Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Biochem ; 169(2): 155-161, 2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33226400

RESUMEN

Extracellular vesicles (EVs) are small particles that are naturally released from various types of cells. EVs contain a wide variety of cellular components, such as proteins, nucleic acids, lipids and metabolites, which facilitate intercellular communication in diverse biological processes. In the tumour microenvironment, EVs have been shown to play important roles in tumour progression, including immune system-tumour interactions. Although previous studies have convincingly demonstrated the immunosuppressive functions of tumour-derived EVs, some studies have suggested that tumour-derived EVs can also stimulate host immunity, especially in therapeutic conditions. Recent studies have revealed the heterogeneous nature of EVs with different structural and biological characteristics that may account for the divergent functions of EVs in tumour immunity. In this review article, we provide a brief summary of our current understanding of tumour-derived EVs in immune activation and inhibition. We also highlight the emerging utility of EVs in the diagnosis and treatment of cancers and discuss the potential clinical applications of tumour-derived EVs.


Asunto(s)
Comunicación Celular/inmunología , Exosomas/inmunología , Vesículas Extracelulares/inmunología , Neoplasias/inmunología , Animales , Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Microambiente Tumoral
2.
Theranostics ; 10(16): 7436-7447, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32642004

RESUMEN

Background: Increasing evidence points to the critical role of extracellular vesicles (EVs) as molecular parcels that carry a diverse array of bioactive payloads for coordination of complex intracellular signaling. Focused ultrasound (FUS) hyperthermia is a technique for non-invasive, non-ionizing sublethal heating of cells in a near-instantaneous manner; while it has been shown to improve drug delivery and immunological recognition of tumors, its impact on EVs has not been explored to date. The goal of this study was to determine whether FUS impacts the release, proteomic profile, and immune-activating properties of tumor-derived EVs. Methods: Monolayered murine glioma cells were seeded within acoustically transparent cell culture chambers, and FUS hyperthermia was applied to achieve complete coverage of the chamber. Glioma-derived EVs (GEVs) were isolated for characterization by Nanoparticle Tracking Analysis, cryo-electron microscopy and mass spectrometry. An in vitro experimental setup was designed to further dissect the impact of GEVs on innate inflammation; immortalized murine dendritic cells (DCs) were pulsed with GEVs (either naïve or FUS hyperthermia-exposed) and assayed for production of IL-12p70, an important regulator of DC maturation and T helper cell polarization toward the interferon-γ-producing type 1 phenotype. Results: We confirmed that FUS hyperthermia significantly augments GEV release (by ~46%) as well as shifts the proteomic profile of these GEVs. Such shifts included enrichment of common EV-associated markers, downregulation of markers associated with cancer progression and resistance and modulation of inflammation-associated markers. When DCs were pulsed with GEVs, we noted that naïve GEVs suppressed IL-12p70 production by DCs in a GEV dose-dependent manner. In contrast, GEVs from cells exposed to FUS hyperthermia promoted a significant upregulation in IL-12p70 production by DCs, consistent with a pro-inflammatory stimulus. Conclusion: FUS hyperthermia triggers release of proteomically distinct GEVs that are capable of facilitating an important component of innate immune activation, lending both to a potential mechanism by which FUS interfaces with the tumor-immune landscape and to a role for GEV-associated biomarkers in monitoring response to FUS.


Asunto(s)
Vesículas Extracelulares/efectos de la radiación , Glioma/terapia , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Hipertermia Inducida/métodos , Animales , Línea Celular Tumoral , Microscopía por Crioelectrón , Células Dendríticas/inmunología , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/ultraestructura , Glioma/inmunología , Glioma/patología , Humanos , Inmunidad Innata , Interleucina-12/inmunología , Interleucina-12/metabolismo , Ratones , Proteómica , Linfocitos T Colaboradores-Inductores/inmunología , Escape del Tumor/efectos de la radiación , Regulación hacia Arriba/efectos de la radiación
3.
Fish Shellfish Immunol ; 106: 79-102, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32731012

RESUMEN

The American lobster (Homarus americanus) is a commercially important crustacean with an unusual long life span up to 100 years and a comparative animal model of longevity. Therefore, research into its immune system and physiology is of considerable importance both for industry and comparative immunology studies. Peptidylarginine deiminases (PADs) are a phylogenetically conserved enzyme family that catalyses post-translational protein deimination via the conversion of arginine to citrulline. This can lead to structural and functional protein changes, sometimes contributing to protein moonlighting, in health and disease. PADs also regulate the cellular release of extracellular vesicles (EVs), which is an important part of cellular communication, both in normal physiology and in immune responses. Hitherto, studies on EVs in Crustacea are limited and neither PADs nor associated protein deimination have been studied in a Crustacean species. The current study assessed EV and deimination signatures in haemolymph of the American lobster. Lobster EVs were found to be a poly-dispersed population in the 10-500 nm size range, with the majority of smaller EVs, which fell within 22-115 nm. In lobster haemolymph, 9 key immune and metabolic proteins were identified to be post-translationally deiminated, while further 41 deiminated protein hits were identified when searching against a Crustacean database. KEGG (Kyoto encyclopedia of genes and genomes) and GO (gene ontology) enrichment analysis of these deiminated proteins revealed KEGG and GO pathways relating to a number of immune, including anti-pathogenic (viral, bacterial, fungal) and host-pathogen interactions, as well as metabolic pathways, regulation of vesicle and exosome release, mitochondrial function, ATP generation, gene regulation, telomerase homeostasis and developmental processes. The characterisation of EVs, and post-translational deimination signatures, reported in lobster in the current study, and the first time in Crustacea, provides insights into protein moonlighting functions of both species-specific and phylogenetically conserved proteins and EV-mediated communication in this long-lived crustacean. The current study furthermore lays foundation for novel biomarker discovery for lobster aquaculture.


Asunto(s)
Proteínas de Artrópodos/inmunología , Citrulinación/inmunología , Vesículas Extracelulares/inmunología , Nephropidae/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Animales , Vesículas Extracelulares/metabolismo , Hemolinfa/inmunología , Nephropidae/metabolismo
4.
Nat Commun ; 11(1): 1126, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32111847

RESUMEN

The efficacy of nano-mediated drug delivery has been impeded by multiple biological barriers such as the mononuclear phagocyte system (MPS), as well as vascular and interstitial barriers. To overcome the abovementioned obstacles, we report a nano-pathogenoid (NPN) system that can in situ hitchhike circulating neutrophils and supplement photothermal therapy (PTT). Cloaked with bacteria-secreted outer membrane vesicles inheriting pathogen-associated molecular patterns of native bacteria, NPNs are effectively recognized and internalized by neutrophils. The neutrophils migrate towards inflamed tumors, extravasate across the blood vessels, and penetrate through the tumors. Then NPNs are rapidly released from neutrophils in response to inflammatory stimuli and subsequently taken up by tumor cells to exert anticancer effects. Strikingly, due to the excellent targeting efficacy, cisplatin-loaded NPNs combined with PTT completely eradicate tumors in all treated mice. Such a nano-platform represents an efficient and generalizable strategy towards in situ cell hitchhiking as well as enhanced tumor targeted delivery.


Asunto(s)
Quimiotaxis de Leucocito , Sistemas de Liberación de Medicamentos , Nanopartículas/administración & dosificación , Neoplasias/terapia , Neutrófilos/fisiología , Fototerapia , Animales , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/inmunología , Materiales Biomiméticos/administración & dosificación , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Cisplatino/administración & dosificación , Cisplatino/química , Cisplatino/farmacocinética , Liberación de Fármacos , Vesículas Extracelulares/química , Vesículas Extracelulares/inmunología , Inmunoterapia Adoptiva , Inflamación/etiología , Ratones , Nanopartículas/química , Nanopartículas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Activación Neutrófila , Infiltración Neutrófila , Neutrófilos/inmunología , Neutrófilos/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Fototerapia/efectos adversos , Microambiente Tumoral/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Sci Rep ; 9(1): 14661, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31601878

RESUMEN

Extracellular vesicles (EVs) are involved in cell-to-cell communication and modulation of numerous physiological and pathological processes. EVs are found in large quantities in milk and contain several inflammation- and immunity-modulating proteins and microRNAs, through which they exert beneficial effects in several inflammatory disease models. Here, we investigated the effects of two EV subsets, concentrated from commercial cow's milk, on a murine model of colitis induced with dextran sodium sulfate (DSS). P35K EVs, isolated by ultracentrifugation at 35,000 g, and P100K EVs, isolated at 100,000 g, were previously characterized and administered by gavage to healthy and DSS-treated mice. P35K EVs and, to a lesser extent, P100K EVs improved several outcomes associated to DSS-induced colitis, modulated the gut microbiota, restored intestinal impermeability and replenished mucin secretion. Also, P35K EVs modulated innate immunity, while P100K EVs decreased inflammation through the downregulation of colitis-associated microRNAs, especially miR-125b, associated with a higher expression of the NFκB inhibitor TNFAIP3 (A20). These results suggest that different milk EV subsets may improve colitis outcomes through different, and possibly complementary, mechanisms. Further unveiling of these mechanisms might offer new opportunities for improving the life of patients with colitis and be of importance for milk processing, infant milk formulation and general public health.


Asunto(s)
Colitis/dietoterapia , Suplementos Dietéticos , Vesículas Extracelulares/inmunología , Mucosa Intestinal/inmunología , Leche/citología , Animales , Colitis/inducido químicamente , Colitis/inmunología , Colitis/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/inmunología , Humanos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Ratones , Leche/inmunología , Mucinas/metabolismo , Ultracentrifugación
6.
FASEB J ; 33(5): 6442-6455, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30776316

RESUMEN

Cellular stress or injury induces release of endogenous danger signals such as ATP, which plays a central role in activating immune cells. ATP is essential for the release of nonclassically secreted cytokines such as IL-1ß but, paradoxically, has been reported to inhibit the release of classically secreted cytokines such as TNF. Here, we reveal that ATP does switch off soluble TNF (17 kDa) release from LPS-treated macrophages, but rather than inhibiting the entire TNF secretion, ATP packages membrane TNF (26 kDa) within microvesicles (MVs). Secretion of membrane TNF within MVs bypasses the conventional endoplasmic reticulum- and Golgi transport-dependent pathway and is mediated by acid sphingomyelinase. These membrane TNF-carrying MVs are biologically more potent than soluble TNF in vivo, producing significant lung inflammation in mice. Thus, ATP critically alters TNF trafficking and secretion from macrophages, inducing novel unconventional membrane TNF signaling via MVs without direct cell-to-cell contact. These data have crucial implications for this key cytokine, particularly when therapeutically targeting TNF in acute inflammatory diseases.-Soni, S., O'Dea, K. P., Tan, Y. Y., Cho, K., Abe, E., Romano, R., Cui, J., Ma, D., Sarathchandra, P., Wilson, M. R., Takata, M. ATP redirects cytokine trafficking and promotes novel membrane TNF signaling via microvesicles.


Asunto(s)
Adenosina Trifosfato/inmunología , Membrana Celular/inmunología , Vesículas Extracelulares/inmunología , Macrófagos/inmunología , Neumonía/inmunología , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Enfermedad Aguda , Adenosina Trifosfato/genética , Animales , Comunicación Celular/genética , Comunicación Celular/inmunología , Membrana Celular/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/inmunología , Vesículas Extracelulares/genética , Aparato de Golgi/genética , Aparato de Golgi/inmunología , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Noqueados , Neumonía/inducido químicamente , Neumonía/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Necrosis Tumoral alfa/genética
7.
J Reprod Immunol ; 120: 34-41, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28441551

RESUMEN

OBJECTIVES: Preeclampsia is characterised by systemic endothelial cell dysfunction thought to be triggered by toxic/dangerous factors from the placenta, including placental extracellular vesicles (EVs). Why placental EVs become toxic is unknown. We previously reported that preeclamptic sera produced toxic/dangerous placental macrovesicles but whether small EVs are also toxic/dangerous in preeclampsia is unknown. STUDY DESIGN: First trimester placental explants were treated with 10% preeclamptic or control sera (n=10) for 24h. Micro- and nano-vesicles were harvested by sequential centrifugation. Micro- or nano-vesicles were also exposed to monolayers of endothelial cells in the presence or absence of nifedipine (50µg/ml) or labetalol (0.5µg/ml) which are well-known anti-hypertensives in clinical practices. MAIN OUTCOMES MEASURES: The number and size of micro- and nano-vesicles were counted. Endothelial cell-surface intercellular adhesion molecule 1 (ICAM-1) and high mobility group box 1 (HMGB1) levels in micro- or nano-vesicles were measured by immunoassays. RESULTS: Neither the amount nor size of both micro- and nano-vesicles was different after treating placental explants with preeclamptic or control sera. The levels of HMGB1 were significantly increased in both micro- and nano-vesicles from preeclamptic sera treated placental explants (p<0.03). Exposing endothelial cells to micro- or nano-vesicles from preeclamptic sera-treated placental explants induced endothelial activation, but it was reversed by co-incubation with nifedipine (p=0.004) or labetalol (p=0.002). CONCLUSION: Our data demonstrate that preeclamptic sera produce toxic/dangerous micro- and nano-placental EVs which activated endothelial cells. This effect was reversed by antihypertensives. The increased levels of HMGB1 in EVs may contribute to endothelial cell activation.


Asunto(s)
Células Endoteliales/inmunología , Proteína HMGB1/metabolismo , Sueros Inmunes/inmunología , Placenta/inmunología , Preeclampsia/inmunología , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Vesículas Extracelulares/inmunología , Femenino , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Labetalol/farmacología , Nanopartículas , Nifedipino/farmacología , Embarazo , Primer Trimestre del Embarazo , Tocolíticos/farmacología , Regulación hacia Arriba
8.
J Proteome Res ; 16(1): 217-227, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-27723984

RESUMEN

Influenza A viruses (IAVs) are aggressive pathogens that cause acute respiratory diseases and annual epidemics in humans. Host defense against IAV infection is initiated by macrophages, which are the principal effector cells of the innate immune system. We have previously shown that IAV infection of human macrophages is associated with robust secretion of proteins via conventional and unconventional protein release pathways. Here we have characterized unconventional, extracellular vesicle (EV)-mediated protein secretion in human macrophages during IAV infection using proteomics, bioinformatics, and functional studies. We demonstrate that at 9 h postinfection a robust EV-mediated protein secretion takes place. We identified 2359 human proteins from EVs of IAV-infected macrophages compared with 1448 proteins identified from EVs of control cells. Bioinformatic analysis shows that many proteins involved in translation, like components of spliceosome machinery and the ribosome, are secreted in EVs in response to IAV infection. Our data also shows that EVs derived from IAV-infected macrophages contain fatty acid-binding proteins, antiviral cytokines, copper metabolism Murr-1 domain proteins, and autophagy-related proteins. In addition, our data suggest that secretory autophagy plays a role in activating EV-mediated protein secretion during IAV infection.


Asunto(s)
Vesículas Extracelulares/genética , Interacciones Huésped-Patógeno , Subtipo H3N2 del Virus de la Influenza A/fisiología , Macrófagos/metabolismo , Proteoma/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/inmunología , Proteínas Relacionadas con la Autofagia/metabolismo , Biología Computacional , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/virología , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/inmunología , Proteínas de Unión a Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Ontología de Genes , Humanos , Macrófagos/inmunología , Macrófagos/virología , Anotación de Secuencia Molecular , Cultivo Primario de Células , Biosíntesis de Proteínas , Proteoma/inmunología , Proteoma/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA