Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 397
Filtrer
1.
Cell Mol Life Sci ; 81(1): 267, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38884678

RÉSUMÉ

Neutralizing antibodies are considered a correlate of protection against severe human respiratory syncytial virus (HRSV) disease. Currently, HRSV neutralization assays are performed on immortalized cell lines like Vero or A549 cells. It is known that assays on these cell lines exclusively detect neutralizing antibodies (nAbs) directed to the fusion (F) protein. For the detection of nAbs directed to the glycoprotein (G), ciliated epithelial cells expressing the cellular receptor CX3CR1 are required, but generation of primary cell cultures is expensive and labor-intensive. Here, we developed a high-throughput neutralization assay based on the interaction between clinically relevant HRSV grown on primary cells with ciliated epithelial cells, and validated this assay using a panel of infant sera. To develop the high-throughput neutralization assay, we established a culture of differentiated apical-out airway organoids (Ap-O AO). CX3CR1 expression was confirmed, and both F- and G-specific monoclonal antibodies neutralized HRSV in the Ap-O AO. In a side-by-side neutralization assay on Vero cells and Ap-O AO, neutralizing antibody levels in sera from 125 infants correlated well, although titers on Ap-O AO were consistently lower. We speculate that these lower titers might be an actual reflection of the neutralizing antibody capacity in vivo. The organoid-based neutralization assay described here holds promise for further characterization of correlates of protection against HRSV disease.


Sujet(s)
Anticorps neutralisants , Récepteur-1 de la chimiokine CX3C , Tests de neutralisation , Organoïdes , Infections à virus respiratoire syncytial , Virus respiratoire syncytial humain , Humains , Virus respiratoire syncytial humain/immunologie , Anticorps neutralisants/immunologie , Organoïdes/métabolisme , Organoïdes/immunologie , Organoïdes/virologie , Organoïdes/cytologie , Animaux , Tests de neutralisation/méthodes , Chlorocebus aethiops , Cellules Vero , Infections à virus respiratoire syncytial/immunologie , Infections à virus respiratoire syncytial/virologie , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/immunologie , Anticorps antiviraux/immunologie , Protéines de fusion virale/immunologie , Protéines de fusion virale/métabolisme , Nourrisson , Cellules épithéliales/métabolisme , Cellules épithéliales/immunologie , Cellules épithéliales/virologie , Anticorps monoclonaux/immunologie
2.
Int J Mol Sci ; 25(11)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38892268

RÉSUMÉ

The cellular distribution and changes in CX3CL1/fractalkine and its receptor CX3CR1 protein levels in the trigeminal subnucleus caudalis (TSC) of rats with unilateral infraorbital nerve ligation (IONL) were investigated on postoperation days 1, 3, 7, and 14 (POD1, POD3, POD7, and POD14, respectively) and compared with those of sham-operated and naïve controls. Behavioral tests revealed a significant increase in tactile hypersensitivity bilaterally in the vibrissal pads of both sham- and IONL-operated animals from POD1 to POD7, with a trend towards normalization in sham controls at POD14. Image analysis revealed increased CX3CL1 immunofluorescence (IF) intensities bilaterally in the TSC neurons of both sham- and IONL-operated rats at all survival periods. Reactive astrocytes in the ipsilateral TSC also displayed CX3CL1-IF from POD3 to POD14. At POD1 and POD3, microglial cells showed high levels of CX3CR1-IF, which decreased by POD7 and POD14. Conversely, CX3CR1 was increased in TSC neurons and reactive astrocytes at POD7 and POD14, which coincided with high levels of CX3CL1-IF and ADAM17-IF. This indicates that CX3CL1/CX3CR1 may be involved in reciprocal signaling between TSC neurons and reactive astrocytes. The level of CatS-IF in microglial cells suggests that soluble CX3CL1 may be involved in neuron-microglial cell signaling at POD3 and POD7, while ADAM17 allows this release at all studied time points. These results indicate an extended CX3CL1/CX3CR1 signaling axis and its role in the crosstalk between TSC neurons and glial cells during the development of trigeminal neuropathic pain.


Sujet(s)
Récepteur-1 de la chimiokine CX3C , Chimiokine CX3CL1 , Transduction du signal , Animaux , Chimiokine CX3CL1/métabolisme , Rats , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/génétique , Mâle , Microglie/métabolisme , Névralgie essentielle du trijumeau/métabolisme , Névralgie essentielle du trijumeau/anatomopathologie , Neurones/métabolisme , Astrocytes/métabolisme , Névralgie/métabolisme , Névralgie/anatomopathologie , Rat Sprague-Dawley
3.
Int J Med Sci ; 21(8): 1385-1398, 2024.
Article de Anglais | MEDLINE | ID: mdl-38903915

RÉSUMÉ

Inflammatory bowel disease (IBD) is a chronic inflammatory intestinal disease, characterized by dysregulated immune response. HDAC3 is reported to be an epigenetic brake in inflammation, playing critical roles in macrophages. However, its role in IBD is unclear. In our study, we found HDAC3 was upregulated in CX3CR1-positive cells in the mucosa from IBD mice. Conditional knockout (cKO) of Hdac3 in CX3CR1 positive cells attenuated the disease severity of Dextran Sulfate Sodium (DSS)-induced colitis. In addition, inhibition of HDAC3 with RGFP966 could also alleviate the DSS-induced tissue injury and inflammation in IBD. The RNA sequencing results revealed that Hdac3 cKO restrained DSS-induced upregulation of genes in the pathways of cytokine-cytokine receptor interaction, complement and coagulation cascades, chemokine signaling, and extracellular matrix receptor interaction. We also identified that Guanylate-Binding Protein 5 (GBP5) was transcriptionally regulated by HDAC3 in monocytes by RNA sequencing. Inhibition of HDAC3 resulted in decreased transcriptional activity of interferon-gamma-induced expression of GBP5 in CX3CR1-positive cells, such as macrophages and microglia. Overexpression of HDAC3 upregulated the transcriptional activity of GBP5 reporter. Lastly, conditional knockout of Hdac3 in macrophages (Hdac3 mKO) attenuated the disease severity of DSS-induced colitis. In conclusion, inhibition of HDAC3 in macrophages could ameliorate the disease severity and inflammatory response in colitis by regulating GBP5-NLRP3 axis, identifying a new therapeutic avenue for the treatment of colitis.


Sujet(s)
Colite , Sulfate dextran , Histone deacetylases , Macrophages , Souris knockout , Protéine-3 de la famille des NLR contenant un domaine pyrine , Transduction du signal , Animaux , Sulfate dextran/toxicité , Sulfate dextran/effets indésirables , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Souris , Macrophages/métabolisme , Macrophages/immunologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Colite/induit chimiquement , Colite/génétique , Colite/anatomopathologie , Colite/métabolisme , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Maladies inflammatoires intestinales/induit chimiquement , Maladies inflammatoires intestinales/génétique , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/traitement médicamenteux , Protéines G/génétique , Protéines G/métabolisme , Protéines G/antagonistes et inhibiteurs , Modèles animaux de maladie humaine , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/génétique , Souris de lignée C57BL , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/effets des médicaments et des substances chimiques , Acrylamides , Phénylènediamines
4.
Nat Commun ; 15(1): 5402, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38926390

RÉSUMÉ

Acute brain slices represent a workhorse model for studying the central nervous system (CNS) from nanoscale events to complex circuits. While slice preparation inherently involves tissue damage, it is unclear how microglia, the main immune cells and damage sensors of the CNS react to this injury and shape neuronal activity ex vivo. To this end, we investigated microglial phenotypes and contribution to network organization and functioning in acute brain slices. We reveal time-dependent microglial phenotype changes influenced by complex extracellular ATP dynamics through P2Y12R and CX3CR1 signalling, which is sustained for hours in ex vivo mouse brain slices. Downregulation of P2Y12R and changes of microglia-neuron interactions occur in line with alterations in the number of excitatory and inhibitory synapses over time. Importantly, functional microglia modulate synapse sprouting, while microglial dysfunction results in markedly impaired ripple activity both ex vivo and in vivo. Collectively, our data suggest that microglia are modulators of complex neuronal networks with important roles to maintain neuronal network integrity and activity. We suggest that slice preparation can be used to model time-dependent changes of microglia-neuron interactions to reveal how microglia shape neuronal circuits in physiological and pathological conditions.


Sujet(s)
Adénosine triphosphate , Encéphale , Récepteur-1 de la chimiokine CX3C , Microglie , Neurones , Récepteurs purinergiques P2Y12 , Synapses , Animaux , Microglie/métabolisme , Adénosine triphosphate/métabolisme , Souris , Neurones/métabolisme , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/génétique , Récepteurs purinergiques P2Y12/métabolisme , Récepteurs purinergiques P2Y12/génétique , Encéphale/métabolisme , Synapses/métabolisme , Souris de lignée C57BL , Phénotype , Mâle , Transduction du signal
5.
Biochemistry (Mosc) ; 89(5): 904-911, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38880650

RÉSUMÉ

Multiple sclerosis (MS) is a complex autoimmune disease of central nervous system (CNS) characterized by the myelin sheath destruction and compromised nerve signal transmission. Understanding molecular mechanisms driving MS development is critical due to its early onset, chronic course, and therapeutic approaches based only on symptomatic treatment. Cytokines are known to play a pivotal role in the MS pathogenesis with interleukin-6 (IL-6) being one of the key mediators. This study investigates contribution of IL-6 produced by microglia and dendritic cells to the development of experimental autoimmune encephalomyelitis (EAE), a widely used mouse model of MS. Mice with conditional inactivation of IL-6 in the CX3CR1+ cells, including microglia, or CD11c+ dendritic cells, displayed less severe symptoms as compared to their wild-type counterparts. Mice with microglial IL-6 deletion exhibited an elevated proportion of regulatory T cells and reduced percentage of pathogenic IFNγ-producing CD4+ T cells, accompanied by the decrease in pro-inflammatory monocytes in the CNS at the peak of EAE. At the same time, deletion of IL-6 from microglia resulted in the increase of CCR6+ T cells and GM-CSF-producing T cells. Conversely, mice with IL-6 deficiency in the dendritic cells showed not only the previously described increase in the proportion of regulatory T cells and decrease in the proportion of TH17 cells, but also reduction in the production of GM-CSF and IFNγ in the secondary lymphoid organs. In summary, IL-6 functions during EAE depend on both the source and localization of immune response: the microglial IL-6 exerts both pathogenic and protective functions specifically in the CNS, whereas the dendritic cell-derived IL-6, in addition to being critically involved in the balance of regulatory T cells and TH17 cells, may stimulate production of cytokines associated with pathogenic functions of T cells.


Sujet(s)
Cellules dendritiques , Modèles animaux de maladie humaine , Encéphalomyélite auto-immune expérimentale , Interleukine-6 , Microglie , Sclérose en plaques , Animaux , Cellules dendritiques/métabolisme , Cellules dendritiques/immunologie , Souris , Interleukine-6/métabolisme , Sclérose en plaques/immunologie , Sclérose en plaques/métabolisme , Sclérose en plaques/anatomopathologie , Microglie/métabolisme , Encéphalomyélite auto-immune expérimentale/métabolisme , Encéphalomyélite auto-immune expérimentale/immunologie , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Souris de lignée C57BL , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/génétique , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Récepteurs CCR6/métabolisme , Récepteurs CCR6/génétique , Femelle
6.
Oncoimmunology ; 13(1): 2355684, 2024.
Article de Anglais | MEDLINE | ID: mdl-38798746

RÉSUMÉ

Identifying tumor-relevant T cell subsets in the peripheral blood (PB) has become a potential strategy for cancer treatment. However, the subset of PB that could be used to treat cancer remains poorly defined. Here, we found that the CX3CR1+ T cell subset in the blood of patients with lung cancer exhibited effector properties and had a higher TCR matching ratio with tumor-infiltrating lymphocytes (TILs) compared to CX3CR1- T cells, as determined by paired single-cell RNA and TCR sequencing. Meanwhile, the anti-tumor activities, effector cytokine production, and mitochondrial function were enhanced in CX3CR1+ T cells both in vitro and in vivo. However, in the co-culture system of H322 cells with T cells, the percentages of apoptotic cells and Fas were substantially higher in CX3CR1+ T cells than those in CX3CR1- T cells. Fas-mediated apoptosis was rescued by treatment with an anti-PD-1 antibody. Accordingly, the combination of adoptive transfer of CX3CR1+ T cells and anti-PD-1 treatment considerably decreased Fas expression and improved the survival of lung xenograft mice. Moreover, an increased frequency of CX3CR1+ T cells in the PB correlated with a better response and prolonged survival of patients with lung cancer who received anti-PD-1 therapy. These findings indicate the promising potential of adoptive transfer of peripheral CX3CR1+ T cells as an individual cancer immunotherapy.


Sujet(s)
Récepteur-1 de la chimiokine CX3C , Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Lymphocytes TIL , Récepteur-1 de mort cellulaire programmée , Animaux , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Récepteur-1 de la chimiokine CX3C/métabolisme , Humains , Souris , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Lymphocytes TIL/immunologie , Lymphocytes TIL/effets des médicaments et des substances chimiques , Lymphocytes TIL/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe , Lignée cellulaire tumorale , Femelle , Apoptose/effets des médicaments et des substances chimiques , Mâle , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/effets des médicaments et des substances chimiques , Sous-populations de lymphocytes T/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme
7.
Article de Anglais | MEDLINE | ID: mdl-38760287

RÉSUMÉ

OBJECTIVE: We aimed to describe the association between CX3CR1, CX3CL1, and ITGAV immunoexpression with PNI and adverse oncologic outcomes in patients with OSCC. STUDY DESIGN: Expression CX3CR1, CX3CL1, and ITGAV was assessed by immunohistochemistry in a cohort of 50 paraffin-embedded resections of OSCC. Survival analysis, Cox, and binary logistic regressions were undertaken to determine the impact on patient survival and predictive value for PNI. RESULTS: CX3CL1 positive nerves exhibited a significant association with tumor budding (TB) (P = .043), whereas nerves positive for ITGAV were associated with PNI (P = .021), T3-T4 tumor size (P = .029), and III-IV stage (P = .044). Cases with ITGAV-positive nerves exhibited an odds ratio of 9.603 (P = .008) for PNI, whereas cases with CX3CL1-positive nerves exhibited and odds ratio of 4.682 (P = .033) for TB. A trend toward decreased 5-year overall survival (P = .078) and 5-year disease-specific survival (P = .09) was observed in relation to ITGAV-positive nerves. However, no independent predictors for poor survival were identified. CONCLUSIONS: The expression of ITGAV was associated with PNI and advanced disease, whereas the expression of CX3CL1 was related to TB, suggesting that ITGAV and CX3CL1 are involved in their respective developments. Therefore, further investigations are encouraged to assess the potential utility of targeted therapies against CX3CL1 receptors in OSCC.


Sujet(s)
Marqueurs biologiques tumoraux , Récepteur-1 de la chimiokine CX3C , Carcinome épidermoïde , Chimiokine CX3CL1 , Immunohistochimie , Tumeurs de la bouche , Humains , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/métabolisme , Femelle , Mâle , Chimiokine CX3CL1/métabolisme , Adulte d'âge moyen , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Sujet âgé , Marqueurs biologiques tumoraux/métabolisme , Récepteur-1 de la chimiokine CX3C/métabolisme , Adulte , Stadification tumorale , Sujet âgé de 80 ans ou plus , Pronostic , Invasion tumorale , Analyse de survie , Valeur prédictive des tests
8.
J Am Heart Assoc ; 13(9): e034731, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38700011

RÉSUMÉ

BACKGROUND: Cardiac damage induced by ischemic stroke, such as arrhythmia, cardiac dysfunction, and even cardiac arrest, is referred to as cerebral-cardiac syndrome (CCS). Cardiac macrophages are reported to be closely associated with stroke-induced cardiac damage. However, the role of macrophage subsets in CCS is still unclear due to their heterogeneity. Sympathetic nerves play a significant role in regulating macrophages in cardiovascular disease. However, the role of macrophage subsets and sympathetic nerves in CCS is still unclear. METHODS AND RESULTS: In this study, a middle cerebral artery occlusion mouse model was used to simulate ischemic stroke. ECG and echocardiography were used to assess cardiac function. We used Cx3cr1GFPCcr2RFP mice and NLRP3-deficient mice in combination with Smart-seq2 RNA sequencing to confirm the role of macrophage subsets in CCS. We demonstrated that ischemic stroke-induced cardiac damage is characterized by severe cardiac dysfunction and robust infiltration of monocyte-derived macrophages into the heart. Subsequently, we identified that cardiac monocyte-derived macrophages displayed a proinflammatory profile. We also observed that cardiac dysfunction was rescued in ischemic stroke mice by blocking macrophage infiltration using a CCR2 antagonist and NLRP3-deficient mice. In addition, a cardiac sympathetic nerve retrograde tracer and a sympathectomy method were used to explore the relationship between sympathetic nerves and cardiac macrophages. We found that cardiac sympathetic nerves are significantly activated after ischemic stroke, which contributes to the infiltration of monocyte-derived macrophages and subsequent cardiac dysfunction. CONCLUSIONS: Our findings suggest a potential pathogenesis of CCS involving the cardiac sympathetic nerve-monocyte-derived macrophage axis.


Sujet(s)
Modèles animaux de maladie humaine , Accident vasculaire cérébral ischémique , Macrophages , Souris de lignée C57BL , Protéine-3 de la famille des NLR contenant un domaine pyrine , Animaux , Macrophages/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Protéine-3 de la famille des NLR contenant un domaine pyrine/déficit , Accident vasculaire cérébral ischémique/physiopathologie , Accident vasculaire cérébral ischémique/métabolisme , Accident vasculaire cérébral ischémique/anatomopathologie , Récepteurs CCR2/génétique , Récepteurs CCR2/métabolisme , Mâle , Souris knockout , Souris , Infarctus du territoire de l'artère cérébrale moyenne/physiopathologie , Infarctus du territoire de l'artère cérébrale moyenne/anatomopathologie , Système nerveux sympathique/physiopathologie , Myocarde/anatomopathologie , Myocarde/métabolisme , Cardiopathies/étiologie , Cardiopathies/physiopathologie , Cardiopathies/anatomopathologie , Récepteur-1 de la chimiokine CX3C/génétique , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/déficit
9.
Front Immunol ; 15: 1383607, 2024.
Article de Anglais | MEDLINE | ID: mdl-38715600

RÉSUMÉ

Background: The crucial role of inflammation in aortic aneurysm (AA) is gaining prominence, while there is still a lack of key cytokines or targets for effective clinical translation. Methods: Mendelian randomization (MR) analysis was performed to identify the causal relationship between 91 circulating inflammatory proteins and AA and between 731 immune traits and AA. Bulk RNA sequencing data was utilized to demonstrate the expression profile of the paired ligand-receptor. Gene enrichment analysis, Immune infiltration, and correlation analysis were employed to deduce the potential role of CX3CR1. We used single-cell RNA sequencing data to pinpoint the localization of CX3CL1 and CX3CR1, which was further validated by multiplex immunofluorescence staining. Cellchat analysis was utilized to infer the CX3C signaling pathway. Trajectory analysis and the Cytosig database were exploited to determine the downstream effect of CX3CL1-CX3CR1. Results: We identified 4 candidates (FGF5, CX3CL1, IL20RA, and SCF) in multiple two-sample MR analyses. Subsequent analysis of the expression profile of the paired receptor revealed the significant upregulation of CX3CR1 in AA and its positive correlation with pro-inflammatory macrophages. Two sample MR between immune cell traits and AA demonstrated the potential causality between intermediate monocytes and AA. We finally deciphered in single-cell sequencing data that CX3CL1 sent by endothelial cells (ECs) acted on CX3CR1 of intermediated monocytes, leading to its recruitment and pro-inflammatory responses. Conclusion: Our study presented a genetic insight into the pathogenetic role of CX3CL1-CX3CR1 in AA, and further deciphered the CX3C signaling pathway between ECs and intermediate monocytes.


Sujet(s)
Anévrysme de l'aorte , Récepteur-1 de la chimiokine CX3C , Chimiokine CX3CL1 , Analyse de randomisation mendélienne , Récepteur-1 de la chimiokine CX3C/génétique , Récepteur-1 de la chimiokine CX3C/métabolisme , Humains , Chimiokine CX3CL1/génétique , Chimiokine CX3CL1/métabolisme , Anévrysme de l'aorte/génétique , Anévrysme de l'aorte/métabolisme , Analyse de profil d'expression de gènes , Transcriptome , Transduction du signal , Prédisposition génétique à une maladie
10.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38731899

RÉSUMÉ

The chemotactic cytokine fractalkine (FKN, chemokine CX3CL1) has unique properties resulting from the combination of chemoattractants and adhesion molecules. The soluble form (sFKN) has chemotactic properties and strongly attracts T cells and monocytes. The membrane-bound form (mFKN) facilitates diapedesis and is responsible for cell-to-cell adhesion, especially by promoting the strong adhesion of leukocytes (monocytes) to activated endothelial cells with the subsequent formation of an extracellular matrix and angiogenesis. FKN signaling occurs via CX3CR1, which is the only known member of the CX3C chemokine receptor subfamily. Signaling within the FKN-CX3CR1 axis plays an important role in many processes related to inflammation and the immune response, which often occur simultaneously and overlap. FKN is strongly upregulated by hypoxia and/or inflammation-induced inflammatory cytokine release, and it may act locally as a key angiogenic factor in the highly hypoxic tumor microenvironment. The importance of the FKN/CX3CR1 signaling pathway in tumorigenesis and cancer metastasis results from its influence on cell adhesion, apoptosis, and cell migration. This review presents the role of the FKN signaling pathway in the context of angiogenesis in inflammation and cancer. The mechanisms determining the pro- or anti-tumor effects are presented, which are the cause of the seemingly contradictory results that create confusion regarding the therapeutic goals.


Sujet(s)
Récepteur-1 de la chimiokine CX3C , Carcinogenèse , Chimiokine CX3CL1 , Inflammation , Néovascularisation pathologique , Transduction du signal , Humains , Chimiokine CX3CL1/métabolisme , Néovascularisation pathologique/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/génétique , Animaux , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Tumeurs/étiologie , Microenvironnement tumoral ,
11.
Cells ; 13(10)2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38786041

RÉSUMÉ

Monocytes, as well as downstream macrophages and dendritic cells, are essential players in the immune system, fulfilling key roles in homeostasis as well as in inflammatory conditions. Conventionally, driven by studies on reporter models, mouse monocytes are categorized into a classical and a non-classical subset based on their inversely correlated surface expression of Ly6C/CCR2 and CX3CR1. Here, we aimed to challenge this concept by antibody staining and reporter mouse models. Therefore, we took advantage of Cx3cr1GFP and Ccr2RFP reporter mice, in which the respective gene was replaced by a fluorescent reporter protein gene. We analyzed the expression of CX3CR1 and CCR2 by flow cytometry using several validated fluorochrome-coupled antibodies and compared them with the reporter gene signal in these reporter mouse strains. Although we were able to validate the specificity of the fluorochrome-coupled flow cytometry antibodies, mouse Ly6Chigh classical and Ly6Clow non-classical monocytes showed no differences in CX3CR1 expression levels in the peripheral blood and spleen when stained with these antibodies. On the contrary, in Cx3cr1GFP reporter mice, we were able to reproduce the inverse correlation of the CX3CR1 reporter gene signal and Ly6C surface expression. Furthermore, differential CCR2 surface expression correlating with the expression of Ly6C was observed by antibody staining, but not in Ccr2RFP reporter mice. In conclusion, our data suggest that phenotyping strategies for mouse monocyte subsets should be carefully selected. In accordance with the literature, the suitability of CX3CR1 antibody staining is limited, whereas for CCR2, caution should be applied when using reporter mice.


Sujet(s)
Récepteur-1 de la chimiokine CX3C , Cytométrie en flux , Monocytes , Récepteurs CCR2 , Animaux , Récepteurs CCR2/métabolisme , Récepteurs CCR2/génétique , Monocytes/métabolisme , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/génétique , Souris , Anticorps/immunologie , Gènes rapporteurs , Phénotype , Souris de lignée C57BL , Souris transgéniques , Protéines à fluorescence verte/métabolisme , Antigènes Ly/métabolisme , Antigènes Ly/génétique
12.
Viruses ; 16(5)2024 05 14.
Article de Anglais | MEDLINE | ID: mdl-38793659

RÉSUMÉ

Respiratory syncytial virus (RSV) is the most prevalent cause of acute lower respiratory infection in young children. Currently, the first RSV vaccines are approved by the FDA. Recently, N6-methyladenosine (m6A) RNA methylation has been implicated in the regulation of the viral life cycle and replication of many viruses, including RSV. m6A methylation of RSV RNA has been demonstrated to promote replication and prevent anti-viral immune responses by the host. Whether m6A is also involved in viral entry and whether m6A can also affect RSV infection via different mechanisms than methylation of viral RNA is poorly understood. Here, we identify m6A reader YTH domain-containing protein 1 (YTHDC1) as a novel negative regulator of RSV infection. We demonstrate that YTHDC1 abrogates RSV infection by reducing the expression of RSV entry receptor CX3C motif chemokine receptor 1 (CX3CR1) on the cell surface of lung epithelial cells. Altogether, these data reveal a novel role for m6A methylation and YTHDC1 in the viral entry of RSV. These findings may contribute to the development of novel treatment options to control RSV infection.


Sujet(s)
Adénosine , Récepteur-1 de la chimiokine CX3C , Infections à virus respiratoire syncytial , Virus respiratoire syncytial humain , Pénétration virale , Humains , Infections à virus respiratoire syncytial/virologie , Infections à virus respiratoire syncytial/métabolisme , Virus respiratoire syncytial humain/génétique , Virus respiratoire syncytial humain/physiologie , Adénosine/analogues et dérivés , Adénosine/métabolisme , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/génétique , Réplication virale , Méthylation , Régulation négative , Facteurs d'épissage des ARN/métabolisme , Facteurs d'épissage des ARN/génétique , Cellules épithéliales/virologie , Cellules épithéliales/métabolisme , Lignée cellulaire , Cellules A549 , ARN viral/génétique , ARN viral/métabolisme , Interactions hôte-pathogène , Protéines de tissu nerveux
13.
Mult Scler Relat Disord ; 87: 105634, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38677127

RÉSUMÉ

BACKGROUND: Exercise may have beneficial effects in MS, remaining controversial its possible disease-modifying effects and which mechanisms might be involved. We evaluated whether exercise-induced lymphocyte redistribution differ in MS patients as compared to controls. METHODS: Exercise was assessed in 12 relapsing-remitting MS patients and 11 controls in a cycle ergometer, obtaining blood samples before exercise, at maximal exercise capacity (T1), and after resting (T2). Peripheral lymphocytes were evaluated by flow cytometry, assessing chemokine receptor expression to study cell trafficking properties. RESULTS: Lymphocyte subsets in all cases increased after exercise and decreased at resting. However, total natural killer (NK) cells in patients as compared to controls had a lower exercise-induced redeployment at T1 (696 ± 581 cells/µL vs.1502 ± 641 cells/µL, p < 0.01). Evaluating NK cell subsets, CD56bright NK cells numbers decreased in peripheral blood in MS patients after resting (T2), contrasting with values remaining above baseline in healthy controls. NK cells mobilized after exercise at T1 in controls, as compared to patients, had a higher CX3CR1 expression (1402 ± 564/µL vs. 615 ± 548 cell//µL, p < 0.01). CONCLUSION: Exercise-induced redeployment of NK cells may be reduced in MS patients, as well as their migration capabilities, pointing to potential immunological mechanisms to be enhanced by exercise training programs.


Sujet(s)
Exercice physique , Cellules tueuses naturelles , Sclérose en plaques récurrente-rémittente , Humains , Cellules tueuses naturelles/immunologie , Femelle , Mâle , Adulte , Sclérose en plaques récurrente-rémittente/immunologie , Sclérose en plaques récurrente-rémittente/sang , Sclérose en plaques récurrente-rémittente/physiopathologie , Exercice physique/physiologie , Adulte d'âge moyen , Épreuve d'effort , Récepteur-1 de la chimiokine CX3C/métabolisme
14.
Arterioscler Thromb Vasc Biol ; 44(6): 1246-1264, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38660801

RÉSUMÉ

BACKGROUND: Heterogeneity in the severity of cerebral cavernous malformations (CCMs) disease, including brain bleedings and thrombosis that cause neurological disabilities in patients, suggests that environmental, genetic, or biological factors act as disease modifiers. Still, the underlying mechanisms are not entirely understood. Here, we report that mild hypoxia accelerates CCM disease by promoting angiogenesis, neuroinflammation, and vascular thrombosis in the brains of CCM mouse models. METHODS: We used genetic studies, RNA sequencing, spatial transcriptome, micro-computed tomography, fluorescence-activated cell sorting, multiplex immunofluorescence, coculture studies, and imaging techniques to reveal that sustained mild hypoxia via the CX3CR1-CX3CL1 (CX3C motif chemokine receptor 1/chemokine [CX3C motif] ligand 1) signaling pathway influences cell-specific neuroinflammatory interactions, contributing to heterogeneity in CCM severity. RESULTS: Histological and expression profiles of CCM neurovascular lesions (Slco1c1-iCreERT2;Pdcd10fl/fl; Pdcd10BECKO) in male and female mice found that sustained mild hypoxia (12% O2, 7 days) accelerates CCM disease. Our findings indicate that a small reduction in oxygen levels can significantly increase angiogenesis, neuroinflammation, and thrombosis in CCM disease by enhancing the interactions between endothelium, astrocytes, and immune cells. Our study indicates that the interactions between CX3CR1 and CX3CL1 are crucial in the maturation of CCM lesions and propensity to CCM immunothrombosis. In particular, this pathway regulates the recruitment and activation of microglia and other immune cells in CCM lesions, which leads to lesion growth and thrombosis. We found that human CX3CR1 variants are linked to lower lesion burden in familial CCMs, proving it is a genetic modifier in human disease and a potential marker for aggressiveness. Moreover, monoclonal blocking antibody against CX3CL1 or reducing 1 copy of the Cx3cr1 gene significantly reduces hypoxia-induced CCM immunothrombosis. CONCLUSIONS: Our study reveals that interactions between CX3CR1 and CX3CL1 can modify CCM neuropathology when lesions are accelerated by environmental hypoxia. Moreover, a hypoxic environment or hypoxia signaling caused by CCM disease influences the balance between neuroinflammation and neuroprotection mediated by CX3CR1-CX3CL1 signaling. These results establish CX3CR1 as a genetic marker for patient stratification and a potential predictor of CCM aggressiveness.


Sujet(s)
Récepteur-1 de la chimiokine CX3C , Chimiokine CX3CL1 , Modèles animaux de maladie humaine , Hémangiome caverneux du système nerveux central , Transduction du signal , Animaux , Femelle , Humains , Mâle , Souris , Chimiokine CX3CL1/métabolisme , Chimiokine CX3CL1/génétique , Récepteur-1 de la chimiokine CX3C/génétique , Récepteur-1 de la chimiokine CX3C/métabolisme , Hémangiome caverneux du système nerveux central/génétique , Hémangiome caverneux du système nerveux central/métabolisme , Hémangiome caverneux du système nerveux central/anatomopathologie , Hypoxie/métabolisme , Hypoxie/complications , Souris de lignée C57BL , Souris knockout , Néovascularisation pathologique/métabolisme , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/anatomopathologie , Maladies neuro-inflammatoires/génétique
15.
Lung ; 202(3): 343-356, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38678499

RÉSUMÉ

BACKGROUND: Severe asthma, characterized by inflammation and airway remodeling, involves fibroblast differentiation into myofibroblasts expressing α-SMA. This process leads to the production of fibronectin and connective tissue growth factor (CTGF), driven by factors such as transforming growth factor (TGF)-ß. Furthermore, the persistent presence of myofibroblasts is associated with resistance to apoptosis and mitochondrial dysfunction. The chemokine (C-X3-C motif) ligand 1 (CX3CL1) plays a role in tissue fibrosis. However, it is currently unknown whether neutralization of CX3CL1 decreases TGF-ß-induced fibroblast differentiation and mitochondrial dysfunction in normal human lung fibroblasts (NHLFs). METHODS: CX3CL1/C-X3-C motif chemokine receptor 1 (CX3CR1), CX3CL1 was analyzed by immunofluorescence (IF) or immunohistochemical (IHC) staining of ovalbumin-challenged mice. CX3CL1 release was detected by ELISA. TGF-ß-induced CTGF, fibronectin, and α-SMA expression were evaluated in NHLFs following neutralization of CX3CL1 (TP213) treatment for the indicated times by Western blotting or IF staining. Mitochondrion function was detected by a JC-1 assay and seahorse assay. Cell apoptosis was observed by a terminal uridine nick-end labeling (TUNEL) assay. RESULTS: An increase in CX3CL1 expression was observed in lung tissues from mice with ovalbumin-induced asthma by IF staining. CX3CR1 was increased in the subepithelial layer of the airway by IHC staining. Moreover, CX3CR1 small interfering (si)RNA downregulated TGF-ß-induced CTGF and fibronectin expression in NHLFs. CX3CL1 induced CTGF and fibronectin expression in NHLFs. TGF-ß-induced CX3CL1 secretion from NHLFs. Furthermore, TP213 decreased TGF-ß-induced CTGF, fibronectin, and α-SMA expression in NHLFs. Mitochondrion-related differentially expressed genes (DEGs) were examined after CX3CL1 neutralization in TGF-ß-treated NHLFs. TP213 alleviated TGF-ß-induced mitochondrial dysfunction and apoptosis resistance in NHLFs. CX3CL1 induced p65, IκBα, and IKKα phosphorylation in a time-dependent manner. Furthermore, CX3CL1-induced fibronectin expression and JC-1 monomer were decreased by p65 siRNA. TP213 reduced TGF-ß-induced p65 and α-SMA expression in NHLFs. CONCLUSIONS: These findings suggest that neutralizing CX3CL1 attenuates lung fibroblast activation and mitochondrial dysfunction. Understanding the impacts of CX3CL1 neutralization on fibroblast mitochondrial function could contribute to the development of therapeutic strategies for managing airway remodeling in severe asthma.


Sujet(s)
Apoptose , Récepteur-1 de la chimiokine CX3C , Différenciation cellulaire , Chimiokine CX3CL1 , Facteur de croissance du tissu conjonctif , Fibroblastes , Fibronectines , Mitochondries , Fibrose pulmonaire , Facteur de croissance transformant bêta , Chimiokine CX3CL1/métabolisme , Chimiokine CX3CL1/génétique , Animaux , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/anatomopathologie , Humains , Facteur de croissance du tissu conjonctif/métabolisme , Facteur de croissance du tissu conjonctif/génétique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/anatomopathologie , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Facteur de croissance transformant bêta/métabolisme , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/génétique , Fibronectines/métabolisme , Souris , Actines/métabolisme , Poumon/anatomopathologie , Poumon/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal , Asthme/métabolisme , Asthme/anatomopathologie , Modèles animaux de maladie humaine , Cellules cultivées , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Myofibroblastes/effets des médicaments et des substances chimiques , Ovalbumine
16.
Microbiol Spectr ; 12(5): e0272023, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38572984

RÉSUMÉ

Gut microbiota has demonstrated an increasingly important role in the onset and development of colorectal cancer (CRC). Nonetheless, the association between gut microbiota and KRAS mutation in CRC remains enigmatic. We conducted 16S rRNA sequencing on stool samples from 94 CRC patients and employed the linear discriminant analysis effect size algorithm to identify distinct gut microbiota between KRAS mutant and KRAS wild-type CRC patients. Transcriptome sequencing data from nine CRC patients were transformed into a matrix of immune infiltrating cells, which was then utilized to explore KRAS mutation-associated biological functions, including Gene Ontology items and Kyoto Encyclopedia of Genes and Genomes pathways. Subsequently, we analyzed the correlations among these KRAS mutation-associated gut microbiota, host immunity, and KRAS mutation-associated biological functions. At last, we developed a predictive random forest (RF) machine learning model to predict the KRAS mutation status in CRC patients, based on the gut microbiota associated with KRAS mutation. We identified a total of 26 differential gut microbiota between both groups. Intriguingly, a significant positive correlation was observed between Bifidobacterium spp. and mast cells, as well as between Bifidobacterium longum and chemokine receptor CX3CR1. Additionally, we also observed a notable negative correlation between Bifidobacterium and GOMF:proteasome binding. The RF model constructed using the KRAS mutation-associated gut microbiota demonstrated qualified efficacy in predicting the KRAS phenotype in CRC. Our study ascertained the presence of 26 KRAS mutation-associated gut microbiota in CRC and speculated that Bifidobacterium may exert an essential role in preventing CRC progression, which appeared to correlate with the upregulation of mast cells and CX3CR1 expression, as well as the downregulation of GOMF:proteasome binding. Furthermore, the RF model constructed on the basis of KRAS mutation-associated gut microbiota exhibited substantial potential in predicting KRAS mutation status in CRC patients.IMPORTANCEGut microbiota has emerged as an essential player in the onset and development of colorectal cancer (CRC). However, the relationship between gut microbiota and KRAS mutation in CRC remains elusive. Our study not only identified a total of 26 gut microbiota associated with KRAS mutation in CRC but also unveiled their significant correlations with tumor-infiltrating immune cells, immune-related genes, and biological pathways (Gene Ontology items and Kyoto Encyclopedia of Genes and Genomes pathways). We speculated that Bifidobacterium may play a crucial role in impeding CRC progression, potentially linked to the upregulation of mast cells and CX3CR1 expression, as well as the downregulation of GOMF:Proteasome binding. Furthermore, based on the KRAS mutation-associated gut microbiota, the RF model exhibited promising potential in the prediction of KRAS mutation status for CRC patients. Overall, the findings of our study offered fresh insights into microbiological research and clinical prediction of KRAS mutation status for CRC patients.


Sujet(s)
Tumeurs colorectales , Microbiome gastro-intestinal , Apprentissage machine , Mutation , Protéines proto-oncogènes p21(ras) , Humains , Tumeurs colorectales/microbiologie , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Microbiome gastro-intestinal/génétique , Protéines proto-oncogènes p21(ras)/génétique , Mâle , Femelle , ARN ribosomique 16S/génétique , Adulte d'âge moyen , Sujet âgé , Fèces/microbiologie , Bifidobacterium/génétique , Bactéries/génétique , Bactéries/classification , Bactéries/isolement et purification , Récepteur-1 de la chimiokine CX3C/génétique , Récepteur-1 de la chimiokine CX3C/métabolisme
17.
Infect Immun ; 92(5): e0000624, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38629806

RÉSUMÉ

Enterococci are common commensal bacteria that colonize the gastrointestinal tracts of most mammals, including humans. Importantly, these bacteria are one of the leading causes of nosocomial infections. This study examined the role of colonic macrophages in facilitating Enterococcus faecalis infections in mice. We determined that depletion of colonic phagocytes resulted in the reduction of E. faecalis dissemination to the gut-draining mesenteric lymph nodes. Furthermore, we established that trafficking of monocyte-derived CX3CR1-expressing macrophages contributed to E. faecalis dissemination in a manner that was not reliant on CCR7, the conventional receptor involved in lymphatic migration. Finally, we showed that E. faecalis mutants with impaired intracellular survival exhibited reduced dissemination, suggesting that E. faecalis can exploit host immune cell migration to disseminate systemically and cause disease. Our findings indicate that modulation of macrophage trafficking in the context of antibiotic therapy could serve as a novel approach for preventing or treating opportunistic infections by disseminating enteric pathobionts like E. faecalis.


Sujet(s)
Récepteur-1 de la chimiokine CX3C , Côlon , Enterococcus faecalis , Macrophages , Récepteurs CCR2 , Récepteurs aux chimiokines , Animaux , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/génétique , Macrophages/microbiologie , Macrophages/immunologie , Souris , Côlon/microbiologie , Côlon/immunologie , Récepteurs CCR2/métabolisme , Récepteurs CCR2/génétique , Récepteurs aux chimiokines/métabolisme , Récepteurs aux chimiokines/génétique , Infections bactériennes à Gram positif/immunologie , Infections bactériennes à Gram positif/microbiologie , Souris de lignée C57BL , Noeuds lymphatiques/microbiologie , Noeuds lymphatiques/immunologie , Récepteurs CCR7/métabolisme , Récepteurs CCR7/génétique
18.
Int J Mol Sci ; 25(8)2024 Apr 18.
Article de Anglais | MEDLINE | ID: mdl-38674036

RÉSUMÉ

CX3CL1, also named fractalkine or neurotactin, is the only known member of the CX3C chemokine family that can chemoattract several immune cells. CX3CL1 exists in both membrane-anchored and soluble forms, with each mediating distinct biological activities. CX3CL1 signals are transmitted through its unique receptor, CX3CR1, primarily expressed in the microglia of the central nervous system (CNS). In the CNS, CX3CL1 acts as a regulator of microglia activation in response to brain disorders or inflammation. Recently, there has been a growing interest in the role of CX3CL1 in regulating cell adhesion, chemotaxis, and host immune response in viral infection. Here, we provide a comprehensive review of the changes and function of CX3CL1 in various viral infections, such as human immunodeficiency virus (HIV), SARS-CoV-2, influenza virus, and cytomegalovirus (CMV) infection, to highlight the emerging roles of CX3CL1 in viral infection and associated diseases.


Sujet(s)
Chimiokine CX3CL1 , Maladies virales , Chimiokine CX3CL1/métabolisme , Humains , Maladies virales/métabolisme , Maladies virales/immunologie , Maladies virales/virologie , Animaux , COVID-19/virologie , COVID-19/métabolisme , COVID-19/immunologie , SARS-CoV-2/pathogénicité , SARS-CoV-2/physiologie , Microglie/métabolisme , Microglie/virologie , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/génétique
19.
PLoS Negl Trop Dis ; 18(4): e0012112, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38669292

RÉSUMÉ

Visceral leishmaniasis (VL) is a potentially fatal parasitic infection caused by Leishmania donovani in India. L. donovani is an obligate intracellular protozoan residing mostly in macrophages of the reticuloendothelial system throughout chronic infection. Monocytic phagocytes are critical in the pathogenesis of different forms of leishmaniasis. Subsets of monocytes are distinguished by their surface markers into CD14+CD16- classical monocytes, CD14+CD16+ intermediate monocytes, and CD16++CD14low non-classical monocyte subsets. During cutaneous leishmaniasis (CL), intermediate monocyte are reported to be a source of inflammatory cytokines IL-1ß and TNF, and they express CCR2 attracting them to sites of inflammatory pathology. We examined monocyte subsets in the blood and bone marrow of patients with VL from an endemic site in Bihar, India, and found these contrasted with the roles of monocytes in CL. During VL, intermediate and non-classical CD16+ monocyte subsets expressed instead a non-inflammatory phenotype with low CCR2, high CX3CR1 and low microbicidal oxidant generation, making them more similar to patrolling monocytes than inflammatory cells. Bone marrow CD16+ monocyte subsets expressed a phenotype that might be more similar to the inflammatory subsets of CL, although our inability to obtain bone marrow from healthy donors in the endemic region hampered this interpretation Overall the data suggest that CD16+ intermediate monocyte subsets in VL patients express a phenotypes that contributes to an immunosuppressed pathologic immune state, but in contrast to CL, these do not mediate localized inflammatory responses.


Sujet(s)
Moelle osseuse , Leishmaniose viscérale , Monocytes , Leishmaniose viscérale/immunologie , Leishmaniose viscérale/parasitologie , Humains , Monocytes/immunologie , Inde , Adulte , Mâle , Moelle osseuse/parasitologie , Femelle , Récepteurs du fragment Fc des IgG/analyse , Récepteurs du fragment Fc des IgG/métabolisme , Leishmania donovani/immunologie , Leishmania donovani/physiologie , Jeune adulte , Adolescent , Récepteurs CCR2/métabolisme , Adulte d'âge moyen , Enfant , Récepteurs aux chimiokines/métabolisme , Récepteur-1 de la chimiokine CX3C/génétique , Récepteur-1 de la chimiokine CX3C/métabolisme , Cytokines/métabolisme
20.
Brain Res Bull ; 211: 110939, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38574865

RÉSUMÉ

PURPOSE: To evaluate the potential efficacy of Triptolide (TP) on cerebral ischemia/reperfusion injury (CIRI) and to uncover the underlying mechanism through which TP regulates CIRI. METHODS: We constructed a middle cerebral artery occlusion/reperfusion (MCAO/R) mouse model to simulate CIRI, and established a lipopolysaccharide (LPS)-stimulated BV-2 cell model to mimic the inflammatory state during CIRI. The neurological deficits score (NS) of mice were measured for assessment of neurologic functions. Both the severity of cerebral infarction and the apoptosis level in mouse brain tissues or cells were respectively evaluated using corresponding techniques. The expression levels of Ionized calcium binding adapter molecule 1 (IBA-1), Inductible Nitric Oxide Synthase (iNOS), Arginase 1 (Arg-1), Tumor necrosis factor-α (TNF-α), Interleukin 1ß (IL-1ß), Cysteine histoproteinase S (CTSS), Fractalkine, chemokine C-X3-C motif receptor 1 (CX3CR1), BCL-2-associated X protein (BAX), and antiapoptotic proteins (Bcl-2) were detected using immunofluorescence, qRT-PCR as well as Western blot, respectively. RESULTS: Relative to the Sham group, treatment with TP attenuated the increased NS, infarct area and apoptosis levels observed in MCAO/R mice. Upregulated expression levels of IBA-1, iNOS, Arg-1, TNF-α and IL-1ß were found in MCAO/R mice, while TP suppressed iNOS, TNF-α and IL-1ß expression, and enhanced Arg-1 expression in both MCAO/R mice and LPS-stimulated BV-2 cells. Besides, TP inhibited the CTSS/Fractalkine/CX3CR1 pathway activation in both MCAO/R mice and LPS-induced BV-2 cells, while overexpression of CTSS reversed such effect. Co-culturing HT-22 cells with TP+LPS-treated BV-2 cells led to enhanced cell viability and decreased apoptosis levels. However, overexpression of CTSS further aggravated HT-22 cell injury. CONCLUSION: TP inhibits not only microglia polarization towards the M1 phenotype by suppressing the CTSS/Fractalkine/CX3CR1 pathway activation, but also HT-22 apoptosis by crosstalk with BV-2 cells, thereby ameliorating CIRI. These findings reveal a novel mechanism of TP in improving CIRI, and offer potential implications for addressing the preventive and therapeutic strategies of CIRI.


Sujet(s)
Encéphalopathie ischémique , Diterpènes , Composés époxy , Infarctus du territoire de l'artère cérébrale moyenne , Phénanthrènes , Lésion d'ischémie-reperfusion , Transduction du signal , Animaux , Mâle , Souris , Apoptose/effets des médicaments et des substances chimiques , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/métabolisme , Chimiokine CX3CL1/effets des médicaments et des substances chimiques , Chimiokine CX3CL1/métabolisme , Récepteur-1 de la chimiokine CX3C/effets des médicaments et des substances chimiques , Récepteur-1 de la chimiokine CX3C/métabolisme , Modèles animaux de maladie humaine , Diterpènes/pharmacologie , Composés époxy/pharmacologie , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Souris de lignée C57BL , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Neuroprotecteurs/pharmacologie , Phénanthrènes/pharmacologie , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...