Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 788
Filtrer
1.
Virol J ; 21(1): 177, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107806

RÉSUMÉ

BACKGROUND: Reticuloendotheliosis virus (REV), a member of the family Retroviridae, is a hot area of research, and a previous study showed that exosomes purified from REV-positive semen were not blocked by REV-specific neutralizing antibodies and established productive infections. METHODS: To further verify the infectivity of exosomes from REV-infected cells, we isolated and purified exosomes from REV-infected DF-1 cells and identified them using Western blot and a transmission electron microscope. We then inoculated 7-day-old embryonated eggs, 1-day-old chicks and 23-week-old hens with and without antibody treatment. REV was administered simultaneously as a control. RESULTS: In the absence of antibodies, the results indicated that REV-exosomes and REV could infect chicks, resulting in viremia and viral shedding, compared with the infection caused by REV, REV-exosomes reduced the hatching rate and increased mortality after hatching, causing severe growth inhibition and immune organ damage in 1-day-old chicks; both REV and REV-exosomes also could infect hens, however, lead to transient infection. In the presence of antibodies, REV-exosomes were not blocked by REV-specific neutralizing antibodies and infected 7-day-old embryonated eggs. However, REV could not infect 1-day-old chicks and 23-week-old hens. CONCLUSION: In this study, we compared the infectious ability of REV-exosomes and REV, REV-exosomes could escape from REV-specific neutralizing antibodies in embryonated eggs, providing new insights into the immune escape mechanism of REV.


Sujet(s)
Anticorps antiviraux , Poulets , Exosomes , Maladies de la volaille , Virus de la réticuloendothéliose , Infections à Retroviridae , Excrétion virale , Animaux , Exosomes/virologie , Exosomes/immunologie , Anticorps antiviraux/immunologie , Poulets/virologie , Virus de la réticuloendothéliose/immunologie , Maladies de la volaille/virologie , Maladies de la volaille/transmission , Maladies de la volaille/immunologie , Infections à Retroviridae/virologie , Infections à Retroviridae/transmission , Infections à Retroviridae/immunologie , Infections à Retroviridae/médecine vétérinaire , Anticorps neutralisants/immunologie , Lignée cellulaire , Virémie/virologie , Femelle
2.
Cancer Immunol Immunother ; 73(10): 196, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39105814

RÉSUMÉ

Anti-cluster of differentiation (CD) 3 × α programmed death-ligand 1 (PD-L1) bispecific T-cell engager (BsTE)-bound T-cells (BsTE:T) are a promising new cancer treatment agent. However, the mechanisms of action of bispecific antibody-armed activated T-cells are poorly understood. Therefore, this study aimed to investigate the anti-tumor mechanism and efficacy of BsTE:T. The BsTE:T migration was assessed in vivo and in vitro using syngeneic and xenogeneic tumor models, flow cytometry, immunofluorescence staining, transwell migration assays, microfluidic chips, Exo View R100, western blotting, and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 technology. In murine B16 melanoma, MC38 colon cancer, and human multiple myeloma cells, BsTE:T exhibited superior tumor elimination relative to that of T-cells or BsTE alone. Moreover, BsTE:T migration into tumors was significantly enhanced owing to the presence of PD-L1 in tumor cells and secretion of PD-L1-containing exosomes. Furthermore, increased infiltration of CD44highCD62Llow effector memory CD8+ T-cells into tumors was closely associated with the anti-tumor effect of BsTE:T. Therefore, BsTE:T is an innovative potential anti-tumor therapy, and exosomal PD-L1 plays a crucial role both in vitro and in vivo in the anti-tumor activity of BsTE:T.


Sujet(s)
Anticorps bispécifiques , Antigène CD274 , Exosomes , Animaux , Anticorps bispécifiques/pharmacologie , Anticorps bispécifiques/immunologie , Exosomes/métabolisme , Exosomes/immunologie , Souris , Antigène CD274/métabolisme , Antigène CD274/antagonistes et inhibiteurs , Humains , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Souris de lignée C57BL , Mélanome expérimental/immunologie , Mélanome expérimental/thérapie , Antigènes CD3/immunologie , Antigènes CD3/métabolisme , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Lignée cellulaire tumorale , Femelle , Mouvement cellulaire , Tests d'activité antitumorale sur modèle de xénogreffe
3.
Int Immunopharmacol ; 139: 112656, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39043104

RÉSUMÉ

Today, cancer treatment is one of the main challenges for researchers. The main cause of tumor cell formation is mutations that lead to uncontrolled proliferation and inhibition of apoptosis in malignant cells. Tumor cells also create a microenvironment that can suppress the immune system cells' responses through various methods, including producing soluble factors and cell-to-cell communication. After being produced from tumor cells, exosomes can also affect the functions of other cells in this microenvironment. Various studies have shown that exosomes from different sources, including tumor cells and immune cells, can be used to treat cancers due to their characteristics. Since tumor cells are rich sources of various types of tumor peptides, they can induce anti-tumor responses. Immune cells also produce exosomes that mimic the functions of their cells of origin, such that exosomes derived from NK cells and CTLs can directly lead to their apoptosis after merging with tumor cells. However, many researchers have pointed out that naïve exosomes have a limited therapeutic function, and their therapeutic potential can be increased by manipulating and engineering them. There are various methods to modify exosomes and improve their therapeutic potential. In general, these methods are divided into two parts, which include changing the cell of origin of the exosome and encapsulating the exosome to carry different drugs. In this review, we will discuss the studies on the therapeutic use of naive and engineered exosomes and provide an update on new studies in this field.


Sujet(s)
Exosomes , Immunothérapie , Tumeurs , Microenvironnement tumoral , Exosomes/immunologie , Exosomes/métabolisme , Humains , Tumeurs/thérapie , Tumeurs/immunologie , Immunothérapie/méthodes , Animaux , Microenvironnement tumoral/immunologie , Cellules tueuses naturelles/immunologie
4.
Front Immunol ; 15: 1436151, 2024.
Article de Anglais | MEDLINE | ID: mdl-39076982

RÉSUMÉ

Introduction: Exosomes produced by the protozoan parasite Leishmania (LeishEXO) are well-established drivers of virulence, though mechanisms underlying their exacerbation of experimental leishmaniasis remain elusive. Expression of Annexin A1 (ANXA1), a protein implicated in exosome-mediated pathologies and viral internalization, has been shown to correlate with cutaneous leishmaniasis severity. Given ANXA1's regulation of myeloid cells - the canonical hosts for Leishmania - we studied the potential role of ANXA1 and its receptors FPR1/2 in exerting LeishEXO's effects. Methods: Murine and in vitro ANXA1-/- models were used to study the generation of protective TH1 responses during experimental L. major infection with and without LeishEXO. Recruitment of inflammatory cells was assessed using a peritoneal cell recruitment assay and immunophenotyping, and production of inflammatory mediators was measured using a cytokine and chemokine array. Treatment of experimental models with FPR2 antagonist WRW4 and FPR1/2 agonist WKYMVm was used to delineate the role of the FPR/ANXA1 axis in LeishEXO-mediated hyperpathogenesis. Results: We established that ANXA1 deficiency prohibits LeishEXO-mediated pathogenesis and myeloid cell infection, with minimal alterations to adaptive and innate immune phenotypes. FPR2 blockade with WRW4 similarly inhibited leishmanial hyperpathogenesis, while direct activation of FPRs with WKYMVm enhanced infection and recapitulated the LeishEXO-mediated phenotype. This research describes LeishEXO's utilization of the ANXA1/FPR axis to facilitate parasitic internalization and pathogenesis, which may be leveraged in the development of therapeutics for leishmaniasis.


Sujet(s)
Annexine A1 , Exosomes , Leishmania major , Leishmaniose cutanée , Souris knockout , Récepteurs aux peptides formylés , Annexine A1/métabolisme , Annexine A1/génétique , Animaux , Exosomes/métabolisme , Exosomes/immunologie , Leishmania major/immunologie , Leishmaniose cutanée/immunologie , Leishmaniose cutanée/parasitologie , Leishmaniose cutanée/métabolisme , Souris , Récepteurs aux peptides formylés/métabolisme , Souris de lignée C57BL , Modèles animaux de maladie humaine , Peau/parasitologie , Peau/immunologie , Peau/anatomopathologie , Peau/métabolisme , Lymphocytes auxiliaires Th1/immunologie , Femelle
5.
Aging (Albany NY) ; 16(14): 11373-11384, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39028365

RÉSUMÉ

OBJECTIVE: To investigate the expression of programmed death ligand-1 (PD-L1) in circulating exosomes, and to define the role of exosomal PD-L1 in promoting immune escape mechanism during chronic hepatitis B infection (CHB) and related liver diseases. METHODS: The levels of PD-L1 expressed in exosomes were detected by ELISA. CD8+T cells were sorted and cytotoxicity test was assessed by flow cytometry. PD-L1 protein expression in hepatocellular carcinoma (HCC) and normal adjacent tissues were detected by immunohistochemistry. RESULTS: Circulating exosomal PD-L1 levels were significantly higher in patients with CHB and HCC than in healthy controls (F =7.46, P=0.001). Levels of CD107a on CD8+T cells in patients with CHB receiving PD-L1 blocking antibody were significantly lower than in patients receiving isotype blocking antibody (t = 4.96, P < 0.01). Levels of TNF-α in cell culture supernatants of the PD-L1 blocking antibody group were significantly higher than in the isotype blocking antibody group (t =5.92, P < 0.01). Compared with patients receiving isotype blocking antibody, levels of CD107a on CD8+T cells significantly increased in patients with HCC receiving anti-PD-L1 antibody (t = 3.51, P<0.05). Compared with adjacent tissues, the levels of PD-L1 protein expression in HCC tissues were slightly higher; however, no significant difference between the two groups was observed. CONCLUSIONS: PD-L1 blockade in exosomes might promote the cytotoxic function of CD8+T cells and inhibit immune evasion during progression of HCC. Blocking PD-L1 in exosomes reduced the cytotoxic function of CD8+T cells in patients with CHB while enhancing the production of proinflammatory cytokines.


Sujet(s)
Antigène CD274 , Lymphocytes T CD8+ , Carcinome hépatocellulaire , Exosomes , Hépatite B chronique , Tumeurs du foie , Échappement de la tumeur à la surveillance immunitaire , Humains , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/métabolisme , Antigène CD274/métabolisme , Tumeurs du foie/immunologie , Tumeurs du foie/métabolisme , Hépatite B chronique/immunologie , Hépatite B chronique/sang , Exosomes/métabolisme , Exosomes/immunologie , Mâle , Femelle , Adulte d'âge moyen , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Échappement de la tumeur à la surveillance immunitaire/immunologie , Adulte
6.
J Extracell Vesicles ; 13(7): e12490, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39051742

RÉSUMÉ

Extracellular vesicles (EVs) are emerging as promising carriers for the delivery of therapeutic biologics. Genetic engineering represents a robust strategy for loading proteins of interest into EVs. Identification of EV-enriched proteins facilitates protein cargo loading efficiency. Many EV-enriched proteins are sorted into EVs via an endosomal sorting complex required for transport (ESCRT)-dependent pathway. In parallel, viruses hijack this EV biosynthesis machinery via conserved late domain motifs to promote egress from host cells. Inspired by the similarity of biogenesis between EVs and viruses, we developed a synthetic, Late domain-based EV scaffold protein that enables the display of a set of single chain variable fragments (scFvs) on the EV surface. We named this scaffold the Late domain-based exosomal antibody surface display platform (LEAP). We applied the LEAP scaffold to reprogramme HEK293T cell-derived EVs to elicit T-cell anti-tumor immunity by simultaneously displaying αPD-L1 and αCD3 scFvs on the EV surface (denoted as αPD-L1×αCD3 bispecific T-cell engaging exosomes, BiTExos). We demonstrated that αPD-L1×αCD3 BiTExos actively redirected T cells to bind to PD-L1+ tumor cells, promoting T-cell activation, proliferation and tumoricidal cytokine production. Furthermore, the αPD-L1×αCD3 BiTExos promoted T-cell infiltration into the tumor microenvironment to mitigate the tumor burden in vivo. Our study suggested that the LEAP scaffold may serve as a platform for EV surface display and could be applied for a broad range of EV-based biomedical applications.


Sujet(s)
Antigène CD274 , Antigènes CD3 , Vésicules extracellulaires , Anticorps à chaîne unique , Lymphocytes T , Humains , Vésicules extracellulaires/immunologie , Vésicules extracellulaires/métabolisme , Antigène CD274/métabolisme , Antigène CD274/immunologie , Animaux , Antigènes CD3/immunologie , Antigènes CD3/métabolisme , Cellules HEK293 , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Souris , Anticorps à chaîne unique/immunologie , Exosomes/métabolisme , Exosomes/immunologie , Tumeurs/immunologie , Tumeurs/thérapie , Activation des lymphocytes/immunologie
7.
Front Immunol ; 15: 1429442, 2024.
Article de Anglais | MEDLINE | ID: mdl-39040099

RÉSUMÉ

Introduction: Allergic rhinitis (AR) is an upper airway inflammatory disease of the nasal mucosa. Conventional treatments such as symptomatic pharmacotherapy and allergen-specific immunotherapy have considerable limitations and drawbacks. As an emerging therapy with regenerative potential and immunomodulatory effect, mesenchymal stem cell-derived exosomes (MSC-Exos) have recently been trialed for the treatment of various inflammatory and autoimmune diseases. Methods: In order to achieve sustained and protected release of MSC-Exos for intranasal administration, we fabricated Poly(lactic-co-glycolic acid) (PLGA) micro and nanoparticles-encapsulated MSC-Exos (PLGA-Exos) using mechanical double emulsion for local treatment of AR. Preclinical in vivo imaging, ELISA, qPCR, flow cytometry, immunohistochemical staining, and multiomics sequencing were used for phenotypic and mechanistic evaluation of the therapeutic effect of PLGA-Exos in vitro and in vivo. Results: The results showed that our PLGA platform could efficiently encapsulate and release the exosomes in a sustained manner. At protein level, PLGA-Exos treatment upregulated IL-2, IL-10 and IFN-γ, and downregulated IL-4, IL-17 and antigen-specific IgE in ovalbumin (OVA)-induced AR mice. At cellular level, exosomes treatment reduced Th2 cells, increased Tregs, and reestablished Th1/Th2 balance. At tissue level, PLGA-Exos significantly attenuated the infiltration of immune cells (e.g., eosinophils and goblet cells) in nasal mucosa. Finally, multiomics analysis discovered several signaling cascades, e.g., peroxisome proliferator-activated receptor (PPAR) pathway and glycolysis pathway, that might mechanistically support the immunomodulatory effect of PLGA-Exos. Discussion: For the first time, we present a biomaterial-facilitated local delivery system for stem cell-derived exosomes as a novel and promising strategy for AR treatment.


Sujet(s)
Exosomes , Cellules souches mésenchymateuses , Copolymère d'acide poly(lactique-co-glycolique) , Rhinite allergique , Exosomes/immunologie , Exosomes/métabolisme , Animaux , Rhinite allergique/thérapie , Rhinite allergique/immunologie , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Cellules souches mésenchymateuses/immunologie , Cellules souches mésenchymateuses/métabolisme , Souris , Souris de lignée BALB C , Immunomodulation , Cytokines/métabolisme , Modèles animaux de maladie humaine , Femelle , Humains , Muqueuse nasale/immunologie , Muqueuse nasale/métabolisme , Administration par voie nasale
8.
Front Immunol ; 15: 1424081, 2024.
Article de Anglais | MEDLINE | ID: mdl-39040108

RÉSUMÉ

Exosomes are found in various tissues of the body and carry abundant contents including nucleic acids, proteins, and metabolites, which continuously flow between cells of various tissues and mediate important intercellular communication. In addition, exosomes from different cellular sources possess different physiopathological immunomodulatory effects, which are closely related to the immune regeneration of normal or abnormal organs and tissues. Here, we focus on the mechanistic interactions between exosomes and the human immune system, introduce the immuno-regenerative therapeutic potential of exosomes in common clinical immune-related diseases, such as infectious diseases, autoimmune diseases, and tumors, and reveal the safety and efficacy of exosomes as a novel cell-free immune regenerative therapy.


Sujet(s)
Exosomes , Immunothérapie , Exosomes/immunologie , Exosomes/métabolisme , Humains , Immunothérapie/méthodes , Animaux , Tumeurs/thérapie , Tumeurs/immunologie , Communication cellulaire/immunologie , Immunomodulation , Maladies auto-immunes/thérapie , Maladies auto-immunes/immunologie
9.
J Immunol ; 213(5): 567-576, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38984872

RÉSUMÉ

Endometriosis, affecting 10% of women, is defined as implantation, survival, and growth of endometrium-like/endometriotic tissue outside the uterine cavity, causing inflammation, infertility, pain, and susceptibility to ovarian cancer. Despite extensive studies, its etiology and pathogenesis are poorly understood and largely unknown. The prevailing view is that the immune system of endometriosis patients fails to clear ectopically disseminated endometrium from retrograde menstruation. Exosomes are small extracellular vesicles that exhibit immunomodulatory properties. We studied the role of endometriotic tissue-secreted exosomes in the pathophysiology of endometriosis. Two exosome-mediated mechanisms known to impair the immune response were investigated: 1) downregulation of NKG2D-mediated cytotoxicity and 2) FasL- and TRAIL-induced apoptosis of activated immune cells. We showed that secreted endometriotic exosomes isolated from supernatants of short-term explant cultures carry the NKG2D ligands MICA/B and ULBP1-3 and the proapoptotic molecules FasL and TRAIL on their surface, i.e., signature molecules of exosome-mediated immune suppression. Acting as decoys, these exosomes downregulate the NKG2D receptor, impair NKG2D-mediated cytotoxicity, and induce apoptosis of activated PBMCs and Jurkat cells through the FasL- and TRAIL pathway. The secreted endometriotic exosomes create an immunosuppressive gradient at the ectopic site, forming a "protective shield" around the endometriotic lesions. This gradient guards the endometriotic lesions against clearance by a cytotoxic attack and creates immunologic privilege by induction of apoptosis in activated immune cells. Taken together, our results provide a plausible, exosome-based mechanistic explanation for the immune dysfunction and the compromised immune surveillance in endometriosis and contribute novel insights into the pathogenesis of this enigmatic disease.


Sujet(s)
Apoptose , Endométriose , Endomètre , Exosomes , Sous-famille K des récepteurs de cellules NK de type lectine , Ligand TRAIL , Humains , Endométriose/immunologie , Endométriose/métabolisme , Endométriose/anatomopathologie , Femelle , Exosomes/métabolisme , Exosomes/immunologie , Sous-famille K des récepteurs de cellules NK de type lectine/métabolisme , Apoptose/immunologie , Endomètre/immunologie , Endomètre/anatomopathologie , Endomètre/métabolisme , Ligand TRAIL/métabolisme , Ligand TRAIL/immunologie , Régulation négative/immunologie , Ligand de Fas/métabolisme , Ligand de Fas/immunologie , Cytotoxicité immunologique , Adulte , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes d'histocompatibilité de classe I/immunologie
10.
Eur J Pharmacol ; 979: 176831, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39047964

RÉSUMÉ

Programmed cell death ligand 1 (PD-L1) interacts with programmed cell death 1 (PD-1), leading to T cell exhaustion and promoting tumor cell survival, ultimately mediating immunosuppression. While FDA-approved monoclonal antibodies targeting the PD-1/PD-L1 interaction have shown success in cancer treatment, some patients experience limited and short-lived therapeutic outcomes. Recent studies have identified PD-L1 expression not only on tumor cell surfaces but also on exosomes, with secretion pathways including both conventional and unconventional endocytosis routes, presenting a unique therapeutic opportunity. Emerging evidence suggests that exosomal PD-L1 contributes to systemic immunosuppression, potentially counteracting the effects of anti-PD-1 checkpoint therapies. However, the significance of exosomal PD-L1 in clinical cancer patients unresponsive to anti-PD-1/PD-L1 immunotherapy, as well as the factors regulating its generation, remain unclear. Moreover, the mechanisms underlying PD-L1 expression on exosomes and its regulation in cancer are yet to be fully elucidated. This review primarily focuses on the mechanisms modulating exosomal PD-L1 generation in cancer, while also outlining its involvement in immunosuppression, tumor proliferation, and response to cancer immunotherapy. Additionally, we explore the potential of exosomal PD-L1 as a cancer biomarker and therapeutic target, aiming to provide a comprehensive overview of this emerging field and its implications for cancer treatment and diagnosis.


Sujet(s)
Antigène CD274 , Exosomes , Tumeurs , Humains , Exosomes/métabolisme , Exosomes/immunologie , Antigène CD274/métabolisme , Tumeurs/immunologie , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Animaux , Immunothérapie , Marqueurs biologiques tumoraux/métabolisme , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie
11.
Eur J Pharmacol ; 979: 176862, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39068974

RÉSUMÉ

We and other groups have documented that bone marrow-mesenchymal stem cells (BM-MSCs) from Systemic lupus erythematosus (SLE) patients demonstrated signs of senescence, including reduced ability of regulating Treg. Treg cell defects or Treg cell deficiency are regarded as significant factors in the progression of SLE. Exosomes, nanoscale vesicles, abound in molecular and genetic contents, play a critical role in intercellular communications. The purpose of this research is to investigate the mechanism of MSCs-exosomes regulating Tregs cells in SLE, further elucidate the mechanism of immune dysregulation of aging BM-MSCs, and provide theoretical basis and data support for new targets of SLE treatment. In the study, BM-MSCs and exosomes were isolated successfully. Exosomes could be up-taken by naïve CD4+T cells. MSCs-exosomes attenuated SLE clinical manifestation in vivo, but MSCs-exosomes from SLE patients were ineffective. MSCs-exosomes from SLE patients dysregulated Treg cells differentiation in vivo and in vitro. Exosomal miR-20a-5p contributed to the effect of MSCs-exosomes regulating Treg cells. Up-regulating the expression of miR-20a-5p in SLE MSCs-exosomes can restore their ability to promote Treg differentiation and treatment effect. This study further elucidated the role of in the immunomodulatory mechanism of BM-MSCs-exosomes and provided new ideas for the non-cellular autologous transplantation therapy of SLE.


Sujet(s)
Exosomes , Lupus érythémateux disséminé , Cellules souches mésenchymateuses , microARN , Lymphocytes T régulateurs , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/génétique , microARN/génétique , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/immunologie , Exosomes/métabolisme , Exosomes/génétique , Exosomes/immunologie , Exosomes/transplantation , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Humains , Animaux , Femelle , Différenciation cellulaire , Souris , Transplantation de cellules souches mésenchymateuses , Adulte , Régulation positive , Mâle
12.
Front Immunol ; 15: 1435426, 2024.
Article de Anglais | MEDLINE | ID: mdl-39007145

RÉSUMÉ

Exosomes are small disk-shaped extracellular vesicles (EVs) that are naturally released into the environment by different types of cells. Exosomes range from 30-150 nm in size and contain complex RNA and proteins. They are widely found in body fluids such as blood, saliva, urine and breast milk and participate in cell communication by functioning as cell messengers. Almost all cell types can transmit information and exchange substances through the production and release of exosomes to regulate proliferation, differentiation, apoptosis, the immune response, inflammation, and other biological functions. Because exosomes exist widely in various body fluids, they are easy to obtain and detect and have the potential for use in disease diagnosis and prognosis detection. Exosomes can be genetically fused with targeted proteins, enhancing their biocompatibility and immunogenicity. Therefore, exosomes are the preferred vector tools for vaccines. In this review, we describe the characteristics of exosomes and discuss their unique and ambiguous functions in the immune microenvironment after infection. In this regard, we explored the ability of exosomes to carry immunogenic virus antigens and to establish adaptive immune responses. Exosomes can provide an interesting platform for antigen presentation and since vaccines are a powerful method for the prevention of infectious diseases, we further review the advantages and disadvantages of the use of exosomes in vaccine preparation. Overall, exosomes are emerging as a promising avenue for vaccine development.


Sujet(s)
Exosomes , Développement de vaccin , Exosomes/immunologie , Exosomes/métabolisme , Humains , Animaux , Vaccins/immunologie , Systèmes de délivrance de médicaments
13.
Front Immunol ; 15: 1401852, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994350

RÉSUMÉ

Exosomes, as a class of small extracellular vesicles closely related to the biological behavior of various types of tumors, are currently attracting research attention in cancer diagnosis and treatment. Regarding cancer diagnosis, the stability of their membrane structure and their wide distribution in body fluids render exosomes promising biomarkers. It is expected that exosome-based liquid biopsy will become an important tool for tumor diagnosis in the future. For cancer treatment, exosomes, as the "golden communicators" between cells, can be designed to deliver different drugs, aiming to achieve low-toxicity and low-immunogenicity targeted delivery. Signaling pathways related to exosome contents can also be used for safer and more effective immunotherapy against tumors. Exosomes are derived from a wide range of sources, and exhibit different biological characteristics as well as clinical application advantages in different cancer therapies. In this review, we analyzed the main sources of exosomes that have great potential and broad prospects in cancer diagnosis and therapy. Moreover, we compared their therapeutic advantages, providing new ideas for the clinical application of exosomes.


Sujet(s)
Marqueurs biologiques tumoraux , Exosomes , Tumeurs , Humains , Exosomes/métabolisme , Exosomes/immunologie , Tumeurs/thérapie , Tumeurs/immunologie , Animaux , Immunothérapie/méthodes , Biopsie liquide/méthodes
14.
Int Immunopharmacol ; 138: 112282, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38936058

RÉSUMÉ

Hypoxia is a hallmark of solid tumors. Cancer-associated fibroblasts (CAFs) are an important component of the tumor microenvironment, and CAF-derived exosomes are involved in cancer genesis and progression. Here, this work investigated the role and mechanism of exosomal circHIF1A derived from hypoxia-induced CAFs in hepatocellular carcinoma (HCC) tumorigenesis. CAFs isolated from fresh HCC tissues were incubated in normoxia or hypoxia condition (N/CAFs or H/CAFs), and then the exosomes from N/CAFs or H/CAFs were isolated for functional analysis. Cell proliferation, migration and invasion were analyzed by cell counting kit-8, colony formation, and transwell assays. Immune evasion was evaluated by measuring the cytotoxicity and viability of CD8+T cells. qRT-PCR and western blotting analyses were used for the level measurement of genes and proteins. The binding between Hu antigen R (HuR) and circHIF1A or Programmed death ligand 1 (PD-L1) was analyzed by RNA immunoprecipitation assay. Functionally, we found that CAFs, especially CAFs under hypoxic stress (H/CAFs), promoted the proliferation, migration, invasion and EMT progression in HCC cells, as well as induced immune escape by suppressing CD8+T cell cytotoxicity and activity in an exosome-dependent manner. H/CAFs-derived exosomes showed highly expressed circHIF1A, and could secrete circHIF1A into HCC cells via exosomes. The oncogenic effects of H/CAFs-secreted exosomes were abolished by circHIF1A knockdown. Mechanistically, circHIF1A interacted with HuR to stabilize PD-L1 expression in HCC cells. Meanwhile, circHIF1A silencing suppressed HCC cell proliferation, mobility and immune escape by regulating PD-L1 expression. In all, exosomal circHIF1A derived from hypoxic-induced CAFs promoted the proliferation, migration, invasion, EMT progression and immune escape in HCC cells by up-regulating PD-L1 expression in a HuR-dependent manner.


Sujet(s)
Antigène CD274 , Fibroblastes associés au cancer , Carcinome hépatocellulaire , Prolifération cellulaire , Exosomes , Tumeurs du foie , Échappement de la tumeur à la surveillance immunitaire , Humains , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/immunologie , Tumeurs du foie/anatomopathologie , Exosomes/métabolisme , Exosomes/immunologie , Fibroblastes associés au cancer/immunologie , Fibroblastes associés au cancer/anatomopathologie , Fibroblastes associés au cancer/métabolisme , Antigène CD274/métabolisme , Antigène CD274/génétique , Lignée cellulaire tumorale , Mouvement cellulaire , Microenvironnement tumoral/immunologie , Lymphocytes T CD8+/immunologie , Animaux
15.
Adv Sci (Weinh) ; 11(31): e2404064, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38887870

RÉSUMÉ

Vitiligo is an autoimmune disease characterized by epidermal melanocyte destruction, with abnormal autoimmune responses and excessive oxidative stress as two cardinal mechanisms. Human umbilical mesenchymal stem cells-derived exosomes (hUMSCs-Exos) are regarded as promising therapeutic choice for autoimmune diseases due to potent immunosuppressive and anti-oxidative properties, which can be potentiated under 3D cell culture condition. Nevertheless, whether exosomes derived from 3D spheroids of hUMSCs (3D-Exos) exhibit considerable therapeutic effect on vitiligo and the underlying mechanism remain elusive. In this study, systemic administration of 3D-Exos showed a remarkable effect in treating mice with vitiligo, as revealed by ameliorated skin depigmentation, less CD8+T cells infiltration, and expanded Treg cells in skin, and 3D-Exos exerted a better effect than 2D-Exos. Mechanistically, 3D-Exos can prominently facilitate the expansion of Treg cells in vitiligo lesion and suppress H2O2-induced melanocytes apoptosis. Forward miRNA profile analysis and molecular experiments have demonstrated that miR-132-3p and miR-125b-5p enriched in 3D-Exos greatly contributed to these biological effects by targeting Sirt1 and Bak1 respectively. In aggregate, 3D-Exos can efficiently ameliorate vitiligo by simultaneously potentiating Treg cells-mediated immunosuppression and suppressing oxidative stress-induced melanocyte damage via the delivery of miR-132-3p and miR-125b-5p. The employment of 3D-Exos will be a promising treament for vitiligo.


Sujet(s)
Modèles animaux de maladie humaine , Exosomes , Mélanocytes , Cellules souches mésenchymateuses , Stress oxydatif , Lymphocytes T régulateurs , Vitiligo , Vitiligo/thérapie , Vitiligo/immunologie , Animaux , Exosomes/métabolisme , Exosomes/immunologie , Souris , Lymphocytes T régulateurs/immunologie , Mélanocytes/immunologie , Humains , Cellules souches mésenchymateuses/immunologie , Cellules souches mésenchymateuses/métabolisme , Immunosuppression thérapeutique/méthodes , microARN/génétique , microARN/métabolisme , Souris de lignée C57BL
16.
Mol Immunol ; 172: 17-22, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38865800

RÉSUMÉ

Acute myocardial infarction (AMI), mainly triggered by vascular occlusion or thrombosis, is the most prevalent cause of morbidity and mortality among all cardiovascular diseases. The devastating consequences of AMI are further aggravated by the intricate cellular processes involved in inflammation. In the past two decades, many studies have reported that regulatory T cells (Tregs), as the main immunoregulatory cells, play a crucial role in AMI progression. This review offers a comprehensive insight into the intricate relationship between Tregs and AMI development. Moreover, it explores emerging therapeutic strategies that focus on Tregs and their exosomes. Furthermore, we underscore the importance of employing noninvasive in vivo imaging techniques to advance the clinical applications of Tregs-based treatments in AMI. Although further research is essential to fully elucidate the molecular mechanisms underlying the effects of Tregs, therapies tailored to these cells hold immense potential for the treatment of patients with AMI.


Sujet(s)
Infarctus du myocarde , Lymphocytes T régulateurs , Humains , Lymphocytes T régulateurs/immunologie , Infarctus du myocarde/immunologie , Infarctus du myocarde/thérapie , Animaux , Exosomes/immunologie
17.
Cancer Lett ; 595: 216989, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-38825162

RÉSUMÉ

Exosomes, a subset of extracellular vesicles, are released by all active cells and play a crucial role in intercellular communications. Exosomes could facilitate the transfer of various biologically active molecules, such as DNA, non-coding RNAs, and proteins, from donor to recipient cells, thereby participating in diverse biological and pathological processes. Besides, exosomes possess unique characteristics, including non-toxicity, low-immunogenicity, and stability within biological systems, rendering them highly advantageous for cancer drug development. Meanwhile, accumulating evidence suggests that exosomes originating from tumor cells and immune cells possess distinct composition profiles that play a direct role in anticancer immunotherapy. Of note, exosomes can transport their contents to specific cells, thereby exerting an impact on the phenotype and immune-regulatory functions of targeted cells. Therapeutic cancer vaccines, an emerging therapeutics of immunotherapy, could enhance antitumor immune responses by delivering a large number of tumor antigens, thereby augmenting the immune response against tumor cells. Therefore, the therapeutic rationale of cancer vaccines and exosome-based immunotherapy are almost similar to some extent, but some challenges have hindered their application in the clinical setting. Here, in this review, we first summarized the biogenesis, structure, compositions, and biological functions of exosomes. Then we described the roles of exosomes in cancer biology, particularly in tumor immunity. We also comprehensively reviewed current exosome-based anticancer vaccine development and we divided them into three types. Finally, we give some insights into clinical translation and clinical trial progress of exosome-based anticancer vaccines for future direction.


Sujet(s)
Vaccins anticancéreux , Exosomes , Immunothérapie , Tumeurs , Humains , Exosomes/immunologie , Exosomes/métabolisme , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Tumeurs/immunologie , Tumeurs/thérapie , Immunothérapie/méthodes , Animaux
18.
Front Immunol ; 15: 1384946, 2024.
Article de Anglais | MEDLINE | ID: mdl-38835784

RÉSUMÉ

Breast cancer has a high incidence and a heightened propensity for metastasis. The absence of precise targets for effective intervention makes it imperative to devise enhanced treatment strategies. Exosomes, characterized by a lipid bilayer and ranging in size from 30 to 150 nm, can be actively released by various cells, including those in tumors. Exosomes derived from distinct subsets of immune cells have been shown to modulate the immune microenvironment within tumors and influence breast cancer progression. In addition, tumor-derived exosomes have been shown to contribute to breast cancer development and progression and may become a new target for breast cancer immunotherapy. Tumor immunotherapy has become an option for managing tumors, and exosomes have become therapeutic vectors that can be used for various pathological conditions. Edited exosomes can be used as nanoscale drug delivery systems for breast cancer therapy, contributing to the remodeling of immunosuppressive tumor microenvironments and influencing the efficacy of immunotherapy. This review discusses the regulatory role of exosomes from different cells in breast cancer and the latest applications of exosomes as nanoscale drug delivery systems and immunotherapeutic agents in breast cancer, showing the development prospects of exosomes in the clinical treatment of breast cancer.


Sujet(s)
Tumeurs du sein , Exosomes , Immunothérapie , Microenvironnement tumoral , Exosomes/immunologie , Exosomes/métabolisme , Humains , Tumeurs du sein/thérapie , Tumeurs du sein/immunologie , Femelle , Immunothérapie/méthodes , Microenvironnement tumoral/immunologie , Animaux , Systèmes de délivrance de médicaments
19.
Front Immunol ; 15: 1400112, 2024.
Article de Anglais | MEDLINE | ID: mdl-38868769

RÉSUMÉ

Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies worldwide and has a poor prognosis. Although immune checkpoint inhibitors have entered a new era of HCC treatment, their response rates are modest, which can be attributed to the immunosuppressive tumor microenvironment within HCC tumors. Accumulating evidence has shown that tumor growth is fueled by cancer stem cells (CSCs), which contribute to therapeutic resistance to the above treatments. Given that CSCs can regulate cellular and physical factors within the tumor niche by secreting various soluble factors in a paracrine manner, there have been increasing efforts toward understanding the roles of CSC-derived secretory factors in creating an immunosuppressive tumor microenvironment. In this review, we provide an update on how these secretory factors, including growth factors, cytokines, chemokines, and exosomes, contribute to the immunosuppressive TME, which leads to immune resistance. In addition, we present current therapeutic strategies targeting CSC-derived secretory factors and describe future perspectives. In summary, a better understanding of CSC biology in the TME provides a rational therapeutic basis for combination therapy with ICIs for effective HCC treatment.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Cellules souches tumorales , Microenvironnement tumoral , Humains , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Microenvironnement tumoral/immunologie , Cellules souches tumorales/immunologie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Tumeurs du foie/immunologie , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Animaux , Exosomes/métabolisme , Exosomes/immunologie , Cytokines/métabolisme , Protéines et peptides de signalisation intercellulaire/métabolisme
20.
Int Immunopharmacol ; 137: 112484, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38885605

RÉSUMÉ

Melanoma is a malignant skin tumor with a high mortality rate. Regulatory T cells (Tregs) are immune cells with immunosuppressive roles, however, the precise mechanisms governing Treg involvement in melanoma remain enigmatic. Experimental findings unveiled different transcription factor switches between normal and tumor T cell, with heightened FOXP3 and BATF in the latter. These factors induced immunosuppressive molecules and Treg maintenance genes, polarizing tumor T cells into Tregs. Spatial transcriptomics illuminated the preferential settlement of Tregs at the melanoma periphery. Within this context, FOXP3 in Tregs facilitated direct enhancement of specific ligand gene expression, fostering communication with neighboring cells. Novel functional molecules bound to FOXP3 or BATF in Tregs, such as SPOCK2, SH2D2A, and ligand molecules ITGB2, LTA, CLEC2C, CLEC2D, were discovered, which had not been previously reported in melanoma Treg studies. Furthermore, we validated our findings in a large number of clinical samples and identified the Melanoma Treg-Specific Regulatory Tag Set (Mel TregS). ELISA analysis showed that the protein levels of Mel TregS in melanoma Tregs were higher than in normal Tregs. We then utilized SERS technology to measure the signal values of Mel TregS in exosome, and successfully discriminated between healthy individuals and melanoma patients, as well as early and late-stage patients. This approach significantly enhanced detection sensitivity. In sum, our research elucidated fresh insights into the mechanisms governing Treg self-maintenance and communication with surrounding cells in melanoma. We also introduced an innovative method for clinical disease monitoring through SERS technology.


Sujet(s)
Facteurs de transcription Forkhead , Mélanome , Tumeurs cutanées , Lymphocytes T régulateurs , Mélanome/immunologie , Mélanome/génétique , Humains , Lymphocytes T régulateurs/immunologie , Facteurs de transcription Forkhead/métabolisme , Facteurs de transcription Forkhead/génétique , Tumeurs cutanées/immunologie , Tumeurs cutanées/génétique , Régulation de l'expression des gènes tumoraux , Exosomes/métabolisme , Exosomes/génétique , Exosomes/immunologie , Facteurs de transcription à motif basique et à glissière à leucines/génétique , Facteurs de transcription à motif basique et à glissière à leucines/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE