Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.126
Filtrer
1.
Methods Mol Biol ; 2854: 9-18, 2025.
Article de Anglais | MEDLINE | ID: mdl-39192113

RÉSUMÉ

Antiviral innate immunity is the first line of defence against viruses. The interferon (IFN) signaling pathway, the DNA damage response (DDR), apoptosis, endoplasmic reticulum (ER) stress, and autophagy are involved in antiviral innate immunity. Viruses abrogate the antiviral immune response of cells to replication in various ways. Viral genes/proteins play a key role in evading antiviral innate immunity. Here, we will discuss the interference of viruses with antiviral innate immunity and the strategy for identifying viral gene/protein immune evasion.


Sujet(s)
Immunité innée , Humains , Protéines virales/immunologie , Protéines virales/génétique , Virus/immunologie , Virus/génétique , Échappement immunitaire , Maladies virales/immunologie , Maladies virales/virologie , Animaux , Gènes viraux , Autophagie/immunologie , Interactions hôte-pathogène/immunologie , Transduction du signal/immunologie
2.
Front Immunol ; 15: 1426204, 2024.
Article de Anglais | MEDLINE | ID: mdl-39139569

RÉSUMÉ

Autophagy is a regulated intracellular catabolic process by which invading pathogens, damaged organelles, aggregated proteins, and other macromolecules are degraded in lysosomes. It has been widely appreciated that autophagic activity plays an important role in regulating the development, fate determination, and function of cells in the immune system, including B lymphocytes. Autophagy encompasses several distinct pathways that have been linked to B cell homeostasis and function. While B cell presentation of major histocompatibility complex (MHC) class II-restricted cytosolic antigens to T cells involves both macroautophagy and chaperone-mediated autophagy (CMA), plasma cells and memory B cells mainly rely on macroautophagy for their survival. Emerging evidence indicates that core autophagy factors also participate in processes related to yet clearly distinct from classical autophagy. These autophagy-related pathways, referred to as noncanonical autophagy or conjugation of ATG8 to single membranes (CASM), contribute to B cell homeostasis and functions, including MHC class II-restricted antigen presentation to T cells, germinal center formation, plasma cell differentiation, and recall responses. Dysregulation of B cell autophagy has been identified in several autoimmune and autoinflammatory diseases such as systemic lupus erythematosus, rheumatoid arthritis, and inflammatory bowel disease. In this review, we discuss recent advances in understanding the role of canonical and noncanonical autophagy in B cells, including B cell development and maturation, antigen processing and presentation, pathogen-specific antibody responses, cytokine secretion, and autoimmunity. Unraveling the molecular mechanisms of canonical and noncanonical autophagy in B cells will improve our understanding of B cell biology, with implications for the development of autophagy-based immunotherapies.


Sujet(s)
Autophagie , Lymphocytes B , Humains , Autophagie/immunologie , Animaux , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Présentation d'antigène/immunologie , Homéostasie/immunologie , Transduction du signal
3.
Front Immunol ; 15: 1425443, 2024.
Article de Anglais | MEDLINE | ID: mdl-39104538

RÉSUMÉ

T cells, as a major lymphocyte population involved in the adaptive immune response, play an important immunomodulatory role in the early stages of autoimmune diseases. Autophagy is a cellular catabolism mediated by lysosomes. Autophagy maintains cell homeostasis by recycling degraded cytoplasmic components and damaged organelles. Autophagy has a protective effect on cells and plays an important role in regulating T cell development, activation, proliferation and differentiation. Autophagy mediates the participation of T cells in the acquired immune response and plays a key role in antigen processing as well as in the maintenance of T cell homeostasis. In autoimmune diseases, dysregulated autophagy of T cells largely influences the pathological changes. Therefore, it is of great significance to study how T cells play a role in the immune mechanism of autoimmune diseases through autophagy pathway to guide the clinical treatment of diseases.


Sujet(s)
Maladies auto-immunes , Autophagie , Lymphocytes T , Humains , Autophagie/immunologie , Maladies auto-immunes/immunologie , Animaux , Lymphocytes T/immunologie , Activation des lymphocytes/immunologie
4.
Immun Inflamm Dis ; 12(8): e1336, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39189796

