Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 580
Filtrer
2.
J Extracell Vesicles ; 13(7): e12457, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39007430

RÉSUMÉ

Seminal plasma induces immune tolerance towards paternal allogenic antigens within the female reproductive tract and during foetal development. Recent evidence suggests a role for extracellular vesicles in seminal plasma (spEVs). We isolated spEVs from seminal plasma that was donated by vasectomized men, thereby excluding any contributions from the testis or epididymis. Previous analysis demonstrated that such isolated spEVs originate mainly from the prostate. Here we observed that when isolated fluorescently labelled spEVs were mixed with peripheral blood mononuclear cells, they were endocytosed predominantly by monocytes, and to a lesser extent also by T-cells. In a mixed lymphocyte reaction, T-cell proliferation was inhibited by spEVs. A direct effect of spEVs on T-cells was demonstrated when isolated T cells were activated by anti-CD3/CD28 coated beads. Again, spEVs interfered with T cell proliferation, as well as with the expression of CD25 and the release of IFN-γ, TNF, and IL-2. Moreover, spEVs stimulated the expression of Foxp3 and IL-10 by CD4+CD25+CD127- T cells, indicating differentiation into regulatory T-cells (Tregs). Prior treatment of spEVs with proteinase K revoked their effects on T-cells, indicating a requirement for surface-exposed spEV proteins. The adenosine A2A receptor-specific antagonist CPI-444 also reduced effects of spEVs on T-cells, consistent with the notion that the development of Tregs and their immune suppressive functions are under the influence of adenosine-A2A receptor signalling. We found that adenosine is highly enriched in spEVs and propose that spEVs are targeted to and endocytosed by T-cells, after which they may release their adenosine content into the lumen of endosomes, thus allowing endosome-localized A2A receptor signalling in spEVs targeted T-cells. Collectively, these data support the idea that spEVs can prime T cells directly for differentiation into Tregs.


Sujet(s)
Différenciation cellulaire , Vésicules extracellulaires , Sperme , Lymphocytes T régulateurs , Humains , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Sperme/métabolisme , Sperme/immunologie , Mâle , Prolifération cellulaire , Activation des lymphocytes/immunologie
3.
Front Immunol ; 15: 1397967, 2024.
Article de Anglais | MEDLINE | ID: mdl-38947317

RÉSUMÉ

Introduction: CD39 plays an important role in the immunoregulation and inhibition of effector cells. It is expressed on immune cells, including Tregs, and on extracellular vesicles (EVs) budding from the plasma membrane. Platelet transfusion may induce alloimmunization against HLA-I antigens, leading to refractoriness to platelet transfusion with severe consequences for patients. Tregs may play a key role in determining whether alloimmunization occurs in patients with hematologic disorders. We hypothesized that CD39+ EVs might play an immunoregulatory role, particularly in the context of platelet transfusions in patients with hematologic disorders. Such alloimmunization leads to the production of alloantibodies and is sensitive to the regulatory action of CD39. Methods: We characterized CD39+ EVs in platelet concentrates by flow cytometry. The absolute numbers and cellular origins of CD39+ EVs were evaluated. We also performed functional tests to evaluate interactions with immune cells and their functions. Results: We found that CD39+ EVs from platelet concentrates had an inhibitory phenotype that could be transferred to the immune cells with which they interacted: CD4+ and CD8+ T lymphocytes (TLs), dendritic cells, monocytes, and B lymphocytes (BLs). Moreover, the concentration of CD39+ EVs in platelet concentrates varied and was very high in 10% of concentrates. The number of these EVs present was determinant for EV-cell interactions. Finally, functional interactions were observed with BLs, CD4+ TLs and CD39+ EVs for immunoglobulin production and lymphoproliferation, with potential implications for the immunological management of patients.


