Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 802
Filtrar
1.
Mol Biol Rep ; 51(1): 1058, 2024 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-39417912

RESUMEN

BACKGROUND: Triple-negative breast cancer (TNBC) exhibits a lower survival rate in comparison to other BC subtypes. Utilizing dendritic cell (DC) vaccines as a form of immunotherapy is becoming a promising new approach to cancer treatment. However, inadequate immunogenicity of tumor antigens leads to unsatisfactory effectiveness of the DC vaccines. Exosomes are the basis for the latest improvements in tumor immunotherapy. This study examined whether TNBC-derived exosomes elicit immunogenicity on the maturation and function of monocyte-derived DCs and the impact of the exosome-treated monocyte-derived DCs (moDCs) on T cell differentiation. METHODS: exosomes were isolated from MDA-MB-231 TNBC cancer cells and characterized. Monocytes were separated from peripheral blood mononuclear cells and differentiated into DCs. Then, monocyte-derived DCs were treated with TNBC-derived exosomes. Furthermore, the mRNA levels of the genes and cytokines involved in DC maturation and function were examined using qRT-PCR and ELISA assays. We also cocultured TNBC-derived exosome-treated moDCs with T cells and investigated the role of the treatment in T cell differentiation by evaluating the expression of some related genes by qRT-PCR. The concentration of the cytokines secreted from T cells cocultured with exosome-treated moDCs was quantified by the ELISA assays. RESULTS: Our findings showed that TNBC-derived exosomes induce immunogenicity by enhancing moDCs' maturation and function. In addition, exosome-treated moDCs promote cocultured T-cell expansion by inducing TH1 differentiation through increasing cytokine production. CONCLUSION: TNBC-derived exosomes could improve vaccine-elicited immunotherapy by inducing an immunogenic response and enhancing the effectiveness of the DC vaccines. However, this needs to be investigated further in future studies.


Asunto(s)
Diferenciación Celular , Células Dendríticas , Exosomas , Monocitos , Linfocitos T , Neoplasias de la Mama Triple Negativas , Humanos , Exosomas/metabolismo , Exosomas/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Neoplasias de la Mama Triple Negativas/inmunología , Diferenciación Celular/inmunología , Femenino , Monocitos/inmunología , Monocitos/metabolismo , Línea Celular Tumoral , Linfocitos T/inmunología , Linfocitos T/metabolismo , Citocinas/metabolismo , Inmunoterapia/métodos , Técnicas de Cocultivo , Vacunas contra el Cáncer/inmunología , Activación de Linfocitos/inmunología
2.
Parasite Immunol ; 46(10): e13069, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39404451

RESUMEN

Leishmaniasis is considered one of the most critical health concerns in the world. Unfortunately, no protective vaccines exist and conventional treatments are relatively ineffective. Therefore, new strategies are necessary against leishmaniasis. In recent years, exosomes have shown promising therapeutic outcomes in various diseases, including infectious diseases. In this regard, we aimed to explore the effect of the exosome, pyrimethamine and their combination on the anti-parasitic function of RAW264.7 cells against Leishmania major. Exosomes were isolated from the C57BL/6 peritoneal macrophages. L. major infected and non-infected RAW264.7 cells treated with exosomes, pyrimethamine (PM), and exosomes along with PM. The effect of the treatments was analysed on phagocytosis, efferocytosis, the intracellular parasite count, arginase activity, nitric oxide (NO) and reactive oxygen species (ROS) production. Exosomes could significantly elevate the phagocytosis, efferocytosis, NO and ROS in both infected and non-infected groups (Pv < 0.05). The exosomes reduced the arginase activity in both groups (Pv < 0.05). The intracellular parasite count was significantly lower after treatment with exosomes (Pv < 0.05). These results demonstrate that MQ-derived exosomes can enhance in vitro anti-parasitic responses against L. major. This provides a potential pathway for more effective treatments and underscores the importance of further research in this area.


Asunto(s)
Exosomas , Leishmania major , Macrófagos Peritoneales , Ratones Endogámicos C57BL , Fagocitosis , Especies Reactivas de Oxígeno , Animales , Ratones , Exosomas/metabolismo , Exosomas/inmunología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/parasitología , Células RAW 264.7 , Leishmania major/inmunología , Especies Reactivas de Oxígeno/metabolismo , Óxido Nítrico/metabolismo , Antiprotozoarios/farmacología , Arginasa/metabolismo
3.
Parasitol Res ; 123(10): 336, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39347812

RESUMEN

Echinococcosis is a zoonotic disease, which seriously endangers human health. The immune game between parasite and host is not fully understood. Exosomes are thought to be one of the ways of information communication between parasite and host. In this study, we attempted to explore the communication between Echinococcus granulosus and its host through the medium of exosomes. We collected plasma from E. granulosus patients (CE-EXO) and healthy donors (HD-EXO) and extracted exosomes from the plasma. The expression profile of miRNA in plasma was determined by second generation sequencing. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to annotate the function of target genes of differential miRNAs. Meanwhile, we co-cultured plasma exosomes from healthy donors and plasma exosomes from E. granulosus patients with Jurkat T cells with or without phytohaemagglutinin (PHA) stimulation. The expression of CD69 on Jurkat T cells was detected by flow cytometry. The results showed that the miRNA of exosomes between healthy donors and E. granulosus patients was significantly different. GO and KEGG were used to annotate the function of target genes of differential miRNAs. The results indicate that many important pathways are involved in inflammation, metabolism, and immune response after parasite infection, such as p53 signaling pathway, PI3K-Akt signaling pathway, and glycolysis/gluconeogenesis. Flow cytometry showed that CE-EXO reduced the expression of CD69 + on Jurkat T cells. Our present results suggest that these differentially expressed miRNAs may be important regulators of parasite-host interactions. Meanwhile, functional prediction of its target genes provides valuable information for understanding the mechanism of host-parasite interactions. These results provide clues for future studies on E. granulosus escape from host immune attack, which could help control E. granulosus infection.


Asunto(s)
Equinococosis , Echinococcus granulosus , MicroARNs , Humanos , Equinococosis/inmunología , Equinococosis/sangre , Equinococosis/parasitología , Equinococosis/genética , MicroARNs/sangre , MicroARNs/genética , Proyectos Piloto , Echinococcus granulosus/genética , Echinococcus granulosus/inmunología , Animales , Exosomas/genética , Exosomas/inmunología , Exosomas/metabolismo , Inmunomodulación , Células Jurkat , Perfilación de la Expresión Génica , Interacciones Huésped-Parásitos/inmunología
4.
ACS Nano ; 18(39): 26858-26871, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39308426

RESUMEN

The anti-PD-L1 and its bispecific antibodies have exhibited durable antitumor immunity but still elicit immunosuppression mainly caused by tumor-derived exosomes (TDEs), leading to difficulty in clinical transformation. Herein, engineered Escherichia coli Nissle 1917 (EcN) coexpressing anti-PD-L1 and anti-CD9 nanobodies (EcN-Nb) are developed and decorated with zinc-based metal-organic frameworks (MOFs) loaded with indocyanine green (ICG), to generate EcN-Nb-ZIF-8CHO-ICG (ENZC) for spatiotemporal lysis of bacteria for immunotherapy. The tumor-homing hybrid system can specifically release nanobodies in response to near-infrared (NIR) radiation, thereby targeting TDEs and changing their biological distribution, remodeling tumor-associated macrophages to M1 states, activating more effective and cytotoxic T lymphocytes, and finally, leading to the inhibition of tumor proliferation and metastasis. Altogether, the microfluidic-enabled MOF-modified engineered probiotics target TDEs and activate the antitumor immune response in a spatiotemporally manipulated manner, offering promising TDE-targeted immune therapy.


Asunto(s)
Exosomas , Estructuras Metalorgánicas , Probióticos , Anticuerpos de Dominio Único , Exosomas/metabolismo , Exosomas/inmunología , Exosomas/química , Animales , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/inmunología , Ratones , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Humanos , Verde de Indocianina/química , Escherichia coli/genética , Inmunoterapia , Proliferación Celular/efectos de los fármacos , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/patología , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/química , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Antígeno B7-H1/antagonistas & inhibidores
5.
Viruses ; 16(9)2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39339919

RESUMEN

HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) is a progressive demyelinating disease of the spinal cord due to chronic inflammation. Hallmarks of disease pathology include dysfunctional anti-viral responses and the infiltration of HTLV-1-infected CD4+ T cells and HTLV-1-specific CD8+ T cells in the central nervous system. HAM/TSP individuals exhibit CD4+ and CD8+ T cells with elevated co-expression of multiple inhibitory immune checkpoint proteins (ICPs), but ICP blockade strategies can only partially restore CD8+ T-cell effector function. Exosomes, small extracellular vesicles, can enhance the spread of viral infections and blunt anti-viral responses. Here, we evaluated the impact of exosomes isolated from HTLV-1-infected cells and HAM/TSP patient sera on dendritic cell (DC) and T-cell phenotypes and function. We observed that exosomes derived from HTLV-infected cell lines (OSP2) elicit proinflammatory cytokine responses in DCs, promote helper CD4+ T-cell polarization, and suppress CD8+ T-cell effector function. Furthermore, exosomes from individuals with HAM/TSP stimulate CD4+ T-cell polarization, marked by increased Th1 and regulatory T-cell differentiation. We conclude that exosomes in the setting of HAM/TSP are detrimental to DC and T-cell function and may contribute to the progression of pathology with HTLV-1 infection.


Asunto(s)
Linfocitos T CD4-Positivos , Células Dendríticas , Exosomas , Infecciones por HTLV-I , Virus Linfotrópico T Tipo 1 Humano , Paraparesia Espástica Tropical , Linfocitos T Citotóxicos , Células Dendríticas/inmunología , Exosomas/inmunología , Exosomas/metabolismo , Humanos , Virus Linfotrópico T Tipo 1 Humano/inmunología , Paraparesia Espástica Tropical/inmunología , Paraparesia Espástica Tropical/virología , Linfocitos T CD4-Positivos/inmunología , Infecciones por HTLV-I/inmunología , Infecciones por HTLV-I/virología , Linfocitos T Citotóxicos/inmunología , Citocinas/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología
6.
ACS Nano ; 18(40): 27230-27260, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39319751

RESUMEN

The tumor microenvironment (TME) plays a crucial role in cancer progression and immune evasion, partially mediated by the activity of the TME-derived exosomes. These extracellular vesicles are pivotal in shaping immune responses through the transfer of proteins, lipids, and nucleic acids between cells, facilitating a complex interplay that promotes tumor growth and metastasis. This review delves into the dual roles of exosomes in the TME, highlighting both their immunosuppressive functions and their emerging therapeutic potential. Exosomes can inhibit T cell function and promote tumor immune escape by carrying immune-modulatory molecules, such as PD-L1, yet they also hold promise for cancer therapy as vehicles for delivering tumor antigens and costimulatory signals. Additionally, the review discusses the intricate crosstalk mediated by exosomes among various cell types within the TME, influencing both cancer progression and responses to immunotherapies. Moreover, this highlights current challenges and future directions. Collectively, elucidating the detailed mechanisms by which TME-derived exosomes mediate T cell function offers a promising avenue for revolutionizing cancer treatment. Understanding these interactions allows for the development of targeted therapies that manipulate exosomal pathways to enhance the immune system's response to tumors.


Asunto(s)
Exosomas , Inmunoterapia , Neoplasias , Linfocitos T , Microambiente Tumoral , Exosomas/inmunología , Exosomas/metabolismo , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Humanos , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/patología , Linfocitos T/inmunología , Animales
7.
Int Immunopharmacol ; 142(Pt B): 113248, 2024 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-39321698

RESUMEN

Most somatic cells secrete vesicles called exosomes, which contain a variety of biomolecules. Recent research indicates that macrophage-derived exosomes are strongly correlated with tumors, infectious diseases, chronic inflammation, and tissue fibrosis. Therefore, the purpose of this review is to delve into the mechanisms of pathological states and how macrophage-derived exosomes react to them. We also discuss the biological effects of exosomes and how they affect disease. In addition, we have examined the possible uses of exosomes in illness treatment, highlighting both the benefits and drawbacks of these applications.


Asunto(s)
Progresión de la Enfermedad , Exosomas , Macrófagos , Exosomas/metabolismo , Exosomas/inmunología , Humanos , Macrófagos/inmunología , Animales , Factores Inmunológicos/uso terapéutico , Neoplasias/terapia , Neoplasias/inmunología
8.
Front Immunol ; 15: 1451003, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39267748

RESUMEN

The health tissue surrounding a solid tumor, namely the tumor microenvironment (TME), is an extremely complex universe of cells, extracellular matrix, and signals of various nature, that support and protect the growth of cancer cells. The interactions taking place between cancer cells and the TME are crucial not only for tumor growth, invasion, and metastasis but they also play a key role in modulating immune system responses to cancer, and vice-versa. Indeed, tumor-infiltrating immune cells (e.g., T lymphocytes and natural killers) activity is greatly affected by signals (mostly ligands/receptors and paracrine) they receive in the TME, which frequently generate an immunosuppressive milieu. In the last years, it has become evident that soluble and receptor signaling is not the only way of communication between cells in the TME, with extracellular vesicles, such as exosomes, playing a central role. Among the different new kind of vesicles recently discovered, migrasomes look like to be of extreme interest as they are not only different from the others, but also have been reported as able to deliver a very heterogeneous kind of messages, able to profoundly affect recipient cells' behavior. Indeed, the role played by the different classes of extracellular vesicles, especially in the TME, relies on their not-directional diffusion from the originating cells, while migrasomes released from migrating cells do have a directional effect. Migrasomes biology and their involvement in cancer progression, dissemination, and resistance to therapy is still a largely obscure field, but with promising development foreseen in the next future.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Microambiente Tumoral , Humanos , Microambiente Tumoral/inmunología , Vesículas Extracelulares/inmunología , Neoplasias/inmunología , Neoplasias/patología , Animales , Comunicación Celular/inmunología , Exosomas/inmunología , Transducción de Señal , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo
9.
J Agric Food Chem ; 72(38): 21030-21040, 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39283309

RESUMEN

Mammalian milk exosomal miRNAs play an important role in maintaining intestinal immune homeostasis and protecting epithelial barrier function, but the specific miRNAs and whether miRNA-mediated mechanisms are responsible for these benefits remain a matter of investigation. This study isolated sheep milk-derived exosomes (sheep MDEs), identifying the enriched miRNAs in sheep MDEs, oar-miR-148a, and oar-let-7b as key components targeting TLR4 and TRAF1, which was validated by a dual-luciferase reporter assay. In dextran sulfate sodium-induced colitis mice, administration of sheep MDEs alleviated colitis symptoms, reduced colonic inflammation, and systemic oxidative stress, as well as significantly increased colonic oar-miR-148a and oar-let-7b while reducing toll-like receptor 4 (TLR4) and TNF-receptor-associated factor 1 (TRAF1) level. Further characterization in TNF-α-challenged Caco-2 cells showed that overexpression of these miRNAs suppressed the TLR4/TRAF1-IκBα-p65 pathway and reduced IL-6 and IL-12 production. These findings indicate that sheep MDEs exert gastrointestinal anti-inflammatory effects through the miRNA-mediated modulation of TLR4 and TRAF1, highlighting their potential in managing colitis.


Asunto(s)
Colitis , Sulfato de Dextran , Exosomas , MicroARNs , Leche , Factor 1 Asociado a Receptor de TNF , Receptor Toll-Like 4 , Animales , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , MicroARNs/inmunología , Sulfato de Dextran/efectos adversos , Leche/química , Leche/metabolismo , Colitis/inducido químicamente , Colitis/genética , Colitis/inmunología , Colitis/metabolismo , Ratones , Ovinos , Humanos , Exosomas/genética , Exosomas/metabolismo , Exosomas/química , Exosomas/inmunología , Factor 1 Asociado a Receptor de TNF/genética , Factor 1 Asociado a Receptor de TNF/metabolismo , Células CACO-2 , Masculino , Ratones Endogámicos C57BL , Femenino
10.
J Immunol ; 213(7): 923-931, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39284119

RESUMEN

Cancer immunotherapy, including immune checkpoint blockade, has been approved for treatment of patients with many cancer types. However, some patients fail to respond to immunotherapy, and emerging evidence indicates that tumor-derived exosomes (TEX) play a major role in reprogramming the host immune cells by inducing their dysfunction. Focusing on effector T cells, this review illustrates mechanisms of suppression that TEX use, thus promoting tumor escape from the host immune system. TEX carry multiple suppressive signals that drive T cell dysfunction and convert the tumor microenvironment into "an immune desert" in which activated T cells either die or are reprogrammed to mediate protumor functions. The reprogrammed T cells produce a new crop of CD3+ immunoinhibitory exosomes that further amplify suppression mediated by TEX. The result is a profound depletion of antitumor immune effector cells that reflects the defective immune competence of the cancer patient and partly explains why TEX are a significant barrier for cancer immunotherapy.


Asunto(s)
Exosomas , Inmunoterapia , Neoplasias , Microambiente Tumoral , Exosomas/inmunología , Exosomas/metabolismo , Humanos , Neoplasias/inmunología , Neoplasias/terapia , Microambiente Tumoral/inmunología , Animales , Inmunoterapia/métodos , Escape del Tumor/inmunología , Linfocitos T/inmunología
11.
Int J Biochem Cell Biol ; 175: 106637, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39147124

RESUMEN

Exosomes, which are nanosized extracellular vesicles, have emerged as crucial mediators of the crosstalk between tumor cells and the immune system. Intercellular adhesion molecule 1 (ICAM1) plays a crucial role in multiple immune functions as well as in the occurrence, development and metastasis of cancer. As a glycoprotein expressed on the cell membrane, ICAM1 is secreted extracellularly on exosomes and regulates the immunosuppressive microenvironment. However, the role of exosomal ICAM1 in the immune microenvironment of breast cancer bone metastases remains unclear. This study aimed to elucidated the role of exosomal ICAM1 in facilitating CD8+ T cell exhaustion and subsequent bone metastasis in triple-negative breast cancer (TNBC). We demonstrated that TNBC cells release ICAM1-enriched exosomes, and the binding of ICAM1 to its receptor is necessary for the suppressive effect of CD8 T cell proliferation and function. This pivotal engagement not only inhibits CD8+ T cell proliferation and activation but also initiates the development of an immunosuppressive microenvironment that is conducive to TNBC tumor growth and bone metastasis. Moreover, ICAM1 blockade significantly impairs the ability of tumor exosomes to bind to CD8+ T cells, thereby inhibiting their immunosuppressive effects. The present study elucidates the complex interaction between primary tumors and the immune system that is mediated by exosomes and provides a foundation for the development of novel cancer immunotherapies that target ICAM1 with the aim of mitigating TNBC bone metastasis.


Asunto(s)
Neoplasias Óseas , Linfocitos T CD8-positivos , Exosomas , Molécula 1 de Adhesión Intercelular , Neoplasias de la Mama Triple Negativas , Microambiente Tumoral , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/metabolismo , Exosomas/metabolismo , Exosomas/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Humanos , Femenino , Neoplasias Óseas/secundario , Neoplasias Óseas/inmunología , Neoplasias Óseas/patología , Neoplasias Óseas/metabolismo , Animales , Microambiente Tumoral/inmunología , Ratones , Línea Celular Tumoral , Proliferación Celular , Ratones Endogámicos BALB C , Agotamiento de Células T
12.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167457, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39134287

RESUMEN

DNA virus infection is a significant cause of morbidity and mortality in patients with multiple myeloma (MM). Monocyte dysfunction in MM patients plays a central role in infectious complications, but the precise molecular mechanism underlying the reduced resistance of monocytes to viruses in MM patients remains to be elucidated. Here, we found that MM cells were able to transfer microRNAs (miRNAs) to host monocytes/macrophages via MM cell-derived exosomes, resulting in the inhibition of innate antiviral immune responses. The screening of miRNAs enriched in exosomes derived from the bone marrow (BM) of MM patients revealed five miRNAs that negatively regulate the cGAS-STING antiviral immune response. Notably, silencing these miRNAs with antagomiRs in MM-bearing C57BL/KaLwRijHsd mice markedly reduced viral replication. These findings identify a novel mechanism whereby MM cells possess the capacity to inhibit the innate immune response of the host, thereby rendering patients susceptible to viral infection. Consequently, targeting the aberrant expression patterns of characteristic miRNAs in MM patients is a promising avenue for therapeutic intervention. Considering the miRNA score and relevant clinical factors, we formulated a practical and efficient model for the optimal assessment of susceptibility to DNA viral infection in patients with MM.


Asunto(s)
Exosomas , Inmunidad Innata , Proteínas de la Membrana , Ratones Endogámicos C57BL , MicroARNs , Mieloma Múltiple , Nucleotidiltransferasas , MicroARNs/genética , MicroARNs/inmunología , Mieloma Múltiple/inmunología , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Animales , Humanos , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/inmunología , Exosomas/inmunología , Exosomas/genética , Exosomas/metabolismo , Ratones , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Infecciones por Virus ADN/inmunología , Línea Celular Tumoral , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Femenino , Replicación Viral
13.
Virol J ; 21(1): 177, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39107806

RESUMEN

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.


Asunto(s)
Anticuerpos Antivirales , Pollos , Exosomas , Enfermedades de las Aves de Corral , Virus de la Reticuloendoteliosis , Infecciones por Retroviridae , Esparcimiento de Virus , Animales , Exosomas/virología , Exosomas/inmunología , Anticuerpos Antivirales/inmunología , Pollos/virología , Virus de la Reticuloendoteliosis/inmunología , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/transmisión , Enfermedades de las Aves de Corral/inmunología , Infecciones por Retroviridae/virología , Infecciones por Retroviridae/transmisión , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/veterinaria , Anticuerpos Neutralizantes/inmunología , Línea Celular , Viremia/virología , Femenino
14.
Cancer Immunol Immunother ; 73(10): 196, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39105814

RESUMEN

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.


Asunto(s)
Anticuerpos Biespecíficos , Antígeno B7-H1 , Exosomas , Animales , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/inmunología , Exosomas/metabolismo , Exosomas/inmunología , Ratones , Antígeno B7-H1/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Humanos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ratones Endogámicos C57BL , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Complejo CD3/inmunología , Complejo CD3/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Línea Celular Tumoral , Femenino , Movimiento Celular , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Biomed Pharmacother ; 179: 117296, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39167842

RESUMEN

Over the past decades, cancer immunotherapy has encountered challenges such as immunogenicity, inefficiency, and cytotoxicity. Consequently, exosome-based cancer immunotherapy has gained rapid traction as a promising alternative. Exosomes, a type of extracellular vesicles (EVs) ranging from 50 to 150 nm, are self-originating and exhibit fewer side effects compared to traditional therapies. Exosome-based immunotherapy encompasses three significant areas: cancer vaccination, co-inhibitory checkpoints, and adoptive T-cell therapy. Each of these fields leverages the inherent advantages of exosomes, demonstrating substantial potential for individualized tumor therapy and precision medicine. This review aims to elucidate the reasons behind the promise of exosome-based nanoparticles as cancer therapies by examining their characteristics and summarizing the latest research advancements in cancer immunotherapy.


Asunto(s)
Exosomas , Inmunoterapia , Nanopartículas , Neoplasias , Humanos , Exosomas/metabolismo , Exosomas/inmunología , Neoplasias/terapia , Neoplasias/inmunología , Animales , Inmunoterapia/métodos , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/administración & dosificación
16.
Front Immunol ; 15: 1401852, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38994350

RESUMEN

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.


Asunto(s)
Biomarcadores de Tumor , Exosomas , Neoplasias , Humanos , Exosomas/metabolismo , Exosomas/inmunología , Neoplasias/terapia , Neoplasias/inmunología , Animales , Inmunoterapia/métodos , Biopsia Líquida/métodos
17.
J Extracell Vesicles ; 13(7): e12490, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39051742

RESUMEN

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.


Asunto(s)
Antígeno B7-H1 , Complejo CD3 , Vesículas Extracelulares , Anticuerpos de Cadena Única , Linfocitos T , Humanos , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/metabolismo , Antígeno B7-H1/metabolismo , Antígeno B7-H1/inmunología , Animales , Complejo CD3/inmunología , Complejo CD3/metabolismo , Células HEK293 , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ratones , Anticuerpos de Cadena Única/inmunología , Exosomas/metabolismo , Exosomas/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Activación de Linfocitos/inmunología
18.
Front Immunol ; 15: 1435426, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39007145

RESUMEN

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.


Asunto(s)
Exosomas , Desarrollo de Vacunas , Exosomas/inmunología , Exosomas/metabolismo , Humanos , Animales , Vacunas/inmunología , Sistemas de Liberación de Medicamentos
19.
Front Immunol ; 15: 1429442, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39040099

RESUMEN

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.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Rinitis Alérgica , Exosomas/inmunología , Exosomas/metabolismo , Animales , Rinitis Alérgica/terapia , Rinitis Alérgica/inmunología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Inmunomodulación , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Administración Intranasal
20.
Front Immunol ; 15: 1424081, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39040108

RESUMEN

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.


Asunto(s)
Exosomas , Inmunoterapia , Exosomas/inmunología , Exosomas/metabolismo , Humanos , Inmunoterapia/métodos , Animales , Neoplasias/terapia , Neoplasias/inmunología , Comunicación Celular/inmunología , Inmunomodulación , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...