RÉSUMÉ

BACKGROUND: Intestinal inflammation has various causes and leads to some inflammatory diseases, of which autophagy pathway dysfunction could be considered as one of them. Probiotics could have a positive effect on reducing inflammation by activating the autophagy pathway. To evaluate the precise effects of probiotics as preventive and therapeutic agents to control the symptoms of inflammatory diseases, we aimed to investigate the efficacy of Lactobacillus spp. in regulating the autophagy signaling pathway. METHODS: A quantitative real-time polymerase chain reaction assay was used to analyze the expression of autophagy genes involved in the formation of phagophores, autophagosomes, and autolysosomes after exposing the HT-29 cell line to sonicated pathogens and adding Lactobacillus spp. before, after, and simultaneously with inflammation. A cytokine assay was also accomplished to evaluate the interleukin (IL)-6 and IL-1ß level following the probiotic treatment. RESULTS: Lactobacillus spp. generally increased autophagy gene expression and consumption of Lactobacillus spp. before, simultaneously, and after inflammation, ultimately leading to activate autophagy pathways. The proinflammatory cytokines including IL-6 and IL-1ß decreased after probiotic treatment. CONCLUSIONS: Our native probiotic Lactobacillus spp. showed beneficial effects on HT-29 cells by increasing autophagy gene expression and decreasing the proinflammatory cytokines production in all treatments. Therefore, this novel probiotic cocktail Lactobacillus spp. can prevent and treat inflammation-related diseases.


Sujet(s)
Autophagie , Inflammation , Lactobacillus , Probiotiques , Transduction du signal , Autophagie/immunologie , Humains , Probiotiques/usage thérapeutique , Probiotiques/pharmacologie , Cellules HT29 , Inflammation/prévention et contrôle , Inflammation/immunologie , Inflammation/thérapie , Cytokines/métabolisme , Interleukine-1 bêta/métabolisme , Interleukine-6/métabolisme
5.
Aging (Albany NY) ; 16(15): 11755-11768, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39120579

RÉSUMÉ

Various immune pathways have been identified in the host, including TH1, TH2, TH3, TH9, TH17, TH22, TH1-like, and THαß immune reactions. While TH2 and TH9 responses primarily target multicellular parasites, host immune pathways directed against viruses, intracellular microorganisms (such as bacteria, protozoa, and fungi), and extracellular microorganisms can employ programmed cell death mechanisms to initiate immune responses or execute effective strategies for pathogen elimination. The types of programmed cell death involved include apoptosis, autophagy, pyroptosis, ferroptosis, necroptosis, and NETosis. Specifically, apoptosis is associated with host anti-virus eradicable THαß immunity, autophagy with host anti-virus tolerable TH3 immunity, pyroptosis with host anti-intracellular microorganism eradicable TH1 immunity, ferroptosis with host anti-intracellular microorganism tolerable TH1-like immunity, necroptosis with host anti-extracellular microorganism eradicable TH22 immunity, and NETosis with host anti-extracellular microorganism tolerable TH17 immunity.


Sujet(s)
Nécroptose , Humains , Animaux , Nécroptose/immunologie , Apoptose/immunologie , Mort cellulaire/immunologie , Autophagie/immunologie , Interactions hôte-pathogène/immunologie , Pyroptose/immunologie
6.
Microbiol Spectr ; 12(8): e0047324, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-38980014

RÉSUMÉ

Innate immune cells, such as macrophages, mount an immune response upon exposure to antigens and pathogens. Emerging evidence shows that macrophages exposed to an antigen can generate a "memory-like" response (a.k.a. trained immunity), which confers a non-specific and enhanced response upon subsequent stimulation with a second antigen/microbe. This trained immunity has been implicated in the enhanced response of macrophages against several invading pathogens. However, the association between the nature of the antigen and the corresponding immune correlate of elicited trained immunity is not fully understood. Similarly, the response of macrophages trained and restimulated with homologous stimulants to subsequent infection by pathogenic Mycobacterium tuberculosis (Mtb) remains unexplored. Here, we report the immune and metabolic profiles of trained immunity in human THP-1-derived macrophages after homologous training and restimulation with BCG, LPS, purified protein Derivative (PPD), heat-killed Mtb strains HN878 (hk-HN), and CDC1551 (hk-CDC). Furthermore, the impact of training on the autophagic and antimicrobial responses of macrophages with or without subsequent infection by clinical Mtb isolates HN878 and CDC1551 was evaluated. Results show that repeated stimulation of macrophages with different antigens displays distinct pro-inflammatory, metabolic, antimicrobial, and autophagy induction profiles. These macrophages also induce a differential antimicrobial response upon infection with clinical Mtb HN878 and CDC1551 isolates. A significantly reduced intracellular bacterial load was noted in the stimulated macrophages, which was augmented by the addition of rapamycin, an autophagy inducer. These observations suggest that the nature of the antigen and the mode of stimulation shape the magnitude and breadth of macrophage innate memory response, which impacts subsequent response to Mtb infection. IMPORTANCE: Trained immunity (a.k.a. innate memory response) is a novel concept that has been rapidly emerging as a mechanism underpinning the non-specific immunity of innate immune cells, such as macrophages. However, the association between the nature of the stimuli and the corresponding immune correlate of trained immunity is not fully understood. Similarly, the kinetics of immunological and metabolic characteristics of macrophages upon "training" by the same antigen as primary and secondary stimuli (homologous stimulation) are not fully characterized. Furthermore, the ability of antigens such as purified protein derivative (PPD) and heat-killed-Mtb to induce trained immunity remains unknown. Similarly, the response of macrophages primed and trained by homologous stimulants to subsequent infection by pathogenic Mtb is yet to be reported. In this study, we evaluated the hypothesis that the nature of the stimuli impacts the depth and breadth of trained immunity in macrophages, which differentially affects their response to Mtb infection.


Sujet(s)
Antigènes bactériens , Immunité innée , Mémoire immunologique , Macrophages , Mycobacterium tuberculosis , Tuberculose , Mycobacterium tuberculosis/immunologie , Macrophages/immunologie , Macrophages/microbiologie , Humains , Immunité innée/immunologie , Antigènes bactériens/immunologie , Mémoire immunologique/immunologie , Tuberculose/immunologie , Tuberculose/microbiologie , Cytokines/métabolisme , Cytokines/immunologie , Autophagie/immunologie , Cellules THP-1
7.
Immun Inflamm Dis ; 12(7): e1339, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38990187

RÉSUMÉ

BACKGROUND: Osteomyelitis (OM) is recognized as a significant challenge in orthopedics due to its complex immune and inflammatory responses. The prognosis heavily depends on timely diagnosis, accurate classification, and assessment of severity. Thus, the identification of diagnostic and classification-related genes from an immunological standpoint is crucial for the early detection and tailored treatment of OM. METHODS: Transcriptomic data for OM was sourced from the Gene Expression Omnibus (GEO) database, leading to the identification of autophagy- and immune-related differentially expressed genes (AIR-DEGs) through differential expression analysis. Diagnostic and classification models were subsequently developed. The CIBERSORT algorithm was utilized to examine immune cell infiltration in OM, and the relationship between OM clusters and various immune cells was explored. Key AIR-DEGs were further validated through the creation of OM animal models. RESULTS: Analysis of the transcriptomic data revealed three AIR-DEGs that played a significant role in immune responses and pathways. Nomogram and receiver operating characteristic curve analyses were performed, demonstrating excellent diagnostic capability for differentiating between OM patients and healthy individuals, with an area under the curve of 0.814. An unsupervised clustering analysis discerned two unique patterns of autophagy- and immune-related genes, as well as gene patterns. Further exploration into immune infiltration exhibited notable variances across different subtypes, especially between OM cluster 1 and gene cluster A, highlighting their potential role in mitigating inflammatory responses by regulating immune activities. Moreover, the mRNA and protein expression levels of three AIR-DEGs in the animal model were aligned with those in the training and validation data sets. CONCLUSIONS: From an immunological perspective, a diagnostic model was successfully developed, and two distinct clustering patterns were identified. These contributions offer a significant resource for the early detection and personalized immunotherapy of patients with OM.


Sujet(s)
Autophagie , Marqueurs biologiques , Modèles animaux de maladie humaine , Analyse de profil d'expression de gènes , Ostéomyélite , Ostéomyélite/diagnostic , Ostéomyélite/immunologie , Ostéomyélite/génétique , Animaux , Autophagie/génétique , Autophagie/immunologie , Humains , Souris , Transcriptome
8.
Semin Immunopathol ; 46(3-4): 6, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39042263

RÉSUMÉ

Galectins, a family of glycan-binding proteins have been shown to bind a wide range of glycans. In the cytoplasm, these glycans can be endogenous (or "self"), originating from damaged endocytic vesicles, or exogenous (or "non-self"), found on the surface of invading microbial pathogens. Galectins can detect these unusual cytosolic exposures to glycans and serve as critical regulators in orchestrating immune responses in innate and adaptive immunity. This review provides an overview of how galectins modulate host cellular responses, such as autophagy, xenophagy, and inflammasome-dependent cell death program, to infection.


Sujet(s)
Autophagie , Galectines , Inflammasomes , Humains , Autophagie/immunologie , Galectines/métabolisme , Galectines/immunologie , Inflammasomes/métabolisme , Animaux , Immunité innée , Interactions hôte-pathogène/immunologie , Transduction du signal , Immunité acquise
9.
Front Biosci (Landmark Ed) ; 29(7): 273, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39082353

RÉSUMÉ

BACKGROUND: Understanding the mechanisms through which interferon (IFN) signaling is negatively regulated is crucial for preserving the equilibrium of innate immune reactions, as the innate immune system functions, such as the original barrier, combat threats to the host. Although the function of the encephalomyocarditis virus (EMCV) viral proteins in antagonizing innate immunity has been related to earlier studies, the precise mechanism underlying the role of viral protein 3 (VP3) in type I IFN has yet to be fully illuminated. METHODS: VP3 expression and many other adaptor molecules belonging to type I IFN pathway expression levels were evaluated using Western blotting. The IFN and other antiviral genes, such as interferon-stimulated genes (ISGs) 15 and 56, were assessed by real-time quantitative polymerase chain reaction (RT-qPCR). A 50% tissue culture infectious dose (TCID50) assay was utilized to explore the effect of VP3 on EMCV proliferation in human embryonic kidney (HEK293) cells. Co-immunoprecipitation (Co-IP) assays and confocal microscope analysis were used to investigate the underlying mechanisms mediated by VP3. RESULTS: We discovered that the VP3 of EMCV acts as a suppressor of innate immune reactions. Increased levels of VP3 enhance viral reproduction through modulation of innate immune signaling pathways and suppression of antiviral responses. Additional information indicated that during viral infection, the VP3 of EMCV enhances autophagy and interacts specifically with mitochondrial antiviral signaling protein (MAVS), leading to its degradation in an autophagy pathway that relies on p62. CONCLUSIONS: Our findings showed that EMCV developed a tactic to combat host antiviral defenses by using autophagy to break down a protein that controls the innate immune response following a viral infection of the host. Notably, VP3 plays an important role in this process. Overall, these discoveries may provide a novel therapeutic target for EMCV.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Autophagie , Virus de l'encéphalomyocardite , Interféron de type I , Transduction du signal , Humains , Virus de l'encéphalomyocardite/immunologie , Virus de l'encéphalomyocardite/métabolisme , Autophagie/immunologie , Interféron de type I/métabolisme , Interféron de type I/immunologie , Cellules HEK293 , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Immunité innée , Protéolyse
10.
Front Immunol ; 15: 1397117, 2024.
Article de Anglais | MEDLINE | ID: mdl-39040107

RÉSUMÉ

Intestinal epithelial cells possess the requisite molecular machinery to initiate cell-intrinsic defensive responses against intracellular pathogens, including intracellular parasites. Interferons(IFNs) have been identified as cornerstones of epithelial cell-intrinsic defense against such pathogens in the gastrointestinal tract. Long non-coding RNAs (lncRNAs) are RNA transcripts (>200 nt) not translated into protein and represent a critical regulatory component of mucosal defense. We report here that lncRNA Nostrill facilitates IFN-γ-stimulated intestinal epithelial cell-intrinsic defense against infection by Cryptosporidium, an important opportunistic pathogen in AIDS patients and a common cause of diarrhea in young children. Nostrill promotes transcription of a panel of genes controlled by IFN-γ through facilitating Stat1 chromatin recruitment and thus, enhances expression of several genes associated with cell-intrinsic defense in intestinal epithelial cells in response to IFN-γ stimulation, including Igtp, iNos, and Gadd45g. Induction of Nostrill enhances IFN-γ-stimulated intestinal epithelial defense against Cryptosporidium infection, which is associated with an enhanced autophagy in intestinal epithelial cells. Our findings reveal that Nostrill enhances the transcription of a set of genes regulated by IFN-γ in intestinal epithelial cells. Moreover, induction of Nostrill facilitates the IFN-γ-mediated epithelial cell-intrinsic defense against cryptosporidial infections.


Sujet(s)
Cryptosporidiose , Interféron gamma , Muqueuse intestinale , ARN long non codant , Interféron gamma/métabolisme , ARN long non codant/génétique , Cryptosporidiose/immunologie , Muqueuse intestinale/immunologie , Muqueuse intestinale/parasitologie , Muqueuse intestinale/métabolisme , Animaux , Humains , Transcription génétique , Cellules épithéliales/immunologie , Cellules épithéliales/métabolisme , Cellules épithéliales/parasitologie , Souris , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-1/génétique , Cryptosporidium/génétique , Cryptosporidium/immunologie , Régulation de l'expression des gènes , Autophagie/immunologie
11.
J Gene Med ; 26(7): e3712, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38949072

RÉSUMÉ

Aggrephagy, a type of autophagy, degrades the aggregation of misfolded protein in cells. However, the role of aggrephagy in multiple myeloma (MM) has not been fully demonstrated. In this study, we first investigated the correlation between aggrephagy signaling, MM immune microenvironment composition and disease prognosis. Single-cell RNA-seq data, including the expression profiles of 12,187 single cells from seven MM bone marrow (BM) and seven healthy BM samples, were analyzed by non-negative matrix factorization for 44 aggrephagy-related genes. Bulk RNA-seq cohorts from the Gene Expression Omnibus database were used to evaluate the prognostic value of aggrephagy-related immune cell subtypes and predict immune checkpoint blockade immunotherapeutic response in MM. Compared with healthy BM, MM BM exhibited different patterns of aggrephagy-related gene expression. In MM BM, macrophages, CD8+ T cells, B cells and natural killer cells could be grouped into four to nine aggrephagy-related subclusters. The signature of aggrephagy signaling molecule expression in the immune cells correlates with the patient's prognosis. Our investigation provides a novel view of aggrephagy signaling in MM tumor microenvironment cells, which might be a prognostic indicator and potential target for MM treatment.


