunomodulatory phenotype distinct from TLR4 activation, exerting differential effects on reversing the osteoarthritic phenotype of chondrocytes; thus indicating that MSCs' paracrine effect regulated by TLRs signaling impacts the efficacy of intra-articular MSCs injection.


Sujet(s)
Chondrocytes , Cellules souches mésenchymateuses , Humains , Rats , Animaux , Chondrocytes/métabolisme , Récepteur de type Toll-4/métabolisme , Récepteur de type Toll-3/génétique , Récepteur de type Toll-3/métabolisme , Cellules cultivées , Lipopolysaccharides/pharmacologie , Lipopolysaccharides/métabolisme , Rat Sprague-Dawley , Cellules souches mésenchymateuses/métabolisme , Récepteurs de type Toll/métabolisme , Phénotype , Poly I/métabolisme , Poly I/pharmacologie
12.
ACS Nano ; 18(15): 10509-10526, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38564478

RÉSUMÉ

Systemic exposure to starch-coated iron oxide nanoparticles (IONPs) can stimulate antitumor T cell responses, even when little IONP is retained within the tumor. Here, we demonstrate in mouse models of metastatic breast cancer that IONPs can alter the host immune landscape, leading to systemic immune-mediated disease suppression. We report that a single intravenous injection of IONPs can inhibit primary tumor growth, suppress metastases, and extend survival. Gene expression analysis revealed the activation of Toll-like receptor (TLR) pathways involving signaling via Toll/Interleukin-1 receptor domain-containing adaptor-inducing IFN-ß (TRIF), a TLR pathway adaptor protein. Requisite participation of TRIF in suppressing tumor progression was demonstrated with histopathologic evidence of upregulated IFN-regulatory factor 3 (IRF3), a downstream protein, and confirmed in a TRIF knockout syngeneic mouse model of metastatic breast cancer. Neither starch-coated polystyrene nanoparticles lacking iron, nor iron-containing dextran-coated parenteral iron replacement agent, induced significant antitumor effects, suggesting a dependence on the type of IONP formulation. Analysis of multiple independent clinical databases supports a hypothesis that upregulation of TLR3 and IRF3 correlates with increased overall survival among breast cancer patients. Taken together, these data support a compelling rationale to re-examine IONP formulations as harboring anticancer immune (nano)adjuvant properties to generate a therapeutic benefit without requiring uptake by cancer cells.


Sujet(s)
Tumeurs du sein , Tumeurs du poumon , Animaux , Souris , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-4/métabolisme , Modèles animaux de maladie humaine , Tumeurs du poumon/traitement médicamenteux , Protéines adaptatrices du transport vésiculaire/génétique , Protéines adaptatrices du transport vésiculaire/métabolisme , Fer , Amidon , Nanoparticules magnétiques d'oxyde de fer
13.
Viruses ; 16(4)2024 04 17.
Article de Anglais | MEDLINE | ID: mdl-38675965

RÉSUMÉ

Epstein-Barr virus (EBV), a Herpesviridae family member, is associated with an increased risk of autoimmune disease development in the host. We previously demonstrated that EBV DNA elevates levels of the pro-inflammatory cytokine IL-17A and that inhibiting Toll-like receptor (TLR) 3, 7, or 9 reduces its levels. Moreover, this DNA exacerbated colitis in a mouse model of inflammatory bowel disease (IBD). In the study at hand, we examined whether inhibition of TLR3, 7, or 9 alleviates this exacerbation. Mice were fed 1.5% dextran sulfate sodium (DSS) water and administered EBV DNA. Then, they were treated with a TLR3, 7, or 9 inhibitor or left untreated. We also assessed the additive impact of combined inhibition of all three receptors. Mice that received DSS, EBV DNA, and each inhibitor alone, or a combination of inhibitors, showed significant improvement. They also had a decrease in the numbers of the pathogenic colonic IL-17A+IFN-γ+ foci. Inhibition of all three endosomal TLR receptors offered no additive benefit over administering a single inhibitor. Therefore, inhibition of endosomal TLRs reduces EBV DNA exacerbation of mouse colitis, offering a potential approach for managing IBD patients infected with EBV.


Sujet(s)
ADN viral , Herpèsvirus humain de type 4 , Maladies inflammatoires intestinales , Récepteurs de type Toll , Animaux , Femelle , Souris , Colite/induit chimiquement , Colite/traitement médicamenteux , Colite/virologie , Sulfate dextran , Modèles animaux de maladie humaine , ADN viral/effets indésirables , ADN viral/pharmacologie , Endosomes/effets des médicaments et des substances chimiques , Endosomes/métabolisme , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/complications , Infections à virus Epstein-Barr/traitement médicamenteux , Maladies inflammatoires intestinales/induit chimiquement , Maladies inflammatoires intestinales/traitement médicamenteux , Maladies inflammatoires intestinales/virologie , Interleukine-17/métabolisme , Souris de lignée C57BL , Récepteur de type Toll-3/antagonistes et inhibiteurs , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-7/antagonistes et inhibiteurs , Récepteur de type Toll-7/métabolisme , Récepteur-9 de type Toll-like/antagonistes et inhibiteurs , Récepteur-9 de type Toll-like/métabolisme , Récepteurs de type Toll/antagonistes et inhibiteurs , Récepteurs de type Toll/métabolisme
14.
Viruses ; 16(4)2024 04 20.
Article de Anglais | MEDLINE | ID: mdl-38675983

RÉSUMÉ

Human immunodeficiency virus type 1 (HIV-1) infection can result in HIV-associated neurocognitive disorder (HAND), a spectrum of disorders characterized by neurological impairment and chronic inflammation. Combined antiretroviral therapy (cART) has elicited a marked reduction in the number of individuals diagnosed with HAND. However, there is continual, low-level viral transcription due to the lack of a transcription inhibitor in cART regimens, which results in the accumulation of viral products within infected cells. To alleviate stress, infected cells can release accumulated products, such as TAR RNA, in extracellular vesicles (EVs), which can contribute to pathogenesis in neighboring cells. Here, we demonstrate that cART can contribute to autophagy deregulation in infected cells and increased EV release. The impact of EVs released from HIV-1 infected myeloid cells was found to contribute to CNS pathogenesis, potentially through EV-mediated TLR3 (Toll-like receptor 3) activation, suggesting the need for therapeutics to target this mechanism. Three HIV-1 TAR-binding compounds, 103FA, 111FA, and Ral HCl, were identified that recognize TAR RNA and reduce TLR activation. These data indicate that packaging of viral products into EVs, potentially exacerbated by antiretroviral therapeutics, may induce chronic inflammation of the CNS observed in cART-treated patients, and novel therapeutic strategies may be exploited to mitigate morbidity.


Sujet(s)
Autophagie , Vésicules extracellulaires , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Récepteur de type Toll-3 , Vésicules extracellulaires/métabolisme , Humains , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Infections à VIH/virologie , Infections à VIH/métabolisme , Infections à VIH/traitement médicamenteux , Autophagie/effets des médicaments et des substances chimiques , ARN viral/métabolisme , ARN viral/génétique
15.
J Pathol ; 263(2): 203-216, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38551071

RÉSUMÉ

Urothelial damage and barrier dysfunction emerge as the foremost mechanisms in Hunner-type interstitial cystitis/bladder pain syndrome (HIC). Although treatments aimed at urothelial regeneration and repair have been employed, their therapeutic effectiveness remains limited due to the inadequate understanding of specific cell types involved in damage and the lack of specific molecular targets within these mechanisms. Therefore, we harnessed single-cell RNA sequencing to elucidate the heterogeneity and developmental trajectory of urothelial cells within HIC bladders. Through reclustering, we identified eight distinct clusters of urothelial cells. There was a significant reduction in UPK3A+ umbrella cells and a simultaneous increase in progenitor-like pluripotent cells (PPCs) within the HIC bladder. Pseudotime analysis of the urothelial cells in the HIC bladder revealed that cells faced challenges in differentiating into UPK3A+ umbrella cells, while PPCs exhibited substantial proliferation to compensate for the loss of UPK3A+ umbrella cells. The urothelium in HIC remains unrepaired, despite the substantial proliferation of PPCs. Thus, we propose that inhibiting the pivotal signaling pathways responsible for the injury to UPK3A+ umbrella cells is paramount for restoring the urothelial barrier and alleviating lower urinary tract symptoms in HIC patients. Subsequently, we identified key molecular pathways (TLR3 and NR2F6) associated with the injury of UPK3A+ umbrella cells in HIC urothelium. Finally, we conducted in vitro and in vivo experiments to confirm the potential of the TLR3-NR2F6 axis as a promising therapeutic target for HIC. These findings hold the potential to inhibit urothelial injury, providing promising clues for early diagnosis and functional bladder self-repair strategies for HIC patients. © 2024 The Pathological Society of Great Britain and Ireland.


Sujet(s)
Cystite interstitielle , Récepteur de type Toll-3 , Urothélium , Animaux , Femelle , Humains , Souris , Différenciation cellulaire , Prolifération cellulaire , Cystite interstitielle/anatomopathologie , Cystite interstitielle/métabolisme , Cystite interstitielle/génétique , Souris de lignée C57BL , Transduction du signal , Analyse sur cellule unique , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/génétique , Vessie urinaire/anatomopathologie , Vessie urinaire/métabolisme , Urothélium/anatomopathologie , Urothélium/métabolisme
16.
Am J Physiol Lung Cell Mol Physiol ; 326(5): L618-L626, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38469627

RÉSUMÉ

Thymic stromal lymphopoietin (TSLP) is an epithelial-derived pleiotropic cytokine that regulates T-helper 2 (Th2) immune responses in the lung and plays a major role in severe uncontrolled asthma. Emerging evidence suggests a role for endoplasmic reticulum (ER) stress in the pathogenesis of asthma. In this study, we determined if ER stress and the unfolded protein response (UPR) signaling are involved in TSLP induction in the airway epithelium. For this, we treated human bronchial epithelial basal cells and differentiated primary bronchial epithelial cells with ER stress inducers and the TSLP mRNA and protein expression was determined. A series of siRNA gene knockdown experiments were conducted to determine the ER stress-induced TSLP signaling pathways. cDNA collected from asthmatic bronchial biopsies was used to determine the gene correlation between ER stress and TSLP. Our results show that ER stress signaling induces TSLP mRNA expression via the PERK-C/EBP homologous protein (CHOP) signaling pathway. AP-1 transcription factor is important in regulating this ER stress-induced TSLP mRNA induction, though ER stress alone cannot induce TSLP protein production. However, ER stress significantly enhances TLR3-induced TSLP protein secretion in the airway epithelium. TSLP and ER stress (PERK) mRNA expression positively correlates in bronchial biopsies from participants with asthma, particularly in neutrophilic asthma. In conclusion, these results suggest that ER stress primes TSLP that is then enhanced further upon TLR3 activation, which may induce severe asthma exacerbations. Targeting ER stress using pharmacological interventions may provide novel therapeutics for severe uncontrolled asthma.NEW & NOTEWORTHY TSLP is an epithelial-derived cytokine and a key regulator in the pathogenesis of severe uncontrolled asthma. We demonstrate a novel mechanism by which endoplasmic reticulum stress signaling upregulates airway epithelial TSLP mRNA expression via the PERK-CHOP signaling pathway and enhances TLR3-mediated TSLP protein secretion.


Sujet(s)
Asthme , Cytokines , Stress du réticulum endoplasmique , Cellules épithéliales , Lymphopoïétine stromale thymique , Récepteur de type Toll-3 , Réponse aux protéines mal repliées , Humains , Cytokines/métabolisme , Récepteur de type Toll-3/métabolisme , Récepteur de type Toll-3/génétique , Asthme/métabolisme , Asthme/anatomopathologie , Asthme/génétique , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Facteur de transcription CHOP/métabolisme , Facteur de transcription CHOP/génétique , Transduction du signal , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/anatomopathologie , Bronches/métabolisme , Bronches/anatomopathologie , eIF-2 Kinase/métabolisme , eIF-2 Kinase/génétique , Cellules cultivées , Femelle , ARN messager/génétique , ARN messager/métabolisme
17.
Shock ; 61(6): 905-914, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38526139

RÉSUMÉ

ABSTRACT: Currently, the coronavirus disease 2019 (COVID-19) is becoming a serious threat to human health worldwide. Therefore, there is a great need to develop effective drugs against viral pneumonia. Diammonium glycyrrhizinate (DG), derived from Glycyrrhiza glabra L., has been demonstrated with significant anti-inflammatory properties. However, the therapeutic effects and mechanisms of DG on pneumonia require further clarification. In this study, mice received intratracheal injection of polyinosinic-polycytidylic acid (poly(I:C)) to induce pneumonia and were treated with DG. First, we evaluated the therapeutic potential of DG on poly(I:C)-induced pneumonia. Second, the anti-inflammatory and antioxidative activities and the impact of DG on the toll-like receptor 3 (TLR3) pathway were investigated. Third, the mechanism of DG was analyzed through untargeted metabolomics techniques. Our results revealed that DG intervention decreased permeability and reduced abnormal lung alterations in poly(I:C)-induced pneumonia model mice. DG intervention also downregulated cytokine levels in bronchoalveolar lavage fluid. Moreover, DG treatment inhibited the activation of TLR3 pathway. Furthermore, untargeted metabolomics analysis revealed that DG intervention could modulate serum metabolites involved in amino and nucleotide sugar metabolism, fructose and mannose metabolism, tyrosine metabolism, and phenylalanine, tyrosine, and tryptophan biosynthesis pathways. In conclusion, our study showed that DG could ameliorate poly(I:C)-induced pneumonia by inactivating the TLR3 pathway and affecting amino and nucleotide sugar, fructose and mannose metabolism, as well as tryptophan, phenylalanine, and tyrosine biosynthesis.


Sujet(s)
Modèles animaux de maladie humaine , Acide glycyrrhizique , Poly I-C , Animaux , Souris , Acide glycyrrhizique/pharmacologie , Acide glycyrrhizique/usage thérapeutique , Mâle , Récepteur de type Toll-3/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Pneumopathie infectieuse/traitement médicamenteux , Pneumopathie infectieuse/métabolisme , Pneumopathie infectieuse/induit chimiquement , Traitements médicamenteux de la COVID-19 , Souris de lignée C57BL , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Poumon/métabolisme , Poumon/effets des médicaments et des substances chimiques
18.
Front Cell Infect Microbiol ; 14: 1308362, 2024.
Article de Anglais | MEDLINE | ID: mdl-38476167

RÉSUMÉ

Infectious peritonitis is a leading cause of peritoneal functional impairment and a primary factor for therapy discontinuation in peritoneal dialysis (PD) patients. Although bacterial infections are a common cause of peritonitis episodes, emerging evidence suggests a role for viral pathogens. Toll-like receptors (TLRs) specifically recognize conserved pathogen-associated molecular patterns (PAMPs) from bacteria, viruses, and fungi, thereby orchestrating the ensuing inflammatory/immune responses. Among TLRs, TLR3 recognizes viral dsRNA and triggers antiviral response cascades upon activation. Epigenetic regulation, mediated by histone deacetylase (HDAC), has been demonstrated to control several cellular functions in response to various extracellular stimuli. Employing epigenetic target modulators, such as epidrugs, is a current therapeutic option in several cancers and holds promise in treating viral diseases. This study aims to elucidate the impact of TLR3 stimulation on the plasticity of human mesothelial cells (MCs) in PD patients and to investigate the effects of HDAC1-3 inhibition. Treatment of MCs from PD patients with the TLR3 agonist polyinosinic:polycytidylic acid (Poly(I:C)), led to the acquisition of a bona fide mesothelial-to-mesenchymal transition (MMT) characterized by the upregulation of mesenchymal genes and loss of epithelial-like features. Moreover, Poly(I:C) modulated the expression of several inflammatory cytokines and chemokines. A quantitative proteomic analysis of MCs treated with MS-275, an HDAC1-3 inhibitor, unveiled altered expression of several proteins, including inflammatory cytokines/chemokines and interferon-stimulated genes (ISGs). Treatment with MS-275 facilitated MMT reversal and inhibited the interferon signature, which was associated with reduced STAT1 phosphorylation. However, the modulation of inflammatory cytokine/chemokine production was not univocal, as IL-6 and CXCL8 were augmented while TNF-α and CXCL10 were decreased. Collectively, our findings underline the significance of viral infections in acquiring a mesenchymal-like phenotype by MCs and the potential consequences of virus-associated peritonitis episodes for PD patients. The observed promotion of MMT reversal and interferon response inhibition by an HDAC1-3 inhibitor, albeit without a general impact on inflammatory cytokine production, has translational implications deserving further analysis.


Sujet(s)
Benzamides , Interféron de type I , Péritonite , Pyridines , Maladies virales , Humains , Interféron de type I/métabolisme , Récepteur de type Toll-3/métabolisme , Épigenèse génétique , Protéomique , Cytokines/métabolisme , Chimiokines/métabolisme , Poly I-C/pharmacologie , Récepteurs de type Toll/métabolisme , Maladies virales/génétique , Phénotype , Histone Deacetylase 1/génétique , Histone Deacetylase 1/métabolisme
19.
Biomed Pharmacother ; 173: 116450, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38503239

RÉSUMÉ

Cisplatin not only targets DNA but also RNA. However, it is largely unknown whether platinated RNA (Pt-RNA) causes apoptosis and thus contributes to the cytotoxic effects of cisplatin. Consequently, cellular RNA was isolated from HepG2 and LS180 cells, exposed to cisplatin, and the resulting Pt-RNA (20 ng Pt/µg RNA) was transfected into these cancer cell lines or used to treat an apoptosis reporter Caenorhabditis elegans (C. elegans) strain (MD701, expressing CED-1::GFP). Cellular and molecular effects of Pt-RNA were evaluated by luminogenic caspase 3/7 assays, PCR array analysis, and fluorescence microscopy-based quantification of apoptosis in C. elegans gonads. Assuming RNA cross-linking (pseudo double-stranded RNA), the contribution of the Toll-like receptor 3 (TLR3, a sensor of double-stranded RNA) to apoptosis induction in cancer cell lines was investigated by pharmacological TLR3 inhibition and overexpression. In contrast to controls, Pt-RNA significantly enhanced apoptosis in C. elegans (2-fold) and in the cancer cell lines (2-fold to 4-fold). TLR3 overexpression significantly enhanced the pro-apoptotic effects of Pt-RNA in HepG2 cells. TLR3 inhibition reduced the pro-apoptotic effects of Pt-RNA and cisplatin, but not of paclitaxel (off-target control). Gene expression analysis showed that Pt-RNA (but not RNA) significantly enhanced the mRNA levels of nuclear factor kappa B subunit 2 and interleukin-8 in HepG2 cells, suggesting that Pt-RNA is a damage-associated molecular pattern that additionally causes pro-inflammatory responses. Together, this data suggests that not only DNA but also cellular RNA is a functionally relevant target of cisplatin, leading to pro-apoptotic and immunogenic effects.


Sujet(s)
Cisplatine , Tumeurs , Animaux , Cisplatine/pharmacologie , Caenorhabditis elegans/génétique , Caenorhabditis elegans/métabolisme , Récepteur de type Toll-3/génétique , Récepteur de type Toll-3/métabolisme , ARN double brin/génétique , ARN double brin/pharmacologie , Apoptose , Lignée cellulaire tumorale , ADN , Tumeurs/traitement médicamenteux , Tumeurs/génétique
20.
Biochem Biophys Res Commun ; 708: 149801, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38531219

RÉSUMÉ

Toll-like receptor (TLR) agonists or pro-inflammatory cytokines converge to activate the nuclear factor κB (NF-κB) signaling pathway, which provokes inflammatory responses. In the present study, we identified amiodarone hydrochloride as a selective inhibitor of the TLR3-mediated NF-κB signaling pathway by screening the RIKEN NPDepo Chemical Library. In human umbilical vein endothelial cells (HUVEC), amiodarone selectively inhibited the expression of intercellular adhesion molecule-1 (ICAM-1) induced by polyinosinic-polycytidylic acid (Poly(I:C)), but not tumor necrosis factor-α, interleukin-1α, or lipopolysaccharide. In response to a Poly(I:C) stimulation, amiodarone at 20 µM reduced the up-regulation of mRNA expression encoding ICAM-1, vascular cell adhesion molecule-1, and E-selectin. The nuclear translocation of the NF-κB subunit RelA was inhibited by amiodarone at 15-20 µM in Poly(I:C)-stimulated HUVEC. Amiodarone diminished the fluorescent dots of LysoTracker® Red DND-99 scattered over the cytoplasm of HUVEC. Therefore, the present study revealed that amiodarone selectively inhibited the TLR3-mediated NF-κB signaling pathway by blocking the acidification of intracellular organelles.


Sujet(s)
Amiodarone , Facteur de transcription NF-kappa B , Humains , Facteur de transcription NF-kappa B/métabolisme , Molécule-1 d'adhérence intercellulaire/métabolisme , Récepteur de type Toll-3/métabolisme , Cellules endothéliales/métabolisme , Amiodarone/pharmacologie , Amiodarone/métabolisme , Cellules cultivées , Transduction du signal , Molécule-1 d'adhérence des cellules vasculaires/métabolisme , Organites/métabolisme , Concentration en ions d'hydrogène
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE