Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 603
Filtrar
1.
J Med Chem ; 67(16): 13703-13722, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39115891

RESUMO

Extracellular vesicles (EVs) can transfer antigens and immunomodulatory molecules, and such EVs released by antigen-presenting cells equipped with immunostimulatory functions have been utilized for vaccine formulations. A prior high-throughput screening campaign led to the identification of compound 634 (1), which enhanced EV release and increased intracellular Ca2+ influx. Here, we performed systematic structure-activity relationship (SAR) studies to investigate the scaffold for its potency as a vaccine adjuvant. Synthesized compounds were analyzed in vitro for CD63 reporter activity (a marker for EV biogenesis) in human THP-1 cells, induction of Ca2+ influx, IL-12 production, and cell viability in murine bone-marrow-derived dendritic cells. The SAR studies indicated that the ester functional group was requisite, and the sulfur atom of the benzothiadiazole ring replaced with a higher selenium atom (9f) or a bioisosteric ethenyl group (9h) retained potency. Proof-of-concept vaccination studies validated the potency of the selected compounds as novel vaccine adjuvants.


Assuntos
Adjuvantes Imunológicos , Tiadiazóis , Relação Estrutura-Atividade , Humanos , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/química , Adjuvantes Imunológicos/síntese química , Animais , Tiadiazóis/química , Tiadiazóis/farmacologia , Tiadiazóis/síntese química , Camundongos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células THP-1 , Vacinas/imunologia , Vacinas/química , Camundongos Endogâmicos C57BL , Feminino , Vesículas Extracelulares/química , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo
2.
Front Immunol ; 15: 1393248, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39114661

RESUMO

Objective: Beta cell destruction in type 1 diabetes (T1D) results from the combined effect of inflammation and recurrent autoimmunity. In recent years, the role played by beta cells in the development of T1D has evolved from passive victims of the immune system to active contributors in their own destruction. We and others have demonstrated that perturbations in the islet microenvironment promote endoplasmic reticulum (ER) stress in beta cells, leading to enhanced immunogenicity. Among the underlying mechanisms, secretion of extracellular vesicles (EVs) by beta cells has been suggested to mediate the crosstalk with the immune cell compartment. Methods: To study the role of cellular stress in the early events of T1D development, we generated a novel cellular model for constitutive ER stress by modulating the expression of HSPA5, which encodes BiP/GRP78, in EndoC-ßH1 cells. To investigate the role of EVs in the interaction between beta cells and the immune system, we characterized the EV miRNA cargo and evaluated their effect on innate immune cells. Results: Analysis of the transcriptome showed that HSPA5 knockdown resulted in the upregulation of signaling pathways involved in the unfolded protein response (UPR) and changes the miRNA content of EVs, including reduced levels of miRNAs involved in IL-1ß signaling. Treatment of primary human monocytes with EVs from stressed beta cells resulted in increased surface expression of CD11b, HLA-DR, CD40 and CD86 and upregulation of IL-1ß and IL-6. Conclusion: These findings indicate that the content of EVs derived from stressed beta cells can be a mediator of islet inflammation.


Assuntos
Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático , Vesículas Extracelulares , Células Secretoras de Insulina , MicroRNAs , Monócitos , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/imunologia , Monócitos/imunologia , Monócitos/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/imunologia , Humanos , Estresse do Retículo Endoplasmático/imunologia , MicroRNAs/genética , Inflamação/imunologia , Inflamação/metabolismo , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Animais , Linhagem Celular , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Transdução de Sinais , Resposta a Proteínas não Dobradas/imunologia
3.
Methods Mol Biol ; 2843: 95-117, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39141296

RESUMO

Bacterial extracellular vesicles (BEVs) are released from the surface of bacterial cells and contain a diverse molecular cargo. Studies conducted primarily with bacterial pathogens of mammals have shown that BEVs are involved in multiple processes such as cell-cell communication, the delivery of RNA, DNA, and proteins to target cells, protection from stresses, manipulation of host immunity, and other functions. Until a decade ago, the roles of BEVs in plant-bacteria interactions were barely investigated. However, recent studies have shown that BEVs of plant pathogens possess similar functions as their mammalian pathogen counterparts, and more research is now devoted to study their roles and interactions with plants. In the following methods chapter, we provide five well-validated assays to examine the interaction of BEVs with the plant immune system. These assays rely on different markers or immune outputs, which indicate the activation of plant immunity (defense marker gene expression, reactive oxygen species burst, seedling inhibition). Furthermore, we offer assays that directly evaluate the priming of the immune system following BEV challenge and the effectiveness of its response to subsequent local or systemic infection. Altogether, these assays provide a thorough examination to the interactions of BEVs and the plant immune system.


Assuntos
Vesículas Extracelulares , Imunidade Vegetal , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/imunologia , Doenças das Plantas/microbiologia , Doenças das Plantas/imunologia , Interações Hospedeiro-Patógeno/imunologia , Espécies Reativas de Oxigênio/metabolismo , Bactérias/imunologia , Bactérias/metabolismo , Plantas/imunologia , Plantas/microbiologia , Plantas/metabolismo
4.
Methods Mol Biol ; 2843: 163-175, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39141300

RESUMO

Extracellular vesicles are nanosized lipid-bilayered spheres secreted from every living cell and they serve physiological and pathophysiological functions. Bacterial membrane vesicles are shed from both Gram-negative and Gram-positive bacteria and harbor many virulence factors, nuclear material, polysaccharides, proteins, and antigenic determinants, which are essential for immune recognition and evasion. Hence, bacterial membrane vesicles are very promising vaccine candidates. Spray drying is a well-established pharmaceutical technique to produce inhalable dry powders with enhanced stability for formulations of vaccines. In this chapter, we illustrate general guidelines for spray drying of bacterial extracellular vesicles to improve their stability without compromising their immunogenic protective effect. We discuss some of the most important experiments to characterize the generated spray-dried bacterial membrane vesicle powder vaccine.


Assuntos
Vesículas Extracelulares , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/imunologia , Vacinas Bacterianas/imunologia , Vacinas Bacterianas/química , Secagem por Atomização , Pós/química , Humanos , Membrana Celular/metabolismo , Membrana Celular/química , Animais
5.
Methods Mol Biol ; 2843: 195-216, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39141302

RESUMO

Engineered outer membrane vesicles (OMVs) derived from Gram-negative bacteria are a promising vaccine technology for developing immunity against diverse pathogens. However, antigen display on OMVs can be challenging to control and highly variable due to bottlenecks in protein expression and localization to the bacterial host cell's outer membrane, especially for bulky and complex antigens. Here, we describe methods related to a universal vaccine technology called AvidVax (avidin-based vaccine antigen crosslinking) for rapid and simplified assembly of antigens on the exterior of OMVs during vaccine development. The AvidVax platform involves remodeling the OMV surface with multiple copies of a synthetic antigen-binding protein (SNAP), which is an engineered fusion protein comprised of an outer membrane scaffold protein linked to a biotin-binding protein. The resulting SNAPs enable efficient decoration of OMVs with a molecularly diverse array of biotinylated subunit antigens, including globular and membrane proteins, glycans and glycoconjugates, haptens, lipids, nucleic acids, and short peptides. We detail the key steps in the AvidVax vaccine production pipeline including preparation and isolation of SNAP-OMVs, biotinylation and enrichment of vaccine antigens, and formulation and characterization of antigen-loaded SNAP-OMVs.


Assuntos
Antígenos de Bactérias , Biotinilação , Vesículas Extracelulares , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/genética , Vacinas Bacterianas/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Desenvolvimento de Vacinas , Membrana Externa Bacteriana/metabolismo , Membrana Externa Bacteriana/imunologia
6.
Immunity ; 57(8): 1752-1768, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39142276

RESUMO

Extracellular vesicles (EVs), such as ectosomes and exosomes, contain DNA, RNA, proteins and are encased in a phospholipid bilayer. EVs provide intralumenal cargo for delivery into the cytoplasm of recipient cells with an impact on the function of immune cells, in part because their biogenesis can also intersect with antigen processing and presentation. Motile EVs from activated immune cells may increase the frequency of immune synapses on recipient cells in a proximity-independent manner for local and long-distance modulation of systemic immunity in inflammation, autoimmunity, organ fibrosis, cancer, and infections. Natural and engineered EVs exhibit the ability to impact innate and adaptive immunity and are entering clinical trials. EVs are likely a component of an optimally functioning immune system, with the potential to serve as immunotherapeutics. Considering the evolving evidence, it is possible that EVs could be the original primordial organic units that preceded the creation of the first cell.


Assuntos
Vesículas Extracelulares , Humanos , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Animais , Imunidade Inata/imunologia , Imunidade Adaptativa/imunologia , Apresentação de Antígeno/imunologia , Imunidade
7.
Front Immunol ; 15: 1440667, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39176090

RESUMO

Foot and mouth disease (FMD) is a highly contagious infection caused by FMD-virus (FMDV) that affects livestock worldwide with significant economic impact. The main strategy for the control is vaccination with FMDV chemically inactivated with binary ethylenimine (FMDVi). In FMDV infection and vaccination, B cell response plays a major role by providing neutralizing/protective antibodies in animal models and natural hosts. Extracellular vesicles (EVs) and small EVs (sEVs) such as exosomes are important in cellular communication. EVs secreted by antigen-presenting cells (APC) like dendritic cells (DCs) participate in the activation of B and T cells through the presentation of native antigen membrane-associated to B cells or by transferring MHC-peptide complexes to T cells and even complete antigens from DCs. In this study, we demonstrate for the first time that APC activated with the FMDVi O1 Campos vaccine-antigens secrete EVs expressing viral proteins/peptides that could stimulate FMDV-specific immune response. The secretion of EVs-FMDVi is a time-dependent process and can only be isolated within the first 24 h post-activation. These vesicles express classical EVs markers (CD9, CD81, and CD63), along with immunoregulatory molecules (MHC-II and CD86). With an average size of 155 nm, they belong to the category of EVs. Studies conducted in vitro have demonstrated that EVs-FMDVi express antigens that can stimulate a specific B cell response against FMDV, including both marginal zone B cells (MZB) and follicular B cells (FoB). These vesicles can also indirectly or directly affect T cells, indicating that they express both B and T epitopes. Additionally, lymphocyte expansion induced by EVs-FMDVi is greater in splenocytes that have previously encountered viral antigens in vivo. The present study sheds light on the role of EVs derived from APC in regulating the adaptive immunity against FMDV. This novel insight contributes to our current understanding of the immune mechanisms triggered by APC during the antiviral immune response. Furthermore, these findings may have practical implications for the development of new vaccine platforms, providing a rational basis for the design of more effective vaccines against FMDV and other viral diseases.


Assuntos
Células Apresentadoras de Antígenos , Antígenos Virais , Linfócitos B , Vesículas Extracelulares , Vírus da Febre Aftosa , Febre Aftosa , Vacinas Virais , Animais , Vírus da Febre Aftosa/imunologia , Vesículas Extracelulares/imunologia , Linfócitos B/imunologia , Febre Aftosa/imunologia , Febre Aftosa/prevenção & controle , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos Virais/imunologia , Vacinas Virais/imunologia , Proteínas Virais/imunologia , Ativação Linfocitária/imunologia , Células Dendríticas/imunologia , Apresentação de Antígeno/imunologia
8.
J Med Microbiol ; 73(8)2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39133547

RESUMO

Introduction. Tuberculosis (TB), an infectious disease caused by Mycobacterium tuberculosis (M. tb), remains a significant global public health concern. It is crucial to develop more effective vaccines for TB in order to achieve global control of the disease. Extracellular vesicles (EVs) are spherical membrane-bound structures released by pathogens and host cells. During the course of an infection, both pathogen- and host-derived EVs are produced and play important roles in determining the course of the infection. EVs offer intriguing tools as potential vaccines due to their ability to deliver multiple pathogen or host antigens.Hypothesis /Gap Statement. We hypothesized that EVs derived from M. tb and EVs from M. tb-infected macrophages may serve as potential vaccine candidates against M. tb infection.Aim. This study aims to compare the immunogenicity and immune protection between M. tb EVs and M. tb-infected macrophage-derived EVs.Methodology. In this study, EVs were extracted from culture supernatants of M. tb and M. tb-infected macrophages, respectively. Mass spectrometry was employed to explore the antigen composition of H37Rv-Mφ-EVs and H37Rv-EVs. Cytokine profiling and antibody response assays were used to analyse the immunogenicity offered by EVs. Additionally, we used histological examination to evaluate and protective efficacy of the EVs.Results. Our results demonstrated that mice immunized by EVs released from M. tb-infected macrophages induced stronger inflammatory cytokine response than M. tb. Moreover, EVs from M. tb-infected macrophages reinforced T-cell activation and antibody response compared to M. tb EVs. Proteomic analysis revealed that EVs from M. tb-infected macrophages containing immunodominant cargos have stronger binding ability with major histocompatibility complex molecules, which may contribute to the protection from M. tb infection. Indeed, immunization of EVs released from M. tb-infected macrophages significantly reduced the bacterial load and better protection against M. tb infection than EVs from M. tb. Importantly, the selected antigens (Ag85B, ESAT-6 and the Rv0580c) from EVs of M. tb-infected macrophages exhibited effective immunogenicity.Conclusion. Our results suggested that EVs derived from M. tb-infected macrophages might serve as a proper antigenic library for vaccine candidates against M. tb challenge.


Assuntos
Antígenos de Bactérias , Vesículas Extracelulares , Macrófagos , Mycobacterium tuberculosis , Vacinas contra a Tuberculose , Tuberculose , Vesículas Extracelulares/imunologia , Mycobacterium tuberculosis/imunologia , Animais , Antígenos de Bactérias/imunologia , Vacinas contra a Tuberculose/imunologia , Vacinas contra a Tuberculose/administração & dosagem , Camundongos , Macrófagos/imunologia , Macrófagos/microbiologia , Tuberculose/prevenção & controle , Tuberculose/imunologia , Tuberculose/microbiologia , Citocinas/metabolismo , Feminino
9.
J Extracell Vesicles ; 13(7): e12490, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39051742

RESUMO

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.


Assuntos
Antígeno B7-H1 , Complexo CD3 , Vesículas Extracelulares , Anticorpos de Cadeia Única , Linfócitos T , Humanos , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Antígeno B7-H1/metabolismo , Antígeno B7-H1/imunologia , Animais , Complexo CD3/imunologia , Complexo CD3/metabolismo , Células HEK293 , Linfócitos T/imunologia , Linfócitos T/metabolismo , Camundongos , Anticorpos de Cadeia Única/imunologia , Exossomos/metabolismo , Exossomos/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Ativação Linfocitária/imunologia
10.
J Extracell Vesicles ; 13(7): e12485, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39051751

RESUMO

Extracellular vesicles (EVs) are major contributors to immunological responses following solid organ transplantation. Donor derived EVs are best known for their role in transplant rejection through transferring donor major histocompatibility complex proteins to recipient antigen presenting cells, a phenomenon known as ?cross-decoration'. In contrast, donor liver-derived EVs are associated with organ tolerance in small animal models. Therefore, the cellular source of EVs and their cargo could influence their downstream immunological effects. To investigate the immunological effects of EVs released by the liver in a physiological and transplant-relevant model, we isolated EVs being produced during normothermic ex vivo liver perfusion (NEVLP), a novel method of liver storage prior to transplantation. We found EVs were produced by the liver during NEVLP, and these EVs contained multiple anti-inflammatory miRNA species. In terms of function, liver-derived EVs were able to cross-decorate allogeneic cells and suppress the immune response in allogeneic mixed lymphocyte reactions in a concentration-dependent fashion. In terms of cytokine response, the addition of 1 × 109 EVs to the mixed lymphocyte reactions significantly decreased the production of the inflammatory cytokines TNF-α, IL-10 and IFN-γ. In conclusion, we determined physiologically produced liver-derived EVs are immunologically regulatory, which has implications for their role and potential modification in solid organ transplantation.


Assuntos
Vesículas Extracelulares , Transplante de Fígado , Fígado , Perfusão , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/imunologia , Fígado/imunologia , Fígado/metabolismo , Animais , Transplante de Fígado/métodos , Perfusão/métodos , MicroRNAs/metabolismo , Citocinas/metabolismo , Masculino , Camundongos , Rejeição de Enxerto/imunologia , Humanos
11.
J Extracell Vesicles ; 13(7): e12480, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38978304

RESUMO

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.


Assuntos
5'-Nucleotidase , Antígeno B7-H1 , Vesículas Extracelulares , Neoplasias de Cabeça e Pescoço , Neutrófilos , Transdução de Sinais , Carcinoma de Células Escamosas de Cabeça e Pescoço , Microambiente Tumoral , Humanos , Antígeno B7-H1/metabolismo , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/imunologia , Neutrófilos/metabolismo , Neutrófilos/imunologia , Microambiente Tumoral/imunologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/imunologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , 5'-Nucleotidase/metabolismo , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Linhagem Celular Tumoral , Imunomodulação , Adenosina/metabolismo , Proteínas Ligadas por GPI
12.
Sci Rep ; 14(1): 16970, 2024 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-39043800

RESUMO

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.


Assuntos
Linfócitos B , Vesículas Extracelulares , Imunoglobulina G , Camundongos Transgênicos , Animais , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Camundongos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Infecções por Orthomyxoviridae/imunologia , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Fluorescência Verde/genética , Antígenos/imunologia , Switching de Imunoglobulina , Camundongos Endogâmicos C57BL , Centro Germinativo/imunologia , Centro Germinativo/metabolismo
13.
Methods Mol Biol ; 2821: 225-236, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38997493

RESUMO

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.


Assuntos
Antígenos , Vesículas Extracelulares , Cadeias Leves de Imunoglobulina , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/química , Humanos , Cadeias Leves de Imunoglobulina/metabolismo , Cadeias Leves de Imunoglobulina/química , Antígenos/imunologia , Citometria de Fluxo/métodos
14.
J Transl Med ; 22(1): 691, 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39075551

RESUMO

Extracellular vesicles (EVs) are nanosized heat-stable vesicles released by virtually all cells in the body, including tumor cells and tumor-infiltrating dendritic cells (DCs). By carrying molecules from originating cells, EVs work as cell-to-cell communicators in both homeostasis and cancer but may also represent valuable therapeutic and diagnostic tools. This review focuses on the role of tumor-derived EVs (TEVs) in the modulation of DC functions and on the therapeutic potential of both tumor- and DC-derived EVs in the context of immunotherapy and DC-based vaccine design. TEVs were originally characterized for their capability to transfer tumor antigens to DCs but are currently regarded as mainly immunosuppressive because of the expression of DC-inhibiting molecules such as PD-L1, HLA-G, PGE2 and others. However, TEVs may still represent a privileged system to deliver antigenic material to DCs upon appropriate engineering to reduce their immunosuppressive cargo or increase immunogenicity. DC-derived EVs are more promising than tumor-derived EVs since they expose antigen-loaded MHC, costimulatory molecules and NK cell-activating ligands in the absence of an immunosuppressive cargo. Moreover, DC-derived EVs possess several advantages as compared to cell-based drugs such as a higher antigen/MHC concentration and ease of manipulation and a lower sensitivity to immunosuppressive microenvironments. Preclinical models showed that DC-derived EVs efficiently activate tumor-specific NK and T cell responses either directly or indirectly by transferring antigens to tumor-infiltrating DCs. By contrast, however, phase I and II trials showed a limited clinical efficacy of EV-based anticancer vaccines. We discuss that the future of EV-based therapy depends on our capability to overcome major challenges such as a still incomplete understanding of their biology and pharmacokinetic and the lack of standardized methods for high-throughput isolation and purification. Despite this, EVs remain in the limelight as candidates for cancer immunotherapy which may outmatch cell-based strategies in the fullness of their time.


Assuntos
Células Dendríticas , Progressão da Doença , Vesículas Extracelulares , Imunoterapia , Neoplasias , Células Dendríticas/imunologia , Humanos , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/patologia , Animais
15.
Front Immunol ; 15: 1362120, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38962016

RESUMO

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.


Assuntos
Vesículas Extracelulares , Neoplasias , Células-Tronco Neoplásicas , Microambiente Tumoral , Microambiente Tumoral/imunologia , Humanos , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Animais , Comunicação Celular/imunologia , Evasão Tumoral , Imunomodulação
17.
Front Immunol ; 15: 1397967, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38947317

RESUMO

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.


Assuntos
Plaquetas , Vesículas Extracelulares , Tetraspanina 29 , Humanos , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Plaquetas/imunologia , Plaquetas/metabolismo , Tetraspanina 29/metabolismo , Comunicação Celular/imunologia , Transfusão de Plaquetas , Feminino , Linfócitos B/imunologia , Linfócitos B/metabolismo , Masculino , Apirase/metabolismo , Apirase/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Antígenos CD
18.
J Extracell Vesicles ; 13(7): e12457, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39007430

RESUMO

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.


Assuntos
Diferenciação Celular , Vesículas Extracelulares , Sêmen , Linfócitos T Reguladores , Humanos , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Sêmen/metabolismo , Sêmen/imunologia , Masculino , Proliferação de Células , Ativação Linfocitária/imunologia
19.
Int Immunopharmacol ; 139: 112621, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39013216

RESUMO

Ferroptosis is a novel iron-dependent form of cell death discovered in recent years, characterized by the accumulation of ferrous iron, the production of reactive oxygen species (ROS) through the Fenton reaction, and lipid peroxidation, ultimately leading to the disruption of the antioxidant system and cell membrane damage. Extensive research has found that ferroptosis plays a significant role in regulating tumor cell immune evasion, tumor development, and remodeling the tumor microenvironment. Small Extracellular vesicles (sEVs), carrying various bioactive molecules (ncRNA, DNA, proteins), are key nanoscale mediators of intercellular communication. Increasing evidence confirms that EVs can regulate the ferroptosis pathway in tumors, promoting tumor cell immune evasion and reshaping the tumor microenvironment. This article aims to comprehensively review the key mechanisms by which sEVs mediate ferroptosis in cancer and provide new insights into targeting tumor immunotherapy.


Assuntos
Vesículas Extracelulares , Ferroptose , Imunoterapia , Neoplasias , Microambiente Tumoral , Humanos , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/imunologia , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/metabolismo , Imunoterapia/métodos , Animais , Microambiente Tumoral/imunologia , Espécies Reativas de Oxigênio/metabolismo , Evasão Tumoral
20.
Microbiol Res ; 287: 127837, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39059097

RESUMO

Klebsiella pneumoniae, a prominent nosocomial pathogen, poses a critical global health threat due to its multidrug-resistant (MDR) and hypervirulent strains. This comprehensive review focuses into the complex approaches undertaken in the development of vaccines against K. pneumoniae. Traditional methods, such as whole-cell and ribosomal-based vaccines, are compared with modern strategies, including DNA and mRNA vaccines, and extracellular vesicles (EVs), among others. Each method presents unique advantages and challenges, emphasising the complexity of developing an effective vaccine against this pathogen. Significant advancements in computational tools and artificial intelligence (AI) have revolutionised antigen identification and vaccine design, enhancing the precision and efficiency of developing multiepitope-based vaccines. The review also highlights the potential of glycomics and immunoinformatics in identifying key antigenic components and elucidating immune evasion mechanisms employed by K. pneumoniae. Despite progress, challenges remain in ensuring the safety, efficacy, and manufacturability of these vaccines. Notably, EVs demonstrate promise due to their intrinsic adjuvant properties and ability to elicit robust immune responses, although concerns regarding inflammation and antigen variability persist. This review provides a critical overview of the current landscape of K. pneumoniae vaccine development, stressing the need for continued innovation and interdisciplinary collaboration to address this pressing public health issue. The integration of advanced computational methods and AI holds the potential to accelerate the development of effective immunotherapies, paving the way for novel vaccines against MDR K. pneumoniae.


Assuntos
Vacinas Bacterianas , Infecções por Klebsiella , Klebsiella pneumoniae , Desenvolvimento de Vacinas , Klebsiella pneumoniae/imunologia , Klebsiella pneumoniae/genética , Humanos , Vacinas Bacterianas/imunologia , Infecções por Klebsiella/prevenção & controle , Infecções por Klebsiella/imunologia , Infecções por Klebsiella/microbiologia , Animais , Vacinas de DNA/imunologia , Inteligência Artificial , Vesículas Extracelulares/imunologia , Antígenos de Bactérias/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...