Sujet(s)
Myélome multiple , Transduction du signal , Analyse sur cellule unique , Microenvironnement tumoral , Myélome multiple/génétique , Myélome multiple/immunologie , Humains , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Analyse sur cellule unique/méthodes , Pronostic , Régulation de l'expression des gènes tumoraux , Autophagie/génétique , Autophagie/immunologie , Analyse de profil d'expression de gènes/méthodes , Marqueurs biologiques tumoraux/génétique , Transcriptome
12.
Allergol Immunopathol (Madr) ; 52(4): 97-103, 2024.
Article de Anglais | MEDLINE | ID: mdl-38970272

RÉSUMÉ

INTRODUCTION AND OBJECTIVES: Macrophage-induced inflammation plays a key role in defense against injury and harmful pathogens. Autophagy and the inflammatory response are associated; however, the relationship between the autophagy pathway and lipopolysaccharide (LPS)- induced inflammatory responses remains unknown. We aimed to determine the effect of autophagy on the LPS-induced myeloid differentiation factor 88 (MyD88)/nuclear transcription factor kB (NF-kB) pathway-mediated inflammatory response in RAW264.7 cells. MATERIALS AND METHODS: To determine the effect of autophagy on the LPS-induced inflammatory response, using various in vitro assays, we determined the effect of autophagy inhibitors and inducers on the inflammatory response in RAW264.7 cells. RESULTS: Chloroquine (CQ), an autophagy inhibitor, suppressed pro-inflammatory cytokines, including interleukin (IL)-1ß, IL-6, and tumor necrosis factor α (TNFα) in LPS-stimulated RAW264.7 cells. CQ also affected inflammatory mediators such as myeloid differentiation factor 88 and NF-kB in LPS-stimulated RAW264.7 cells. CONCLUSION: This study demonstrated that CQ regulates the LPS-induced inflammatory response in RAW264.7 cells. We propose that targeting the regulation of pro-inflammatory cytokine levels and inflammatory mediators using CQ is a promising therapeutic approach for preventing inflammatory injury. CQ serves as a potential therapeutic target for treating various inflammatory diseases.


Sujet(s)
Chloroquine , Cytokines , Lipopolysaccharides , Macrophages , Facteur de différenciation myéloïde-88 , Facteur de transcription NF-kappa B , Animaux , Souris , Chloroquine/pharmacologie , Cellules RAW 264.7 , Facteur de transcription NF-kappa B/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Macrophages/métabolisme , Cytokines/métabolisme , Facteur de différenciation myéloïde-88/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Autophagie/immunologie , Inflammation/immunologie , Inflammation/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Anti-inflammatoires/pharmacologie , Médiateurs de l'inflammation/métabolisme
13.
Front Immunol ; 15: 1321657, 2024.
Article de Anglais | MEDLINE | ID: mdl-38975346

RÉSUMÉ

Tuberculosis (TB) remains a significant global health challenge, with approximately 1.5 million deaths per year. The Bacillus Calmette-Guérin (BCG) vaccine against TB is used in infants but shows variable protection. Here, we introduce a novel approach using a double gene knockout mutant (DKO) from wild-type Mycobacterium tuberculosis (Mtb) targeting fbpA and sapM genes. DKO exhibited enhanced anti-TB gene expression in mouse antigen-presenting cells, activating autophagy and inflammasomes. This heightened immune response improved ex vivo antigen presentation to T cells. Subcutaneous vaccination with DKO led to increased protection against TB in wild-type C57Bl/6 mice, surpassing the protection observed in caspase 1/11-deficient C57Bl/6 mice and highlighting the critical role of inflammasomes in TB protection. The DKO vaccine also generated stronger and longer-lasting protection than the BCG vaccine in C57Bl/6 mice, expanding both CD62L-CCR7-CD44+/-CD127+ effector T cells and CD62L+CCR7+/-CD44+CD127+ central memory T cells. These immune responses correlated with a substantial ≥ 1.7-log10 reduction in Mtb lung burden. The DKO vaccine represents a promising new approach for TB immunization that mediates protection through autophagy and inflammasome pathways.


Sujet(s)
Macrophages , Souris de lignée C57BL , Mycobacterium tuberculosis , Vaccins antituberculeux , Tuberculose , Animaux , Mycobacterium tuberculosis/immunologie , Souris , Macrophages/immunologie , Tuberculose/immunologie , Tuberculose/prévention et contrôle , Vaccins antituberculeux/immunologie , Antigènes bactériens/immunologie , Antigènes bactériens/génétique , Inflammasomes/immunologie , Femelle , Vaccin BCG/immunologie , Autophagie/immunologie , Protéines bactériennes/immunologie , Protéines bactériennes/génétique , Modèles animaux de maladie humaine
14.
Fish Shellfish Immunol ; 151: 109719, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38914181

RÉSUMÉ

Sequestosome 1 (SQSTM1/p62) is a selective autophagy adapter protein that participates in antiviral and bacterial immune responses and plays an important regulatory role in clearing the proteins to be degraded and maintaining intracellular protein homeostasis. In this study, two p62 genes were cloned from common carp (Cyprinus carpio), namely Ccp62-1 and Ccp62-2, and conducted bioinformatics analysis on them. The results showed that Ccp62s had the same structural domain (Phox and Bem1 domain, ZZ-type zinc finger domain, and ubiquitin-associated domain) as p62 from other species. Ccp62s were widely expressed in various tissues of fish, and highly expressed in immune organs such as gills, spleen, head kidney, etc. Subcellular localization study showed that they were mainly distributed in punctate aggregates in the cytoplasm. After stimulation with Aeromonas hydrophila and spring viraemia of carp virus (SVCV), the expression level of Ccp62s was generally up-regulated. Overexpression of Ccp62s in EPC cells could inhibit SVCV replication. Upon A. hydrophila challenge, the bacterial load in Ccp62s-overexpressing group was significantly reduced, the expression levels of pro-inflammatory cytokines and interferon factors were increased, and the survival rate of the fish was improved. These results indicated that Ccp62s were involved in the immune response of common carp to bacterial and viral infections.


Sujet(s)
Aeromonas hydrophila , Carpes (poisson) , Maladies des poissons , Protéines de poisson , Infections bactériennes à Gram négatif , Immunité innée , Phylogenèse , Infections à Rhabdoviridae , Rhabdoviridae , Animaux , Carpes (poisson)/immunologie , Carpes (poisson)/génétique , Maladies des poissons/immunologie , Protéines de poisson/génétique , Protéines de poisson/immunologie , Aeromonas hydrophila/physiologie , Immunité innée/génétique , Rhabdoviridae/physiologie , Infections bactériennes à Gram négatif/immunologie , Infections bactériennes à Gram négatif/médecine vétérinaire , Infections à Rhabdoviridae/immunologie , Infections à Rhabdoviridae/médecine vétérinaire , Régulation de l'expression des gènes/immunologie , Séquestosome-1/génétique , Séquestosome-1/immunologie , Analyse de profil d'expression de gènes/médecine vétérinaire , Alignement de séquences/médecine vétérinaire , Séquence d'acides aminés , Autophagie/immunologie
15.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38892443

RÉSUMÉ

Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis (TB), a prevalent infectious disease affecting populations worldwide. A classic trait of TB pathology is the formation of granulomas, which wall off the pathogen, via the innate and adaptive immune systems. Some key players involved include tumor necrosis factor-alpha (TNF-α), foamy macrophages, type I interferons (IFNs), and reactive oxygen species, which may also show overlap with cell death pathways. Additionally, host cell death is a primary method for combating and controlling Mtb within the body, a process which is influenced by both host and bacterial factors. These cell death modalities have distinct molecular mechanisms and pathways. Programmed cell death (PCD), encompassing apoptosis and autophagy, typically confers a protective response against Mtb by containing the bacteria within dead macrophages, facilitating their phagocytosis by uninfected or neighboring cells, whereas necrotic cell death benefits the pathogen, leading to the release of bacteria extracellularly. Apoptosis is triggered via intrinsic and extrinsic caspase-dependent pathways as well as caspase-independent pathways. Necrosis is induced via various pathways, including necroptosis, pyroptosis, and ferroptosis. Given the pivotal role of host cell death pathways in host defense against Mtb, therapeutic agents targeting cell death signaling have been investigated for TB treatment. This review provides an overview of the diverse mechanisms underlying Mtb-induced host cell death, examining their implications for host immunity. Furthermore, it discusses the potential of targeting host cell death pathways as therapeutic and preventive strategies against Mtb infection.


Sujet(s)
Mycobacterium tuberculosis , Tuberculose , Humains , Mycobacterium tuberculosis/immunologie , Mycobacterium tuberculosis/pathogénicité , Tuberculose/immunologie , Tuberculose/microbiologie , Tuberculose/anatomopathologie , Animaux , Mort cellulaire/immunologie , Interactions hôte-pathogène/immunologie , Apoptose , Immunité innée , Autophagie/immunologie , Transduction du signal , Macrophages/immunologie , Macrophages/microbiologie
16.
Nat Commun ; 15(1): 4988, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862534

RÉSUMÉ

Cancer-associated fibroblasts (CAFs) have emerged as a dominant non-hematopoietic cell population in the tumour microenvironment, serving diverse functions in tumour progression. However, the mechanisms via which CAFs influence the anti-tumour immunity remain poorly understood. Here, using multiple tumour models and biopsies from cancer patients, we report that α-SMA+ CAFs can form immunological synapses with Foxp3+ regulatory T cells (Tregs) in tumours. Notably, α-SMA+ CAFs can phagocytose and process tumour antigens and exhibit a tolerogenic phenotype which instructs movement arrest, activation and proliferation in Tregs in an antigen-specific manner. Moreover, α-SMA+ CAFs display double-membrane structures resembling autophagosomes in their cytoplasm. Single-cell transcriptomic data showed an enrichment in autophagy and antigen processing/presentation pathways in α-SMA-expressing CAF clusters. Conditional knockout of Atg5 in α-SMA+ CAFs promoted inflammatory re-programming in CAFs, reduced Treg cell infiltration and attenuated tumour development. Overall, our findings reveal an immunosuppressive mechanism entailing the formation of synapses between α-SMA+ CAFs and Tregs in an autophagy-dependent manner.


Sujet(s)
Autophagie , Fibroblastes associés au cancer , Synapses immunologiques , Lymphocytes T régulateurs , Microenvironnement tumoral , Lymphocytes T régulateurs/immunologie , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/immunologie , Fibroblastes associés au cancer/anatomopathologie , Humains , Synapses immunologiques/métabolisme , Synapses immunologiques/immunologie , Animaux , Microenvironnement tumoral/immunologie , Souris , Autophagie/immunologie , Actines/métabolisme , Protéine-5 associée à l'autophagie/génétique , Protéine-5 associée à l'autophagie/métabolisme , Tumeurs/immunologie , Tumeurs/génétique , Tumeurs/anatomopathologie , Souris de lignée C57BL , Facteurs de transcription Forkhead/métabolisme , Facteurs de transcription Forkhead/génétique , Femelle , Souris knockout
17.
Immun Inflamm Dis ; 12(6): e1310, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38888464

RÉSUMÉ

BACKGROUND: The PI3K/Akt/mTOR pathway and autophagy are important physiological processes. But their roles in eCRSwNP remains controversial. METHODS: In this study, we used the eCRSwNP mouse model, PI3K/Akt/mTOR pathway inhibitors, and autophagy inhibitors and activators to investigate the regulatory effects of the PI3K/Akt/mTOR pathway on autophagy, and their effects on eosinophilic inflammation, and tissue remodeling. The role of ILC2s in eCRSwNP was also studied, and the relationship between ILC2s and autophagy was preliminarily determined. RESULTS: Our results show that eosinophilic inflammation in eCRSwNP mice could be inhibited by promoting the autophagy; otherwise, eosinophilic inflammation could be promoted. Meanwhile, inhibition of the PI3K/Akt/mTOR pathway can further promote autophagy and inhibit eosinophilic inflammation. Meanwhile, inhibiting the PI3K/Akt/mTOR pathway and promoting autophagy can reduce the number of ILC2s and the severity of tissue remodeling in the nasal polyps of eCRSwNP mice. CONCLUSIONS: We conclude that the PI3K/Akt/mTOR pathway plays roles in eosinophilic inflammation and tissue remodeling of eCRSwNP, in part by regulating the level of autophagy. The downregulation of autophagy is a pathogenesis of eCRSwNP; therefore, the recovery of normal autophagy levels might be a new target for eCRSwNP therapy. Furthermore, autophagy might inhibit eosinophilic inflammation and tissue remodeling, in part by reducing the number of ILC2s.


Sujet(s)
Autophagie , Immunité innée , Lymphocytes , Polypes du nez , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Sinusite , Sérine-thréonine kinases TOR , Animaux , Sérine-thréonine kinases TOR/métabolisme , Souris , Sinusite/immunologie , Sinusite/anatomopathologie , Sinusite/métabolisme , Autophagie/immunologie , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Lymphocytes/immunologie , Lymphocytes/métabolisme , Maladie chronique , Polypes du nez/immunologie , Polypes du nez/anatomopathologie , Modèles animaux de maladie humaine , Éosinophilie/immunologie , Éosinophilie/anatomopathologie , Granulocytes éosinophiles/immunologie , Granulocytes éosinophiles/anatomopathologie , Granulocytes éosinophiles/métabolisme , Souris de lignée BALB C
18.
Cell Commun Signal ; 22(1): 305, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38831299

RÉSUMÉ

As a major component of innate immunity and a positive regulator of interferons, the Stimulator of interferon gene (STING) has an immunotherapy potential to govern a variety of infectious diseases. Despite the recent advances regarding vaccines against COVID-19, nontoxic novel adjuvants with the potential to enhance vaccine efficacy are urgently desired. In this connection, it has been well-documented that STING agonists are applied to combat COVID-19. This approach is of major significance for boosting immune responses most likely through an autophagy-dependent manner in susceptible individuals against infection induced by severe acute respiratory syndrome Coronavirus (SARS­CoV­2). Given that STING agonists exert substantial immunomodulatory impacts under a wide array of pathologic conditions, these agents could be considered novel adjuvants for enhancing immunogenicity against the SARS-related coronavirus. Here, we intend to discuss the recent advances in STING agonists' recruitment to boost innate immune responses upon vaccination against SARS-related coronavirus infections. In light of the primordial role of autophagy modulation, the potential of being an antiviral vaccine adjuvant was also explored.


Sujet(s)
Autophagie , COVID-19 , Protéines membranaires , SARS-CoV-2 , Autophagie/immunologie , Autophagie/effets des médicaments et des substances chimiques , Humains , Protéines membranaires/immunologie , SARS-CoV-2/immunologie , COVID-19/immunologie , COVID-19/prévention et contrôle , Animaux , Vaccins contre la COVID-19/immunologie , Immunité innée/effets des médicaments et des substances chimiques , Adjuvants vaccinaux/usage thérapeutique , Adjuvants vaccinaux/pharmacologie , Adjuvants immunologiques/pharmacologie
19.
Immunity ; 57(7): 1603-1617.e7, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38761804

RÉSUMÉ

Recent evidence reveals hyper T follicular helper (Tfh) cell responses in systemic lupus erythematosus (SLE); however, molecular mechanisms responsible for hyper Tfh cell responses and whether they cause SLE are unclear. We found that SLE patients downregulated both ubiquitin ligases, casitas B-lineage lymphoma (CBL) and CBLB (CBLs), in CD4+ T cells. T cell-specific CBLs-deficient mice developed hyper Tfh cell responses and SLE, whereas blockade of Tfh cell development in the mutant mice was sufficient to prevent SLE. ICOS was upregulated in SLE Tfh cells, whose signaling increased BCL6 by attenuating BCL6 degradation via chaperone-mediated autophagy (CMA). Conversely, CBLs restrained BCL6 expression by ubiquitinating ICOS. Blockade of BCL6 degradation was sufficient to enhance Tfh cell responses. Thus, the compromised expression of CBLs is a prevalent risk trait shared by SLE patients and causative to hyper Tfh cell responses and SLE. The ICOS-CBLs axis may be a target to treat SLE.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Protéine inductible de costimulation du lymphocyte T , Lupus érythémateux disséminé , Souris knockout , Protéines proto-oncogènes c-bcl-6 , Protéines proto-oncogènes c-cbl , Lymphocytes T auxiliaires folliculaires , Animaux , Femelle , Humains , Souris , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Autophagie/immunologie , Protéine inductible de costimulation du lymphocyte T/métabolisme , Protéine inductible de costimulation du lymphocyte T/génétique , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/génétique , Souris de lignée C57BL , Protéolyse , Protéines proto-oncogènes c-bcl-6/métabolisme , Protéines proto-oncogènes c-bcl-6/génétique , Protéines proto-oncogènes c-cbl/métabolisme , Protéines proto-oncogènes c-cbl/génétique , Protéines proto-oncogènes c-cbl/déficit , Transduction du signal/immunologie , Lymphocytes T auxiliaires folliculaires/immunologie , Lymphocytes T auxiliaires/immunologie , Ubiquitination
20.
Free Radic Biol Med ; 222: 1-15, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38763209

RÉSUMÉ

Non-small cell lung cancer (NSCLC), particularly lung adenocarcinoma (LUAD), significantly influences cancer-related mortality and is frequently considered by poor therapeutic responses due to genetic alterations. Cancer cells possess an inclination to develop resistance to individual treatment modalities, thus it is necessary to investigate several pathways simultaneously to obtain insights that will aid in the establishment of improved therapeutic approaches. Exploring regulated cell death (RCD) mechanisms offers promising avenues to augment immunotherapy by reshaping the tumor microenvironment (TME). Here, we investigated the prospective of microwave plasma-infused nitric oxide water (NOW) to initiate immunogenic cell death (ICD) while concurrently modulating autophagy and ferroptosis signaling in LUAD-associated A549 cells. Plasma treatment results in stable NO species nitrite/nitrate (NO2-/NO3-) in the water, altering its physicochemical properties. Analysis of ICD markers reveals increased expression of damage-associated molecular patterns (DAMPs) at both protein and mRNA levels post-NOW exposure. Intracellular reactive oxygen and nitrogen species (RONS) accumulation suggests NO-mediated mitochondrial dysfunction, triggering autophagy induction. Flow cytometry and western blotting confirm alterations in autophagy regulators Beclin 1 and SQSTM1. Furthermore, NOW treatment induces lipid peroxidation and upregulates ferroptosis-associated genes, as determined by qRT-PCR. Transmission electron microscopy (TEM) imaging reveals autophagosome formation and loss of cristae structures, corroborating the occurrence of autophagy and ferroptosis. Our findings propose that NOW may considered as inducer of ICD and the stimulation of other RCD-related proteins may enhance the anti-tumor immunogenicity.


Sujet(s)
Adénocarcinome pulmonaire , Mort cellulaire par autophagie , Ferroptose , Tumeurs du poumon , Mitochondries , Monoxyde d'azote , Humains , Monoxyde d'azote/métabolisme , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Adénocarcinome pulmonaire/immunologie , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/génétique , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Mort cellulaire par autophagie/immunologie , Cellules A549 , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Eau/métabolisme , Eau/composition chimique , Microenvironnement tumoral/immunologie , Espèces réactives de l'oxygène/métabolisme , Autophagie/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE