Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 759
Filtrar
1.
Front Immunol ; 15: 1357378, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38720885

RESUMEN

Exosomes carry proteins, metabolites, nucleic acids and lipids from their parent cell of origin. They are derived from cells through exocytosis, are ingested by target cells, and can transfer biological signals between local or distant cells. Therefore, exosomes are often modified in reaction to pathological processes, including infection, cancer, cardiovascular diseases and in response to metabolic perturbations such as obesity and diabetes, all of which involve a significant inflammatory aspect. Here, we discuss how immune cell-derived exosomes origin from neutrophils, T lymphocytes, macrophages impact on the immune reprogramming of diabetes and the associated complications. Besides, exosomes derived from stem cells and their immunomodulatory properties and anti-inflammation effect in diabetes are also reviewed. Moreover, As an important addition to previous reviews, we describes promising directions involving engineered exosomes as well as current challenges of clinical applications in diabetic therapy. Further research on exosomes will explore their potential in translational medicine and provide new avenues for the development of effective clinical diagnostics and therapeutic strategies for immunoregulation of diabetes.


Asunto(s)
Diabetes Mellitus , Exosomas , Inmunomodulación , Exosomas/inmunología , Exosomas/metabolismo , Humanos , Diabetes Mellitus/inmunología , Diabetes Mellitus/terapia , Animales , Macrófagos/inmunología , Macrófagos/metabolismo
2.
Front Immunol ; 15: 1395332, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38726017

RESUMEN

PD-1/PD-L1 signaling is a key factor of local immunosuppression in the tumor microenvironment. Immune checkpoint inhibitors targeting PD-1/PD-L1 signaling have achieved tremendous success in clinic. However, several types of cancer are particularly refractory to the anti-PD-1/PD-L1 treatment. Recently, a series of studies reported that IFN-γ can stimulate cancer cells to release exosomal PD-L1 (exoPD-L1), which possesses the ability to suppress anticancer immune responses and is associated with anti-PD-1 response. In this review, we introduce the PD-1/PD-L1 signaling, including the so-called 'reverse signaling'. Furthermore, we summarize the immune treatments of cancers and pay more attention to immune checkpoint inhibitors targeting PD-1/PD-L1 signaling. Additionally, we review the action mechanisms and regulation of exoPD-L1. We also introduce the function of exoPD-L1 as biomarkers. Finally, we review the methods for analyzing and quantifying exoPD-L1, the therapeutic strategies targeting exoPD-L1 to enhance immunotherapy and the roles of exoPD-L1 beyond cancer. This comprehensive review delves into recent advances of exoPD-L1 and all these findings suggest that exoPD-L1 plays an important role in both cancer and other fields.


Asunto(s)
Antígeno B7-H1 , Exosomas , Inmunoterapia , Neoplasias , Microambiente Tumoral , Humanos , Neoplasias/inmunología , Neoplasias/metabolismo , Antígeno B7-H1/metabolismo , Antígeno B7-H1/inmunología , Exosomas/metabolismo , Exosomas/inmunología , Microambiente Tumoral/inmunología , Animales , Inmunoterapia/métodos , Transducción de Señal , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Biomarcadores de Tumor
3.
J Biochem Mol Toxicol ; 38(6): e23719, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38764138

RESUMEN

Cancer stem cells (CSCs) are associated with the tumor microenvironment (TME). CSCs induce tumorigenesis, tumor recurrence and progression, and resistance to standard therapies. Indeed, CSCs pose an increasing challenge to current cancer therapy due to their stemness or self-renewal properties. The molecular and cellular interactions between heterogeneous CSCs and surrounding TME components and tumor-supporting immune cells show synergistic effects toward treatment failure. In the immunosuppressive TME, CSCs express various immunoregulatory proteins, growth factors, metabolites and cytokines, and also produce exosomes, a type of extracellular vesicles, to protect themselves from host immune surveillance. Among these, the identification and application of CSC-derived exosomes could be considered for the development of therapeutic approaches to eliminate CSCs or cancer, in addition to targeting the modulators that remodel the composition of the TME, as reviewed in this study. Here, we introduce the role of CSCs and how their interaction with TME complicates immunotherapies, and then present the CSC-based immunotherapy and the limitation of these therapies. We describe the biology and role of tumor/CSC-derived exosomes that induce immune suppression in the TME, and finally, introduce their potentials for the development of CSC-based targeted immunotherapy in the future.


Asunto(s)
Células Dendríticas , Exosomas , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia , Células Madre Neoplásicas , Microambiente Tumoral , Humanos , Exosomas/inmunología , Exosomas/metabolismo , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Inmunoterapia/métodos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Vacunas contra el Cáncer/inmunología , Animales
4.
Hum Vaccin Immunother ; 20(1): 2345940, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-38714324

RESUMEN

Traditional vaccines have limits against some persistent infections and pathogens. The development of novel vaccine technologies is particularly critical for the future. Exosomes play an important role in physiological and pathological processes. Exosomes present many advantages, such as inherent capacity being biocompatible, non-toxic, which make them a more desirable candidate for vaccines. However, research on exosomes are in their infancy and the barriers of low yield, low purity, and weak targeting of exosomes limit their applications in vaccines. Accordingly, further exploration is necessary to improve these problems and subsequently facilitate the functional studies of exosomes. In this study, we reviewed the origin, classification, functions, modifications, separation and purification, and characterization methods of exosomes. Meanwhile, we focused on the role and mechanism of exosomes for cancer and COVID-19 vaccines.


Asunto(s)
Vacunas contra la COVID-19 , Vacunas contra el Cáncer , Exosomas , Exosomas/inmunología , Humanos , Vacunas contra la COVID-19/inmunología , Vacunas contra el Cáncer/inmunología , COVID-19/prevención & control , COVID-19/inmunología , SARS-CoV-2/inmunología , Neoplasias/inmunología , Animales , Desarrollo de Vacunas
5.
Elife ; 132024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38743056

RESUMEN

Mutations in the gene for ß-catenin cause liver cancer cells to release fewer exosomes, which reduces the number of immune cells infiltrating the tumor.


Asunto(s)
Escape del Tumor , Humanos , beta Catenina/metabolismo , beta Catenina/genética , Exosomas/inmunología , Exosomas/metabolismo , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/genética , Mutación , Sistema Inmunológico/inmunología , Neoplasias/inmunología , Neoplasias/genética
6.
Int J Nanomedicine ; 19: 3943-3956, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38708179

RESUMEN

Autoimmune diseases refer to a group of conditions where the immune system produces an immune response against self-antigens, resulting in tissue damage. These diseases have profound impacts on the health of patients. In recent years, with the rapid development in the field of biomedicine, engineered exosomes have emerged as a noteworthy class of biogenic nanoparticles. By precisely manipulating the cargo and surface markers of exosomes, engineered exosomes have gained enhanced anti-inflammatory, immunomodulatory, and tissue reparative abilities, providing new prospects for the treatment of autoimmune diseases. Engineered exosomes not only facilitate the efficient delivery of bioactive molecules including nucleic acids, proteins, and cytokines, but also possess the capability to modulate immune cell functions, suppress inflammation, and restore immune homeostasis. This review mainly focuses on the applications of engineered exosomes in several typical autoimmune diseases. Additionally, this article comprehensively summarizes the current approaches for modification and engineering of exosomes and outlines their prospects in clinical applications. In conclusion, engineered exosomes, as an innovative therapeutic approach, hold promise for the management of autoimmune diseases. However, while significant progress has been made, further rigorous research is still needed to address the challenges that engineered exosomes may encounter in the therapeutic intervention process, in order to facilitate their successful translation into clinical practice and ultimately benefit a broader population of patients.


Asunto(s)
Enfermedades Autoinmunes , Exosomas , Exosomas/inmunología , Humanos , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/inmunología , Animales , Nanopartículas/química
7.
J Am Chem Soc ; 146(15): 10293-10298, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38569597

RESUMEN

Fractionating and characterizing target samples are fundamental to the analysis of biomolecules. Extracellular vesicles (EVs), containing information regarding the cellular birthplace, are promising targets for biology and medicine. However, the requirement for multiple-step purification in conventional methods hinders analysis of small samples. Here, we apply a DNA origami tripod with a defined aperture of binders (e.g., antibodies against EV biomarkers), which allows us to capture the target molecule. Using exosomes as a model, we show that our tripod nanodevice can capture a specific size range of EVs with cognate biomarkers from a broad distribution of crude EV mixtures. We further demonstrate that the size of captured EVs can be controlled by changing the aperture of the tripods. This simultaneous selection with the size and biomarker approach should simplify the EV purification process and contribute to the precise analysis of target biomolecules from small samples.


Asunto(s)
Biotecnología , Fraccionamiento Celular , ADN , Exosomas , Nanotecnología , ADN/química , Exosomas/química , Exosomas/inmunología , Nanotecnología/métodos , Fraccionamiento Celular/métodos , Anticuerpos/inmunología , Biomarcadores/análisis , Biotecnología/métodos , Microscopía Fluorescente , Imagen Individual de Molécula
8.
Pathol Res Pract ; 257: 155288, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38653088

RESUMEN

Tumor-mediated immunosuppression is a fundamental obstacle to the development of dendritic cell (DC)-based cancer vaccines, which despite their ability to stimulate host anti-tumor CD8 T cell immunity, have not been able to generate meaningful therapeutic responses. Exosomes are inactive membrane vesicles that are nanoscale in size and are produced by the endocytic pathway. They are essential for intercellular communication. Additionally, DC-derived exosomes (DEXs) contained MHC class I/II (MHCI/II), which is frequently complexed with antigens and co-stimulatory molecules and is therefore able to prime CD4 and CD8 T cells that are specific to particular antigens. Indeed, vaccines with DEXs have been shown to exhibit better anti-tumor efficacy in eradicating tumors compared to DC vaccines in pre-clinical models of digestive system tumors. Also, there is room for improvement in the tumor antigenic peptide (TAA) selection process. DCs release highly targeted exosomes when the right antigenic peptide is chosen, which could aid in the creation of DEX-based antitumor vaccines that elicit more targeted immune responses. Coupled with their resistance to tumor immunosuppression, DEXs-based cancer vaccines have been heralded as the superior alternative cell-free therapeutic vaccines over DC vaccines to treat digestive system tumors. In this review, current studies of DEXs cancer vaccines as well as potential future directions will be deliberated.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas , Exosomas , Exosomas/inmunología , Humanos , Células Dendríticas/inmunología , Vacunas contra el Cáncer/uso terapéutico , Vacunas contra el Cáncer/inmunología , Neoplasias del Sistema Digestivo/inmunología , Neoplasias del Sistema Digestivo/terapia , Neoplasias del Sistema Digestivo/patología , Animales , Inmunoterapia/métodos
9.
Front Immunol ; 15: 1385571, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38680498

RESUMEN

Surgery, chemotherapy, and endocrine therapy have improved the overall survival and postoperative recurrence rates of Luminal A, Luminal B, and HER2-positive breast cancers but treatment modalities for triple-negative breast cancer (TNBC) with poor prognosis remain limited. The effective application of the rapidly developing chimeric antigen receptor (CAR)-T cell therapy in hematological tumors provides new ideas for the treatment of breast cancer. Choosing suitable and specific targets is crucial for applying CAR-T therapy for breast cancer treatment. In this paper, we summarize CAR-T therapy's effective targets and potential targets in different subtypes based on the existing research progress, especially for TNBC. CAR-based immunotherapy has resulted in advancements in the treatment of breast cancer. CAR-macrophages, CAR-NK cells, and CAR-mesenchymal stem cells (MSCs) may be more effective and safer for treating solid tumors, such as breast cancer. However, the tumor microenvironment (TME) of breast tumors and the side effects of CAR-T therapy pose challenges to CAR-based immunotherapy. CAR-T cells and CAR-NK cells-derived exosomes are advantageous in tumor therapy. Exosomes carrying CAR for breast cancer immunotherapy are of immense research value and may provide a treatment modality with good treatment effects. In this review, we provide an overview of the development and challenges of CAR-based immunotherapy in treating different subtypes of breast cancer and discuss the progress of CAR-expressing exosomes for breast cancer treatment. We elaborate on the development of CAR-T cells in TNBC therapy and the prospects of using CAR-macrophages, CAR-NK cells, and CAR-MSCs for treating breast cancer.


Asunto(s)
Neoplasias de la Mama , Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos , Microambiente Tumoral , Humanos , Inmunoterapia Adoptiva/métodos , Inmunoterapia Adoptiva/efectos adversos , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/genética , Femenino , Microambiente Tumoral/inmunología , Neoplasias de la Mama/terapia , Neoplasias de la Mama/inmunología , Animales , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/trasplante , Neoplasias de la Mama Triple Negativas/terapia , Neoplasias de la Mama Triple Negativas/inmunología , Linfocitos T/inmunología , Exosomas/inmunología
10.
Int J Nanomedicine ; 18: 3643-3662, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37427367

RESUMEN

Pathological scars are the result of over-repair and excessive tissue proliferation of the skin injury. It may cause serious dysfunction, resulting in psychological and physiological burdens on the patients. Currently, mesenchymal stem cells-derived exosomes (MSC-Exo) displayed a promising therapeutic effect on wound repair and scar attenuation. But the regulatory mechanisms are opinions vary. In view of inflammation has long been proven as the initial factor of wound healing and scarring, and the unique immunomodulation mechanism of MSC-Exo, the utilization of MSC-Exo may be promising therapeutic for pathological scars. However, different immune cells function differently during wound repair and scar formation. The immunoregulatory mechanism of MSC-Exo would differ among different immune cells and molecules. Herein, this review gave a comprehensive summary of MSC-Exo immunomodulating different immune cells in wound healing and scar formation to provide basic theoretical references and therapeutic exploration of inflammatory wound healing and pathological scars.


Asunto(s)
Cicatriz , Exosomas , Sistema Inmunológico , Inmunomodulación , Células Madre Mesenquimatosas , Humanos , Cicatriz/inmunología , Cicatriz/patología , Cicatriz/terapia , Exosomas/inmunología , Exosomas/patología , Sistema Inmunológico/inmunología , Sistema Inmunológico/patología , Inmunomodulación/inmunología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/patología , Cicatrización de Heridas/inmunología
11.
Cells ; 12(8)2023 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-37190011

RESUMEN

Leishmaniasis is a parasitic disease caused by different species of Leishmania and transmitted through the bite of sand flies vector. Macrophages (MΦ), the target cells of Leishmania parasites, are phagocytes that play a crucial role in the innate immune microbial defense and are antigen-presenting cells driving the activation of the acquired immune response. Exploring parasite-host communication may be key in restraining parasite dissemination in the host. Extracellular vesicles (EVs) constitute a group of heterogenous cell-derived membranous structures, naturally produced by all cells and with immunomodulatory potential over target cells. This study examined the immunogenic potential of EVs shed by L. shawi and L. guyanensis in MΦ activation by analyzing the dynamics of major histocompatibility complex (MHC), innate immune receptors, and cytokine generation. L. shawi and L. guyanensis EVs were incorporated by MΦ and modulated innate immune receptors, indicating that EVs cargo can be recognized by MΦ sensors. Moreover, EVs induced MΦ to generate a mix of pro- and anti-inflammatory cytokines and favored the expression of MHCI molecules, suggesting that EVs antigens can be present to T cells, activating the acquired immune response of the host. Since nano-sized vesicles can be used as vehicles of immune mediators or immunomodulatory drugs, parasitic EVs can be exploited by bioengineering approaches for the development of efficient prophylactic or therapeutic tools for leishmaniasis.


Asunto(s)
Micropartículas Derivadas de Células , Exosomas , Interacciones Huésped-Patógeno , Inmunomodulación , Leishmania guyanensis , Leishmania , Activación de Macrófagos , Macrófagos , Leishmania guyanensis/inmunología , Interacciones Huésped-Patógeno/inmunología , Leishmania/inmunología , Animales , Ratones , Línea Celular , Macrófagos/inmunología , Macrófagos/parasitología , Micropartículas Derivadas de Células/inmunología , Micropartículas Derivadas de Células/parasitología , Exosomas/inmunología , Exosomas/parasitología , Péptido Hidrolasas/metabolismo , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Citocinas/metabolismo , Inmunidad Innata
12.
Microb Biotechnol ; 16(7): 1524-1535, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37212362

RESUMEN

Exosomes, membrane vesicles released extracellularly from cells, contain nucleic acids, proteins, lipids and other components, allowing the transfer of material information between cells. Recent studies reported the role of exosomes in pathogenic microbial infection and host immune mechanisms. Brucella-invasive bodies can survive in host cells for a long time and cause chronic infection, which causes tissue damage. Whether exosomes are involved in host anti-Brucella congenital immune responses has not been reported. Here, we extracted and identified exosomes secreted by Brucella melitensis M5 (Exo-M5)-infected macrophages, and performed in vivo and in vitro studies to examine the effects of exosomes carrying antigen on the polarization of macrophages and immune activation. Exo-M5 promoted the polarization of M1 macrophages, which induced the significant secretion of M1 cytokines (tumour necrosis factor-α and interferon-γ) through NF-κB signalling pathways and inhibited the secretion of M2 cytokines (IL-10), thereby inhibiting the intracellular survival of Brucella. Exo-M5 activated innate immunity and promoted the release of IgG2a antibodies that protected mice from Brucella infection and reduced the parasitaemia of Brucella in the spleen. Furthermore, Exo-M5 contained Brucella antigen components, including Omp31 and OmpA. These results demonstrated that exosomes have an important role in immune responses against Brucella, which might help elucidate the mechanisms of host immunity against Brucella infection and aid the search for Brucella biomarkers and the development of new vaccine candidates.


Asunto(s)
Brucelosis , Exosomas , Macrófagos , Brucella melitensis , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/microbiología , Exosomas/inmunología , Exosomas/microbiología , Animales , Ratones , Polaridad Celular , Antígenos Bacterianos/inmunología , Brucelosis/inmunología , Brucelosis/metabolismo , Transducción de Señal , Espacio Intracelular/microbiología , Viabilidad Microbiana
13.
Cell Transplant ; 32: 9636897221148775, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36661068

RESUMEN

Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune disease associated with impaired organ functions that can seriously affect the daily life of patients. Recent SLE therapies frequently elicit adverse reactions and side effects in patients, and clinical heterogeneity is considerable. Mesenchymal stromal cells (MSCs) have anti-inflammatory, tissue repair, and immunomodulatory properties. Their ability to treat autoimmune diseases largely depends on secreted extracellular vesicles, especially exosomes. The effects of exosomes and microRNAs (miRNAs) on SLE have recently attracted interest. This review summarizes the applications of MSCs derived from bone marrow, adipocyte tissue, umbilical cord, synovial membrane, and gingival tissue, as well as exosomes to treating SLE and the key roles of miRNAs. The efficacy of MSCs infusion in SLE patients with impaired autologous MSCs are reviewed, and the potential of exosomes and their contents as drug delivery vectors for treating SLE and other autoimmune diseases in the future are briefly described.


Asunto(s)
Exosomas , Lupus Eritematoso Sistémico , Células Madre Mesenquimatosas , MicroARNs , Humanos , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Exosomas/genética , Exosomas/inmunología , Vesículas Extracelulares/genética , Vesículas Extracelulares/inmunología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/terapia , MicroARNs/genética , MicroARNs/inmunología , Células Madre Mesenquimatosas/inmunología
14.
JCI Insight ; 7(19)2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36214220

RESUMEN

Intercellular communication is critical for homeostasis in mammalian systems, including the gastrointestinal (GI) tract. Exosomes are nanoscale lipid extracellular vesicles that mediate communication between many cell types. Notably, the roles of immune cell exosomes in regulating GI homeostasis and inflammation are largely uncharacterized. By generating mouse strains deficient in cell-specific exosome production, we demonstrate deletion of the small GTPase Rab27A in CD11c+ cells exacerbated murine colitis, which was reversible through administration of DC-derived exosomes. Profiling RNAs within colon exosomes revealed a distinct subset of miRNAs carried by colon- and DC-derived exosomes. Among antiinflammatory exosomal miRNAs, miR-146a was transferred from gut immune cells to myeloid and T cells through a Rab27-dependent mechanism, targeting Traf6, IRAK-1, and NLRP3 in macrophages. Further, we have identified a potentially novel mode of exosome-mediated DC and macrophage crosstalk that is capable of skewing gut macrophages toward an antiinflammatory phenotype. Assessing clinical samples, RAB27A, select miRNAs, and RNA-binding proteins that load exosomal miRNAs were dysregulated in ulcerative colitis patient samples, consistent with our preclinical mouse model findings. Together, our work reveals an exosome-mediated regulatory mechanism underlying gut inflammation and paves the way for potential use of miRNA-containing exosomes as a novel therapeutic for inflammatory bowel disease.


Asunto(s)
Antígenos CD11 , Colitis , Exosomas , Inflamación , Células Mieloides , Animales , Antígenos CD11/genética , Antígenos CD11/inmunología , Colitis/genética , Colitis/inmunología , Exosomas/genética , Exosomas/inmunología , Inflamación/genética , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Intestinos/inmunología , Lípidos , Mamíferos/genética , Mamíferos/inmunología , Ratones , MicroARNs/inmunología , Proteínas de Unión al GTP Monoméricas/inmunología , Células Mieloides/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Factor 6 Asociado a Receptor de TNF/inmunología
15.
Int J Med Sci ; 19(8): 1265-1274, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35928722

RESUMEN

Objective: To investigate the efficiency and potential mechanisms of exosomes from dendritic cells (DCs) transfected with Forkhead box protein P3 (FOXP3) in the development of experimental autoimmune encephalomyelitis (EAE). Method: Mouse bone marrow-derived immature DCs were loaded with adenovirus carrying FOXP3 gene, and exosomes were generated. Then the exosomes with FOXP3 (FOXP3-EXOs) were co-cultured with CD4+T cell in vitro to evaluate their potential on CD4+T cell proliferation and differentiation, and injected into EAE mice to assess their effects on the development of EAE. Result: FOXP3-EXOs were effective to inhibit the CD4+T cell proliferation and the production of Interferon gamma (IFN-γ), interleukin (IL)-6, and IL-17, while they promoted the production of IL-10 in vitro. Moreover, FOXP3-EXOs treatment significantly decreased the neurological scores, reduced the infiltration of inflammatory cells into the spinal cord, and decreased demyelination in comparison to saline and Con-EXOs treated EAE mice. Moreover, the FOXP3-EXOs treatment resulted in obvious increases in the levels of regulatory T (Treg) cells and IL-10, whereas levels of T helper 1 (Th1) cells, Th17 cells, IFN-γ, IL-6, and IL-17 decreased significantly in the splenocyte culture of EAE mice. Conclusion: The present study preliminarily investigated the effects and potential mechanisms of FOXP3-EXOs in EAE and revealed that the FOXP3-EXOs could inhibit the production of Th1 and Th17 cells and promote the production of Treg cells as well as ameliorate the development of EAE. The neuroprotective effects of FOXP3-EXOs on EAE are likely due to the regulation of Th/Treg balance.


Asunto(s)
Células Dendríticas , Encefalomielitis Autoinmune Experimental , Exosomas , Factores de Transcripción Forkhead , Animales , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Exosomas/genética , Exosomas/inmunología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Interferón gamma/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-17/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Ratones , Ratones Endogámicos C57BL , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores , Células Th17
16.
Front Immunol ; 13: 817942, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35154134

RESUMEN

Tumor immunity is involved in malignant tumor progression. Myeloid-derived suppressor cells (MDSCs) play an irreplaceable role in tumor immunity. MDSCs are composed of immature myeloid cells and exhibit obvious immunomodulatory functions. Exosomes released by MDSCs (MDSCs-Exos) have similar effects to parental MDSCs in regulating tumor immunity. In this review, we provided a comprehensive description of the characteristics, functions and mechanisms of exosomes. We analyzed the immunosuppressive, angiogenesis and metastatic effects of MDSCs-Exos in different tumors through multiple perspectives. Immunotherapy targeting MDSCs-Exos has demonstrated great potential in cancers and non-cancerous diseases.


Asunto(s)
Células Supresoras de Origen Mieloide/inmunología , Neoplasias/inmunología , Exosomas/inmunología , Humanos , Inmunoterapia , Células Supresoras de Origen Mieloide/patología , Neoplasias/patología , Neoplasias/terapia , Microambiente Tumoral/inmunología
17.
Nutrients ; 14(2)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35057528

RESUMEN

Gut bacteria release extracellular vesicles (BEVs) as an intercellular communication mechanism that primes the host innate immune system. BEVs from E. coli activate dendritic cells (DCs) and subsequent T-cell responses in a strain-specific manner. The specific immunomodulatory effects were, in part, mediated by differential regulation of miRNAs. This study aimed to deepen understanding of the mechanisms of BEVs to drive specific immune responses by analyzing their impact on DC-secreted cytokines and exosomes. DCs were challenged with BEVs from probiotic and commensal E. coli strains. The ability of DC-secreted factors to activate T-cell responses was assessed by cytokine quantification in indirect DCs/naïve CD4+ T-cells co-cultures on Transwell supports. DC-exosomes were characterized in terms of costimulatory molecules and miRNAs cargo. In the absence of direct cellular contacts, DC-secreted factors triggered secretion of effector cytokines by T-cells with the same trend as direct DC/T-cell co-cultures. The main differences between the strains influenced the production of Th1- and Treg-specific cytokines. Exosomes released by BEV-activated DCs were enriched in surface proteins involved in antigen presentation and T-cell activation, but differed in the content of immune-related miRNA, depending on the origin of the BEVs. These differences were consistent with the derived immune responses.


Asunto(s)
Citocinas/metabolismo , Células Dendríticas/microbiología , Exosomas/microbiología , Vesículas Extracelulares/inmunología , Microbioma Gastrointestinal/inmunología , Presentación de Antígeno , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Comunicación Celular/inmunología , Técnicas de Cocultivo , Escherichia coli/inmunología , Exosomas/inmunología , Humanos , Activación de Linfocitos/inmunología , MicroARNs/metabolismo , Probióticos/administración & dosificación , Linfocitos T/inmunología , Linfocitos T/microbiología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/microbiología
18.
J Exp Clin Cancer Res ; 41(1): 41, 2022 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-35086548

RESUMEN

BACKGROUND: Cancer cells have an imbalance in oxidation-reduction (redox) homeostasis. Understanding the precise mechanisms and the impact of the altered redox microenvironment on the immunologic reaction to tumors is limited. METHODS: We isolated exosomes from ovarian cancer cells through ultracentrifuge and characterized by Western-blots and Nanoparticle Tracking Analysis. 2D, 3D-coculture tumor model, and 3D live cell imaging were used to study the interactions between tumor cells, macrophages and CD3 T cells in vitro. The role of exosomal miR-155-5p in tumor growth was evaluated in xenograft nude mice models and immune-competent mice models. Flow cytometry and flow sorting were used to determine the expression levels of miR-155-5p and PD-L1 in ascites and splenic macrophages, and the percentages of CD3 T cells subpopulations. RESULTS: The elevation of reactive oxygen species (ROS) greatly downregulated exosomal miR-155-5p expression in tumor cells. Neutralization of ROS with N-acetyl-L-cysteine (NAC) increased the levels of miR-155-5p in tumor exosomes that were taken up by macrophages, leading to reduction of macrophage migration and tumor spheroid infiltration. We further found that programmed death ligand 1 (PD-L1) is a functional target of miR-155-5p. Co-culture of macrophages pre-treated with NAC-derived tumor exosomes or exosomal miR-155-5p with T-lymphocytes leading to an increased percentage of CD8+ T-lymphocyte and a decreased CD3+ T cell apoptosis through PD-L1 downregulation. Tumor growth in nude mice was delayed by treatment with NAC-derived tumor exosomes. Delivery of tumor exo-miR-155-5p in immune-intact mice suppressed ovarian cancer progression and macrophage infiltration, and activated CD8+ T cell function. It is of note that exo-miR-155-5p inhibited tumor growth more potently than the PD-L1 antibody, suggesting that in addition to PD-L1, other pathways may also be targeted by this approach. CONCLUSIONS: Our findings demonstrate a novel mechanism, ROS-induced down-regulation of miR-155-5p, by which tumors modulate the microenvironment that favors tumor growth. Understanding of the negative impact of ROS on the tumor immune response will improve current therapeutic strategies. Targeting miR-155-5p can be an alternative approach to prevent formation of an immunosuppressive TME through downregulation of PD-L1 and other immunosuppressive factors.


Asunto(s)
Antígeno B7-H1/metabolismo , Exosomas/inmunología , Inmunidad/inmunología , Macrófagos/metabolismo , MicroARNs/metabolismo , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Desnudos , Especies Reactivas de Oxígeno
19.
Arthritis Rheumatol ; 74(1): 92-104, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34224653

RESUMEN

OBJECTIVE: T cells play a critical role in the pathogenesis of systemic lupus erythematosus (SLE). Serum-derived exosomes are increased in SLE patients and are correlated with disease severity. This study was undertaken to investigate whether T cell-derived exosomal proteins play a role in SLE pathogenesis. METHODS: We characterized proteins in T cell-derived exosomes from SLE patients and healthy controls by MACSPlex exosome analysis and proteomics. To study the potential pathogenic functions of the exosomal protein identified, we generated and characterized T cell-specific transgenic mice that overexpressed that protein in T cells. RESULTS: We identified eosinophil cationic protein (ECP, also called human RNase III) as overexpressed in SLE T cell-derived exosomes. T cell-specific ECP-transgenic mice (n = 5 per group) displayed early induction of serum interferon-γ (IFNγ) levels (P = 0.062) and inflammation of multiple tissue types. Older T cell-specific ECP-transgenic mice (n = 3 per group) also displayed an increase in follicular helper T cell and plasma B cell numbers, and in autoantibody levels (P < 0.01). Single-cell RNA sequencing showed the induction of IFNγ messenger RNA (P = 2.2 × 10-13 ) and inflammatory pathways in ECP-transgenic mouse T cells. Notably, adoptively transferred ECP-containing exosomes stimulated serum autoantibody levels (P < 0.01) and tissue IFNγ levels in the recipient mice (n = 3 per group). The transferred exosomes infiltrated into multiple tissues of the recipient mice, resulting in hepatitis, nephritis, and arthritis. CONCLUSION: Our findings indicate that ECP overexpression in T cells or T cell-derived exosomes may be a biomarker and pathogenic factor for nephritis, hepatitis, and arthritis associated with SLE.


Asunto(s)
Proteína Catiónica del Eosinófilo/biosíntesis , Exosomas/inmunología , Inflamación/inmunología , Interferón gamma/inmunología , Lupus Eritematoso Sistémico/inmunología , Linfocitos T/inmunología , Animales , Humanos , Ratones , Ratones Transgénicos
20.
Inflammation ; 45(1): 460-475, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34596768

RESUMEN

Mesenchymal stem cells induce kidney transplant tolerance by increasing regulatory T (Treg) cells. Bone marrow mesenchymal stem cell exosomes (BMMSC-Ex) promote Treg cell differentiation. Long non-coding RNA differentiation antagonizing non-protein coding RNA (DANCR) is expressed in BMMSCs and can be encapsulated in exosomes. We aimed to explore the role of DANCR in BMMSC-Ex in immune tolerance after kidney transplantation and related mechanism. The isogenic/allograft kidney transplantation mouse model was established, and levels of serum creatinine (SCr) were determined. Hematoxylin-eosin staining was conducted to detect the inflammation, and immunohistochemistry was performed to detect the infiltration of CD4+ T cells. Levels of IFN-γ, IL-17, and IL-2 were examined by ELISA. Flow cytometry was conducted to determine Treg cells. In the allograft group, the inflammatory response was severe, CD4+ T cell infiltration, SCr levels, and plasma rejection-related factors were up-regulated, while injection of BMMSC-Ex reversed the results. BMMSC-Ex increased Treg cells in kidney transplantation mice. Interference with DANCR reversed the promoting effect of BMMSC-Ex on Treg cell differentiation. DANCR bound to SIRT1, promoted ubiquitination and accelerated its degradation. The injection of BMMSC-Ex (after interference with DANCR) promoted SIRT1 levels, inflammatory response, CD4+ T cell infiltration, SCr levels, and plasma rejection related factors' expression, while Treg cells were decreased. LncRNA DANCR in BMMSC-Ex promoted Treg cell differentiation and induced immune tolerance of kidney transplantation by down-regulating SIRT1 expression in CD4+ T cells.


Asunto(s)
Exosomas/inmunología , Tolerancia Inmunológica , Trasplante de Riñón , Células Madre Mesenquimatosas/inmunología , ARN Largo no Codificante/inmunología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Sirtuina 1/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...