Sujet(s)
Plaquettes , Vésicules extracellulaires , Antigène CD9 , Humains , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/métabolisme , Plaquettes/immunologie , Plaquettes/métabolisme , Antigène CD9/métabolisme , Communication cellulaire/immunologie , Transfusion de plaquettes , Femelle , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Mâle , Apyrase/métabolisme , Apyrase/immunologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Antigènes CD
4.
Gut Microbes ; 16(1): 2379633, 2024.
Article de Anglais | MEDLINE | ID: mdl-39024479

RÉSUMÉ

Gut microbiota-derived extracellular vesicles (mEVs) are reported to regulate inflammatory response by delivering bacterial products into host cells. The complement receptor of the immunoglobulin superfamily macrophages (CRIg+ Mφ) could clear invading bacteria and their derivatives. Here, we investigate the role of CRIg+ Mφ and the mechanism by which mEVs regulate intestinal inflammation. We found that it is exacerbated in IBD patients and colitis mice by mEVs' leakage from disturbed gut microbiota, enriching microbial DNA in the intestinal mucosa. CRIg+ Mφ significantly decrease in IBD patients, allowing the spread of mEVs into the mucosa. The microbial DNA within mEVs is the key trigger for inflammation and barrier function damage. The cGAS/STING pathway is crucial in mEVs-mediated inflammatory injury. Blocking cGAS/STING signaling effectively alleviates inflammation caused by mEVs leakage and CRIg+ Mφ deficiency. Microbial DNA-containing mEVs, along with CRIg+ Mφ deficiency, stimulate inflammation in IBD, with the cGAS/STING pathway playing a crucial role.


Sujet(s)
ADN bactérien , Vésicules extracellulaires , Microbiome gastro-intestinal , Inflammation , Maladies inflammatoires intestinales , Muqueuse intestinale , Macrophages , Protéines membranaires , Nucleotidyltransferases , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Animaux , Souris , Macrophages/immunologie , Macrophages/microbiologie , Macrophages/métabolisme , Maladies inflammatoires intestinales/microbiologie , Maladies inflammatoires intestinales/anatomopathologie , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/immunologie , Humains , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Nucleotidyltransferases/métabolisme , Nucleotidyltransferases/génétique , Muqueuse intestinale/microbiologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/immunologie , Inflammation/microbiologie , Inflammation/métabolisme , ADN bactérien/génétique , Souris de lignée C57BL , Mâle , Femelle , Transduction du signal , Colite/microbiologie , Colite/anatomopathologie
5.
Front Immunol ; 15: 1362120, 2024.
Article de Anglais | MEDLINE | ID: mdl-38962016

RÉSUMÉ

Cancer stem cells (CSCs), accounting for only a minor cell proportion (< 1%) within tumors, have profound implications in tumor initiation, metastasis, recurrence, and treatment resistance due to their inherent ability of self-renewal, multi-lineage differentiation, and tumor-initiating potential. In recent years, accumulating studies indicate that CSCs and tumor immune microenvironment act reciprocally in driving tumor progression and diminishing the efficacy of cancer therapies. Extracellular vesicles (EVs), pivotal mediators of intercellular communications, build indispensable biological connections between CSCs and immune cells. By transferring bioactive molecules, including proteins, nucleic acids, and lipids, EVs can exert mutual influence on both CSCs and immune cells. This interaction plays a significant role in reshaping the tumor immune microenvironment, creating conditions favorable for the sustenance and propagation of CSCs. Deciphering the intricate interplay between CSCs and immune cells would provide valuable insights into the mechanisms of CSCs being more susceptible to immune escape. This review will highlight the EV-mediated communications between CSCs and each immune cell lineage in the tumor microenvironment and explore potential therapeutic opportunities.


Sujet(s)
Vésicules extracellulaires , Tumeurs , Cellules souches tumorales , Microenvironnement tumoral , Microenvironnement tumoral/immunologie , Humains , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/métabolisme , Cellules souches tumorales/immunologie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Tumeurs/immunologie , Tumeurs/anatomopathologie , Tumeurs/thérapie , Animaux , Communication cellulaire/immunologie , Échappement de la tumeur à la surveillance immunitaire , Immunomodulation
6.
J Extracell Vesicles ; 13(7): e12480, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38978304

RÉSUMÉ

Head and neck squamous cell carcinoma (HNSCC) is a global cancer burden with a 5-year overall survival rate of around 50%, stagnant for decades. A tumour-induced immunosuppressive microenvironment contributes to HNSCC progression, with the adenosine (ADO) pathway and an upregulated expression of inhibitory immune checkpoint regulators playing a key role in this context. The correlation between high neutrophil-to-lymphocyte ratio (NLR) with advanced tumour staging suggests involvement of neutrophils (NØ) in cancer progression. Interestingly, we associated a high NLR with an increased intracellular PD-L1 localization in primary HNSCC samples, potentially mediating more aggressive tumour characteristics and therefore synergistically favouring tumour progression. Still, further research is needed to harness this knowledge for effective treatments and overcome resistance. Since it is hypothesized that the tumour microenvironment (TME) may be influenced by small extracellular vesicles (sEVs) secreted by tumours (TEX), this study aims to investigate the impact of HNSCC-derived TEX on NØ and blockade of ADO receptors as a potential strategy to reverse the pro-tumour phenotype of NØ. UMSCC47-TEX exhibited CD73 enzymatic activity involved in ADO signalling, as well as the immune checkpoint inhibitor PD-L1. Data revealed that TEX induce chemotaxis of NØ and the sustained interaction promotes a shift into a pro-tumour phenotype, dependent on ADO receptors (P1R), increasing CD170high subpopulation, CD73 and PD-L1 expression, followed by an immunosuppressive secretome. Blocking A3R reduced CD73 and PD-L1 expression. Co-culture experiments with HNSCC cells demonstrated that TEX-modulated NØ increase the CD73/PD-L1 axis, through Cyclin D-CDK4/6 signalling. To support these findings, the CAM model with primary tumour was treated with NØ supernatant. Moreover, these NØ promoted an increase in migration, invasion, and reduced cell death. Targeting P1R on NØ, particularly A3R, exhibited potential therapeutic strategy to counteract immunosuppression in HNSCC. Understanding the TEX-mediated crosstalk between tumours and NØ offers insights into immunomodulation for improving cancer therapies.


Sujet(s)
5'-Nucleotidase , Antigène CD274 , Vésicules extracellulaires , Tumeurs de la tête et du cou , Granulocytes neutrophiles , Transduction du signal , Carcinome épidermoïde de la tête et du cou , Microenvironnement tumoral , Humains , Antigène CD274/métabolisme , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/immunologie , Microenvironnement tumoral/immunologie , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/métabolisme , 5'-Nucleotidase/métabolisme , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/métabolisme , Tumeurs de la tête et du cou/anatomopathologie , Lignée cellulaire tumorale , Immunomodulation , Adénosine/métabolisme , Protéines liées au GPI
7.
Methods Mol Biol ; 2821: 225-236, 2024.
Article de Anglais | MEDLINE | ID: mdl-38997493

RÉSUMÉ

Many researchers are interested in the possibility of manipulating the targeting specificity of extracellular vesicles (EVs) for their use as physiological delivery vehicles for drugs and bioactive molecules. Our studies demonstrated the possibility of directing EVs toward the desired acceptor cell by coating them with antigen-specific antibody light chains. Here, we describe the methods for detection of the presence of antibody light chains on the EV surface, proving their ability to specifically bind the antigen and for separating the antigen-binding EV subpopulation.


Sujet(s)
Antigènes , Vésicules extracellulaires , Chaines légères des immunoglobulines , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/composition chimique , Humains , Chaines légères des immunoglobulines/métabolisme , Chaines légères des immunoglobulines/composition chimique , Antigènes/immunologie , Cytométrie en flux/méthodes
8.
J Extracell Vesicles ; 13(7): e12484, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39041344

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is characterised by immune evasion that contribute to poor prognosis. Cancer-associated fibroblasts (CAFs) play a pivotal role in orchestrating the PDAC tumour microenvironment. We investigated the role of CAF-derived extracellular vesicle (EV)-packaged long non-coding RNAs (lncRNAs) in immune evasion and explored gene therapy using engineered EVs loading small interfering RNAs (siRNAs) as a potential therapeutic strategy. Our findings highlight the significance of EV-packaged lncRNA RP11-161H23.5 from CAF in promoting PDAC immune evasion by downregulating HLA-A expression, a key component of antigen presentation. Mechanistically, RP11-161H23.5 forms a complex with CNOT4, a subunit of the mRNA deadenylase CCR4-NOT complex, enhancing the degradation of HLA-A mRNA by shortening its poly(A) tail. This immune evasion mechanism compromises the anti-tumour immune response. To combat this, we propose an innovative approach utilising engineered EVs as natural and biocompatible nanocarriers for siRNA-based gene therapy and this strategy holds promise for enhancing the effectiveness of immunotherapy in PDAC. Overall, our study sheds light on the critical role of CAF-derived EV-packaged lncRNA RP11-161H23.5/CNOT4/HLA-A axis in PDAC immune evasion and presents a novel avenue for therapeutic intervention.


Sujet(s)
Fibroblastes associés au cancer , Carcinome du canal pancréatique , Vésicules extracellulaires , Antigènes HLA-A , Tumeurs du pancréas , ARN long non codant , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/immunologie , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/thérapie , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/thérapie , Lignée cellulaire tumorale , Antigènes HLA-A/génétique , Antigènes HLA-A/immunologie , Antigènes HLA-A/métabolisme , Échappement immunitaire , Régulation de l'expression des gènes tumoraux , Régulation négative , Petit ARN interférent , Microenvironnement tumoral/immunologie , Animaux , Échappement de la tumeur à la surveillance immunitaire , Souris
9.
Sci Rep ; 14(1): 16970, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39043800

RÉSUMÉ

B cells and the antibodies they produce are critical in host defense against pathogens and contribute to various immune-mediated diseases. B cells responding to activating signals in vitro release extracellular vesicles (EV) that carry surface antibodies, yet B cell production of EVs that express antibodies and their function in vivo is incompletely understood. Using transgenic mice expressing the Cre recombinase in B cells switching to IgG1 to induce expression of fusion proteins between emerald green fluorescent protein (emGFP) and the EV tetraspanin CD63 as a model, we identify emGFP expression in B cells responding to foreign antigen in vivo and characterize the emGFP+ EVs they release. Our data suggests that emGFP+ germinal center B cells undergoing immunoglobulin class switching to express IgG and their progeny memory B cells and plasma cells, also emGFP+, are sources of circulating antigen-specific IgG+ EVs. Furthermore, using a mouse model of influenza virus infection, we find that IgG+ EVs specific for the influenza hemagglutinin antigen protect against virus infection. In addition, crossing the B cell Cre driver EV reporter mice onto the Nba2 lupus-prone strain revealed increased circulating emGFP+ EVs that expressed surface IgG against nuclear antigens linked to autoimmunity. These data identify EVs loaded with antibodies as a novel route for antibody secretion in B cells that contribute to adaptive immune responses, with important implications for different functions of IgG+ EVs in infection and autoimmunity.


Sujet(s)
Lymphocytes B , Vésicules extracellulaires , Immunoglobuline G , Souris transgéniques , Animaux , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/métabolisme , Immunoglobuline G/immunologie , Immunoglobuline G/métabolisme , Souris , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Infections à Orthomyxoviridae/immunologie , Protéines à fluorescence verte/métabolisme , Protéines à fluorescence verte/génétique , Antigènes/immunologie , Commutation de classe des immunoglobulines , Souris de lignée C57BL , Centre germinatif/immunologie , Centre germinatif/métabolisme
10.
An Acad Bras Cienc ; 96(2): e20231212, 2024.
Article de Anglais | MEDLINE | ID: mdl-38922279

RÉSUMÉ

The tumor microenvironment (TME) harbors several cell types, such as tumor cells, immune cells, and non-immune cells. These cells communicate through several mechanisms, such as cell-cell contact, cytokines, chemokines, and extracellular vesicles (EVs). Tumor-derived vesicles are known to have the ability to modulate the immune response. Monocytes are a subset of circulating innate immune cells and play a crucial role in immune surveillance, being recruited to tissues where they differentiate into macrophages. In the context of tumors, it has been observed that tumor cells can attract monocytes to the TME and induce their differentiation into tumor-associated macrophages with a pro-tumor phenotype. Tumor-derived EVs have emerged as essential structures mediating this process. Through the transfer of specific molecules and signaling factors, tumor-derived EVs can shape the phenotype and function of monocytes, inducing the expression of cytokines and molecules by these cells, thus modulating the TME towards an immunosuppressive environment.


Sujet(s)
Vésicules extracellulaires , Macrophages , Monocytes , Tumeurs , Microenvironnement tumoral , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/physiologie , Humains , Monocytes/immunologie , Microenvironnement tumoral/immunologie , Tumeurs/immunologie , Tumeurs/anatomopathologie , Macrophages/immunologie , Cytokines , Différenciation cellulaire
11.
J Extracell Vesicles ; 13(7): e12471, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38944672

RÉSUMÉ

Haematopoiesis dysregulation with the presence of immature myeloid and erythroid immunosuppressive cells are key characteristics of the immune escape phase of tumour development. Here, the role of in vitro generated B16F10 tumour cell-derived extracellular vesicles (tEVs) as indirect cellular communicators, participating in tumour-induced dysregulation of haematopoiesis, was explored. The isolated tEVs displayed features of small EVs with a size range of 100-200 nm, expressed the common EV markers CD63, CD9, and Alix, and had a spherical shape with a lipid bilayer membrane. Proteomic profiling revealed significant levels of angiogenic factors, particularly vascular endothelial growth factor (VEGF), osteopontin, and tissue factor, associated with the tEVs. Systemic administration of these tEVs in syngeneic mice induced splenomegaly and disrupted haematopoiesis, leading to extramedullary haematopoiesis, expansion of splenic immature erythroid progenitors, reduced bone marrow cellularity, medullary expansion of granulocytic myeloid suppressor cells, and the development of anaemia. These effects closely mirrored those observed in tumour-bearing mice and were not seen after heat inactivating the tEVs. In vitro studies demonstrated that tEVs independently induced the expansion of bone marrow granulocytic myeloid suppressor cells and B cells while reducing the frequency of cells in the erythropoietic lineage. These effects of tEVs were significantly abrogated by the blockade of VEGF or heat inactivation. Our findings underscore the important role of tEVs in dysregulating haematopoiesis during the immune escape phase of cancer immunoediting, suggesting their potential as targets for addressing immune evasion and reinstating normal hematopoietic processes.


Sujet(s)
Vésicules extracellulaires , Hématopoïèse , Animaux , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Souris , Mélanome expérimental/métabolisme , Mélanome expérimental/immunologie , Mélanome expérimental/anatomopathologie , Souris de lignée C57BL , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Lignée cellulaire tumorale
12.
J Clin Invest ; 134(14)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38833312

RÉSUMÉ

BACKGROUNDPredicting immune effector cell-associated neurotoxicity syndrome (ICANS) in patients infused with CAR T cells is still a conundrum. This complication, thought to be consequent to CAR T cell activation, arises a few days after infusion, when circulating CAR T cells are scarce and specific CAR T cell-derived biomarkers are lacking.METHODSCAR+ extracellular vesicle (CAR+EV) release was assessed in human CD19.CAR T cells cocultured with CD19+ target cells. A prospective cohort of 100 patients with B cell lymphoma infused with approved CD19.CAR T cell products was assessed for plasma CAR+EVs as biomarkers of in vivo CD19.CAR T cell activation. Human induced pluripotent stem cell-derived (iPSC-derived) neural cells were used as a model for CAR+EV-induced neurotoxicity.RESULTSIn vitro release of CAR+EVs occurs within 1 hour after target engagement. Plasma CAR+EVs are detectable 1 hour after infusion. A concentration greater than 132.8 CAR+EVs/µL at hour +1 or greater than 224.5 CAR+EVs/µL at day +1 predicted ICANS in advance of 4 days, with a sensitivity and a specificity outperforming other ICANS predictors. ENO2+ nanoparticles were released by iPSC-derived neural cells upon CAR+EV exposure and were increased in plasma of patients with ICANS.CONCLUSIONPlasma CAR+EVs are an immediate signal of CD19.CAR T cell activation, are suitable predictors of neurotoxicity, and may be involved in ICANS pathogenesis.TRIAL REGISTRATIONNCT04892433, NCT05807789.FUNDINGLife Science Hub-Advanced Therapies (financed by Health Ministry as part of the National Plan for Complementary Investments to the National Recovery and Resilience Plan [NRRP]: E.3 Innovative health ecosystem for APC fees and immunomonitoring).


Sujet(s)
Antigènes CD19 , Vésicules extracellulaires , Immunothérapie adoptive , Lymphome B , Humains , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Antigènes CD19/immunologie , Lymphome B/immunologie , Lymphome B/thérapie , Lymphome B/sang , Adulte , Sujet âgé , Récepteurs chimériques pour l'antigène/immunologie , Études prospectives
13.
J Agric Food Chem ; 72(26): 14713-14726, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38885172

RÉSUMÉ

Extracellular vesicles released by probiotics have been demonstrated to effectively alleviate intestinal inflammation, yet the precise underlying mechanisms remain unclear. In this research, for the first time, Lactobacillus plantarum UJS001 (LP-UJS) was isolated from fermented sauerkraut in Zhenjiang, China. Thereafter, the therapeutic effect of LP-UJS-derived extracellular vesicles (LP-UJS-EVs) on dextran sulfate sodium-induced ulcerative colitis (UC) in mice was analyzed to elucidate the immune mechanisms. According to our findings, LP-UJS-EVs played a pivotal role in restoring the intestinal barrier and alleviating intestinal inflammation. Notably, LP-UJS-EVs induced M2 polarization of macrophages, promoted the release of IL-10 and TGF-ß, inhibited the release of histamine, IL-6, and TNF-α, and exerted regulatory effects on intestinal microflora, as evidenced by the reduced abundances of Coprococcus, Parabacteroides, Staphylococcus, and Allobaculum, alongside the enhanced abundance of Prevotella. Furthermore, both LP-UJS and LP-UJS-EVs affected the lysine degradation pathway and significantly increased the abundance of related metabolites, especially oxoadipic acid. In summary, our results underscore the substantial therapeutic potential of LP-UJS and its secreted EVs in the treatment of UC.


Sujet(s)
Rectocolite hémorragique , Vésicules extracellulaires , Microbiome gastro-intestinal , Lactobacillus plantarum , Macrophages , Souris de lignée C57BL , Probiotiques , Rectocolite hémorragique/microbiologie , Rectocolite hémorragique/thérapie , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/immunologie , Lactobacillus plantarum/métabolisme , Animaux , Souris , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/composition chimique , Macrophages/immunologie , Macrophages/métabolisme , Probiotiques/pharmacologie , Probiotiques/administration et posologie , Mâle , Humains , Homéostasie , Interleukine-10/métabolisme , Interleukine-10/génétique , Interleukine-10/immunologie , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/immunologie , Facteur de nécrose tumorale alpha/génétique , Sulfate dextran/effets indésirables , Facteur de croissance transformant bêta/métabolisme
14.
Front Cell Infect Microbiol ; 14: 1411196, 2024.
Article de Anglais | MEDLINE | ID: mdl-38873097

RÉSUMÉ

Bacterial extracellular vesicles (EVs) are crucial mediators of information transfer between bacteria and host cells. Macrophages, as key effector cells in the innate immune system, have garnered widespread attention for their interactions with bacterial EVs. Increasing evidence indicates that bacterial EVs can be internalized by macrophages through multiple pathways, thereby influencing their immune functions. These functions include inflammatory responses, antimicrobial activity, antigen presentation, and programmed cell death. Therefore, this review summarizes current research on the interactions between bacterial EVs and macrophages. This will aid in the deeper understanding of immune modulation mediated by pathogenic microorganisms and provide a basis for developing novel antibacterial therapeutic strategies.


Sujet(s)
Bactéries , Vésicules extracellulaires , Immunité innée , Macrophages , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/métabolisme , Macrophages/immunologie , Macrophages/microbiologie , Humains , Animaux , Bactéries/immunologie , Interactions hôte-pathogène/immunologie
15.
Front Cell Infect Microbiol ; 14: 1398077, 2024.
Article de Anglais | MEDLINE | ID: mdl-38836056

RÉSUMÉ

Mycobacterium tuberculosis (M.tb), the causative agent of Tuberculosis, is an intracellular bacterium well known for its ability to subvert host energy and metabolic pathways to maintain its intracellular survival. For this purpose, the bacteria utilize various mechanisms of which extracellular vehicles (EVs) related mechanisms attracted more attention. EVs are nanosized particles that are released by almost all cell types containing active biomolecules from the cell of origin and can target bioactive pathways in the recipient cells upon uptake. It is hypothesized that M.tb dictates the processes of host EV biogenesis pathways, selectively incorporating its molecules into the host EV to direct immune responses in its favor. During infection with Mtb, both mycobacteria and host cells release EVs. The composition of these EVs varies over time, influenced by the physiological and nutritional state of the host environment. Additionally, different EV populations contribute differently to the pathogenesis of disease at various stages of illness participating in a complex interplay between host cells and pathogens. These interactions ultimately influence immune responses and disease outcomes. However, the precise mechanisms and roles of EVs in pathogenicity and disease outcomes remain to be fully elucidated. In this review, we explored the properties and function of EVs in the context of M.tb infection within the host microenvironment and discussed their capacity as a novel therapeutic strategy to combat tuberculosis.


Sujet(s)
Vésicules extracellulaires , Interactions hôte-pathogène , Mycobacterium tuberculosis , Tuberculose , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Mycobacterium tuberculosis/immunologie , Humains , Tuberculose/immunologie , Tuberculose/microbiologie , Tuberculose/métabolisme , Interactions hôte-pathogène/immunologie , Animaux
16.
Front Immunol ; 15: 1418061, 2024.
Article de Anglais | MEDLINE | ID: mdl-38903499

RÉSUMÉ

Extracellular vesicles (EVs), characterized by low immunogenicity, high biocompatibility and targeting specificity along with excellent blood-brain barrier permeability, are increasingly recognized as promising drug delivery vehicles for treating a variety of diseases, such as cancer, inflammation and viral infection. However, recent findings demonstrate that the intracellular delivery efficiency of EVs fall short of expectations due to phagocytic clearance mediated by the host mononuclear phagocyte system through Fcγ receptors, complement receptors as well as non-opsonic phagocytic receptors. In this text, we investigate a range of bacterial virulence proteins that antagonize host phagocytic machinery, aiming to explore their potential in engineering EVs to counteract phagocytosis. Special emphasis is placed on IdeS secreted by Group A Streptococcus and ImpA secreted by Pseudomonas aeruginosa, as they not only counteract phagocytosis but also bind to highly upregulated surface biomarkers αVß3 on cancer cells or cleave the tumor growth and metastasis-promoting factor CD44, respectively. This suggests that bacterial anti-phagocytic proteins, after decorated onto EVs using pre-loading or post-loading strategies, can not only improve EV-based drug delivery efficiency by evading host phagocytosis and thus achieve better therapeutic outcomes but also further enable an innovative synergistic EV-based cancer therapy approach by integrating both phagocytosis antagonism and cancer targeting or deactivation.


Sujet(s)
Vésicules extracellulaires , Phagocytose , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/métabolisme , Phagocytose/immunologie , Humains , Animaux , Protéines bactériennes/métabolisme , Protéines bactériennes/immunologie , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/métabolisme , Intégrine alphaVbêta3/métabolisme , Intégrine alphaVbêta3/immunologie , Antigènes CD44/métabolisme , Antigènes CD44/immunologie , Pseudomonas aeruginosa/immunologie
17.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38892397

RÉSUMÉ

Pathogenic bacteria have several mechanisms to evade the host's immune response and achieve an efficient infection. Bacterial extracellular vesicles (EVs) are a relevant cellular communication mechanism, since they can interact with other bacterial cells and with host cells. In this review, we focus on the EVs produced by some World Health Organization (WHO) priority Gram-negative and Gram-positive pathogenic bacteria; by spore-producing bacteria; by Mycobacterium tuberculosis (a bacteria with a complex cell wall); and by Treponema pallidum (a bacteria without lipopolysaccharide). We describe the classification and the general properties of bacterial EVs, their role during bacterial infections and their effects on the host immune response. Bacterial EVs contain pathogen-associated molecular patterns that activate innate immune receptors, which leads to cytokine production and inflammation, but they also contain antigens that induce the activation of B and T cell responses. Understanding the many effects of bacterial EVs on the host's immune response can yield new insights on the pathogenesis of clinically important infections, but it can also lead to the development of EV-based diagnostic and therapeutic strategies. In addition, since EVs are efficient activators of both the innate and the adaptive immune responses, they constitute a promising platform for vaccine development.


Sujet(s)
Vésicules extracellulaires , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/métabolisme , Humains , Animaux , Immunité innée , Interactions hôte-pathogène/immunologie , Infections bactériennes/immunologie , Infections bactériennes/microbiologie , Bactéries/immunologie
18.
Front Immunol ; 15: 1360618, 2024.
Article de Anglais | MEDLINE | ID: mdl-38827737

RÉSUMÉ

Psoriasis is a chronic inflammatory disease affecting skin and joints characterized by a chronically altered immune and inflammatory response. Several factors occur from the onset to the development of this disease due to different types of cells spatially and temporally localized in the affected area, such as, keratinocytes, macrophages, neutrophils and T helper lymphocytes. This scenario leads to the chronic release of high levels of inflammatory mediators (i.e., IL-17, IL-23, IL-22, TNF-α, S100 proteins, Defensins) and lastly parakeratosis and thickening of the stratum spinosum. Extracellular vesicles (EVs) are small double membraned biological nanoparticles that are secreted by all cell types and classified, based on dimension and biogenesis, into exosomes, microvesicles and apoptotic bodies. Their role as vessels for long range molecular signals renders them key elements in the pathogenesis of psoriasis, as well as innovative platforms for potential biomarker discovery and delivery of fine-tuned anti-inflammatory therapies. In this review, the role of EVs in the pathogenesis of psoriasis and the modulation of cellular microenvironment has been summarized. The biotechnological implementation of EVs for therapy and research for new biomarkers has been also discussed.


Sujet(s)
Marqueurs biologiques , Vésicules extracellulaires , Psoriasis , Humains , Psoriasis/immunologie , Psoriasis/métabolisme , Psoriasis/étiologie , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Animaux , Peau/anatomopathologie , Peau/immunologie , Peau/métabolisme , Microenvironnement cellulaire/immunologie
19.
J Extracell Vesicles ; 13(6): e12446, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38844736

RÉSUMÉ

Dendritic cells (DCs) are essential orchestrators of immune responses and represent potential targets for immunomodulation in autoimmune diseases. Human amniotic fluid secretome is abundant in immunoregulatory factors, with extracellular vesicles (EVs) being a significant component. However, the impact of these EVs on dendritic cells subsets remain unexplored. In this study, we investigated the interaction between highly purified dendritic cell subsets and EVs derived from amniotic fluid stem cell lines (HAFSC-EVs). Our results suggest that HAFSC-EVs are preferentially taken up by conventional dendritic cell type 2 (cDC2) through CD29 receptor-mediated internalization, resulting in a tolerogenic DC phenotype characterized by reduced expression and production of pro-inflammatory mediators. Furthermore, treatment of cDC2 cells with HAFSC-EVs in coculture systems resulted in a higher proportion of T cells expressing the regulatory T cell marker Foxp3 compared to vehicle-treated control cells. Moreover, transfer of HAFSC-EV-treated cDC2s into an EAE mouse model resulted in the suppression of autoimmune responses and clinical improvement. These results suggest that HAFSC-EVs may serve as a promising tool for reprogramming inflammatory cDC2s towards a tolerogenic phenotype and for controlling autoimmune responses in the central nervous system, representing a potential platform for the study of the effects of EVs in DC subsets.


Sujet(s)
Liquide amniotique , Cellules dendritiques , Modèles animaux de maladie humaine , Encéphalomyélite auto-immune expérimentale , Vésicules extracellulaires , Sclérose en plaques , Animaux , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Souris , Liquide amniotique/cytologie , Liquide amniotique/métabolisme , Encéphalomyélite auto-immune expérimentale/immunologie , Encéphalomyélite auto-immune expérimentale/thérapie , Encéphalomyélite auto-immune expérimentale/métabolisme , Humains , Sclérose en plaques/thérapie , Sclérose en plaques/immunologie , Sclérose en plaques/métabolisme , Femelle , Cellules souches/métabolisme , Cellules souches/cytologie , Souris de lignée C57BL
20.
Int Immunopharmacol ; 137: 112442, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38889508

RÉSUMÉ

Liver disease ranks as the eleventh leading cause of mortality, leading to approximately 2 million deaths annually worldwide. Neutrophils are a type of immune cell that are abundant in peripheral blood and play a vital role in innate immunity by quickly reaching the site of liver injury. They exert their influence on liver diseases through autocrine, paracrine, and immunomodulatory mechanisms. Extracellular vesicles, phospholipid bilayer vesicles, transport a variety of substances, such as proteins, nucleic acids, lipids, and pathogenic factors, for intercellular communication. They regulate cell communication and perform their functions by delivering biological information. Current research has revealed the involvement of the interaction between neutrophils and extracellular vesicles in the pathogenesis of liver disease. Moreover, more research has focused on targeting neutrophils as a therapeutic strategy to attenuate disease progression. Therefore, this article summarizes the roles of neutrophils, extracellular vesicles, and their interactions in noncancerous liver diseases.


Sujet(s)
Communication cellulaire , Vésicules extracellulaires , Maladies du foie , Granulocytes neutrophiles , Humains , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Maladies du foie/immunologie , Maladies du foie/métabolisme , Maladies du foie/anatomopathologie , Animaux , Communication cellulaire/immunologie , Immunité innée , Foie/métabolisme , Foie/anatomopathologie , Foie/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE