Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 24(2): 239-254, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36604547

RESUMEN

Metastasis is the leading cause of cancer-related deaths and myeloid cells are critical in the metastatic microenvironment. Here, we explore the implications of reprogramming pre-metastatic niche myeloid cells by inducing trained immunity with whole beta-glucan particle (WGP). WGP-trained macrophages had increased responsiveness not only to lipopolysaccharide but also to tumor-derived factors. WGP in vivo treatment led to a trained immunity phenotype in lung interstitial macrophages, resulting in inhibition of tumor metastasis and survival prolongation in multiple mouse models of metastasis. WGP-induced trained immunity is mediated by the metabolite sphingosine-1-phosphate. Adoptive transfer of WGP-trained bone marrow-derived macrophages reduced tumor lung metastasis. Blockade of sphingosine-1-phosphate synthesis and mitochondrial fission abrogated WGP-induced trained immunity and its inhibition of lung metastases. WGP also induced trained immunity in human monocytes, resulting in antitumor activity. Our study identifies the metabolic sphingolipid-mitochondrial fission pathway for WGP-induced trained immunity and control over metastasis.


Asunto(s)
Neoplasias Pulmonares , beta-Glucanos , Animales , Ratones , Humanos , Inmunidad Entrenada , Macrófagos , Lisofosfolípidos/metabolismo , Monocitos , Neoplasias Pulmonares/patología , beta-Glucanos/metabolismo , beta-Glucanos/farmacología , Microambiente Tumoral
2.
Small ; 20(20): e2308680, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38225709

RESUMEN

Gut microbiota function has numerous effects on humans and the diet humans consume has emerged as a pivotal determinant of gut microbiota function. Here, a new concept that gut microbiota can be trained by diet-derived exosome-like nanoparticles (ELNs) to release healthy outer membrane vesicles (OMVs) is introduced. Specifically, OMVs released from garlic ELN (GaELNs) trained human gut Akkermansia muciniphila (A. muciniphila) can reverse high-fat diet-induced type 2 diabetes (T2DM) in mice. Oral administration of OMVs released from GaELNs trained A. muciniphila can traffick to the brain where they are taken up by microglial cells, resulting in inhibition of high-fat diet-induced brain inflammation. GaELNs treatment increases the levels of OMV Amuc-1100, P9, and phosphatidylcholines. Increasing the levels of Amuc-1100 and P9 leads to increasing the GLP-1 plasma level. Increasing the levels of phosphatidylcholines is required for inhibition of cGas and STING-mediated inflammation and GLP-1R crosstalk with the insulin pathway that leads to increasing expression of Insulin Receptor Substrate (IRS1 and IRS2) on OMV targeted cells. These findings reveal a molecular mechanism whereby OMVs from plant nanoparticle-trained gut bacteria regulate genes expressed in the brain, and have implications for the treatment of brain dysfunction caused by a metabolic syndrome.


Asunto(s)
Eje Cerebro-Intestino , Diabetes Mellitus Tipo 2 , Exosomas , Ajo , Microbioma Gastrointestinal , Nanopartículas , Diabetes Mellitus Tipo 2/metabolismo , Ajo/química , Animales , Nanopartículas/química , Exosomas/metabolismo , Ratones , Akkermansia , Humanos , Masculino , Dieta Alta en Grasa , Ratones Endogámicos C57BL , Encéfalo/metabolismo , Encéfalo/patología
3.
Hepatology ; 77(4): 1164-1180, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35689610

RESUMEN

BACKGROUND AND AIMS: Intestinal farnesoid X receptor (FXR) plays a critical role in alcohol-associated liver disease (ALD). We aimed to investigate whether alcohol-induced dysbiosis increased intestinal microRNA194 (miR194) that suppressed Fxr transcription and whether Lactobacillus rhamnosus GG-derived exosome-like nanoparticles (LDNPs) protected against ALD through regulation of intestinal miR194-FXR signaling in mice. APPROACH AND RESULTS: Binge-on-chronic alcohol exposure mouse model was utilized. In addition to the decreased ligand-mediated FXR activation, alcohol feeding repressed intestinal Fxr transcription and increased miR194 expression. This transcriptional suppression of Fxr by miR194 was confirmed in intestinal epithelial Caco-2 cells and mouse enteriods. The alcohol feeding-reduced intestinal FXR activation was further demonstrated by the reduced FXR reporter activity in fecal samples and by the decreased fibroblast growth factor 15 (Fgf15) messenger RNA (mRNA) in intestine and protein levels in the serum, which caused an increased hepatic bile acid synthesis and lipogeneses. We further demonstrated that alcohol feeding increased-miR194 expression was mediated by taurine-upregulated gene 1 (Tug1) through gut microbiota regulation of taurine metabolism. Importantly, 3-day oral administration of LDNPs increased bile salt hydrolase (BSH)-harboring bacteria that decreased conjugated bile acids and increased gut taurine concentration, which upregulated Tug1, leading to a suppression of intestinal miR194 expression and recovery of FXR activation. Activated FXR upregulated FGF15 signaling and subsequently reduced hepatic bile acid synthesis and lipogenesis and attenuated ALD. These protective effects of LDNPs were eliminated in intestinal FxrΔIEC and Fgf15-/- mice. We further showed that miR194 was upregulated, whereas BSH activity and taurine levels were decreased in fecal samples of patients with ALD. CONCLUSIONS: Our results demonstrated that gut microbiota-mediated miR194 regulation contributes to ALD pathogenesis and to the protective effects of LDNPs through modulating intestinal FXR signaling.


Asunto(s)
Hepatopatías Alcohólicas , MicroARNs , Animales , Humanos , Ratones , Ácidos y Sales Biliares/metabolismo , Células CACO-2 , Etanol/farmacología , Hígado/patología , Hepatopatías Alcohólicas/metabolismo , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Taurina/farmacología , Nanopartículas
4.
EMBO Rep ; 23(3): e53365, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-34994476

RESUMEN

Bark protects the tree against environmental insults. Here, we analyzed whether this defensive strategy could be utilized to broadly enhance protection against colitis. As a proof of concept, we show that exosome-like nanoparticles (MBELNs) derived from edible mulberry bark confer protection against colitis in a mouse model by promoting heat shock protein family A (Hsp70) member 8 (HSPA8)-mediated activation of the AhR signaling pathway. Activation of this pathway in intestinal epithelial cells leads to the induction of COP9 Constitutive Photomorphogenic Homolog Subunit 8 (COPS8). Utilizing a gut epithelium-specific knockout of COPS8, we demonstrate that COPS8 acts downstream of the AhR pathway and is required for the protective effect of MBELNs by inducing an array of anti-microbial peptides. Our results indicate that MBELNs represent an undescribed mode of inter-kingdom communication in the mammalian intestine through an AhR-COPS8-mediated anti-inflammatory pathway. These data suggest that inflammatory pathways in a microbiota-enriched intestinal environment are regulated by COPS8 and that edible plant-derived ELNs may hold the potential as new agents for the prevention and treatment of gut-related inflammatory disease.


Asunto(s)
Colitis , Exosomas , Morus , Nanopartículas , Animales , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/prevención & control , Modelos Animales de Enfermedad , Exosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Corteza de la Planta/metabolismo
5.
Small ; 18(6): e2105385, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34897972

RESUMEN

Microglia modulate pro-inflammatory and neurotoxic activities. Edible plant-derived factors improve brain function. Current knowledge of the molecular interactions between edible plant-derived factors and the microglial cell is limited. Here an alcohol-induced chronic brain inflammation model is used to identify that the microglial cell is the novel target of oat nanoparticles (oatN). Oral administration of oatN inhibits brain inflammation and improves brain memory function of mice that are fed alcohol. Mechanistically, ethanol activates dectin-1 mediated inflammatory pathway. OatN is taken up by microglial cells via ß-glucan mediated binding to microglial hippocalcin (HPCA) whereas oatN digalactosyldiacylglycerol (DGDG) prevents assess of oatN ß-glucan to dectin-1. Subsequently endocytosed ß-glucan/HPCA is recruited in an endosomal recycling compartment (ERC) via interaction with Rab11a. This complex then sequesters the dectin-1 in the ERC in an oatN ß-glucan dependent manner and alters the location of dectin-1 from Golgi to early endosomes and lysosomes and increases exportation of dectin-1 into exosomes in an Rab11a dependent manner. Collectively, these cascading actions lead to preventing the activation of the alcoholic induced brain inflammation signing pathway(s). This coordinated assembling of the HPCA/Rab11a/dectin-1 complex by oral administration of oatN may contribute to the prevention of brain inflammation.


Asunto(s)
Exosomas , Lectinas Tipo C , Memoria , Microglía , Nanopartículas , Animales , Avena , Encéfalo , Etanol/administración & dosificación , Lectinas Tipo C/metabolismo , Memoria/fisiología , Ratones , Microglía/metabolismo
6.
Mol Ther ; 29(8): 2424-2440, 2021 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-33984520

RESUMEN

Lung inflammation is a hallmark of coronavirus disease 2019 (COVID-19). In this study, we show that mice develop inflamed lung tissue after being administered exosomes released from the lung epithelial cells exposed to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Nsp12 and Nsp13 (exosomesNsp12Nsp13). Mechanistically, we show that exosomesNsp12Nsp13 are taken up by lung macrophages, leading to activation of nuclear factor κB (NF-κB) and the subsequent induction of an array of inflammatory cytokines. Induction of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1ß from exosomesNsp12Nsp13-activated lung macrophages contributes to inducing apoptosis in lung epithelial cells. Induction of exosomesNsp12Nsp13-mediated lung inflammation was abolished with ginger exosome-like nanoparticle (GELN) microRNA (miRNA aly-miR396a-5p. The role of GELNs in inhibition of the SARS-CoV-2-induced cytopathic effect (CPE) was further demonstrated via GELN aly-miR396a-5p- and rlcv-miR-rL1-28-3p-mediated inhibition of expression of Nsp12 and spike genes, respectively. Taken together, our results reveal exosomesNsp12Nsp13 as potentially important contributors to the development of lung inflammation, and GELNs are a potential therapeutic agent to treat COVID-19.


Asunto(s)
COVID-19/metabolismo , Exosomas/metabolismo , MicroARNs/metabolismo , Plantas/metabolismo , Neumonía/metabolismo , Células A549 , Animales , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Citocinas/metabolismo , Células Epiteliales/metabolismo , Humanos , Interleucina-6/metabolismo , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , SARS-CoV-2/patogenicidad , Factor de Necrosis Tumoral alfa/metabolismo , Células U937 , Células Vero
7.
J Immunol ; 202(5): 1623-1634, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30665937

RESUMEN

The role of the mTOR signaling pathway in different myeloid cell subsets is poorly understood in the context of tumor development. In this study, myeloid cell-specific Raptor knockout (KO) mice were used to determine the roles of mechanistic target of rapamycin complex 1 (mTORC1) in regulating macrophage function from Lewis lung carcinoma (LLC) s.c. tumors and lung tumor metastasis. We found no difference in tumor growth between conditional Raptor KO and control mice in the s.c. tumor models, although depletion of mTORC1 decreased the immunosuppressive function of tumor-associated macrophages (TAM). Despite the decreased immunosuppressive activity of TAM, M1-like TAM differentiation was impaired in the s.c. tumor microenvironment of mTORC1 conditional Raptor KO mice due to downregulated CD115 expression on macrophages. In addition, TNF-α production by mTORC1-deficient myeloid cells was also decreased in the s.c. LLC tumors. On the contrary, disruption of mTORC1 in myeloid cells promoted lung cancer metastasis. Accordingly, immunosuppressive interstitial macrophages/metastasis-associated macrophages (CD11b+F4/80high) were accumulated in the lungs of Raptor KO mice in the LLC lung metastasis model, leading to decreased Th1 responses. Taken together, our results demonstrate that differential tumor microenvironment dictates the immunological outcomes of myeloid cells, with mTORC1 disruption leading to different tumor growth phenotypes.


Asunto(s)
Neoplasias Pulmonares/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/inmunología , Células Mieloides/inmunología , Microambiente Tumoral/inmunología , Animales , Neoplasias Pulmonares/patología , Ratones , Ratones Noqueados , Ratones Transgénicos
8.
Immunity ; 35(4): 596-610, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21982596

RESUMEN

Interleukin-23 (IL-23) and CD4(+) T helper 17 (Th17) cells are thought to be critical in psoriasis pathogenesis. Here, we report that IL-23 predominantly stimulated dermal γδ T cells to produce IL-17 that led to disease progression. Dermal γδ T cells constitutively expressed the IL-23 receptor (IL-23R) and transcriptional factor RORγt. IL-17 production from dermal γδ T cells was independent of αß T cells. The epidermal hyperplasia and inflammation induced by IL-23 were significantly decreased in T cell receptor δ-deficient (Tcrd(-/-)) and IL-17 receptor-deficient (Il17ra(-/-)) mice but occurred normally in Tcra(-/-) mice. Imiquimod-induced skin pathology was also significantly decreased in Tcrd(-/-) mice. Perhaps further promoting disease progression, IL-23 stimulated dermal γδ T cell expansion. In psoriasis patients, γδ T cells were greatly increased in affected skin and produced large amounts of IL-17. Thus, IL-23-responsive dermal γδ T cells are the major IL-17 producers in the skin and may represent a novel target for the treatment of psoriasis.


Asunto(s)
Dermatitis/inmunología , Interleucina-17/biosíntesis , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Animales , Dermatitis/patología , Humanos , Interleucina-17/inmunología , Interleucina-17/metabolismo , Interleucina-23/biosíntesis , Interleucina-23/inmunología , Macrófagos/inmunología , Ratones , Ratones Noqueados , Fenotipo , Psoriasis/inmunología , Psoriasis/patología , Receptores de Antígenos de Linfocitos T gamma-delta/deficiencia , Receptores de Interleucina-17/inmunología , Linfocitos T/metabolismo
9.
J Immunol ; 196(5): 2167-80, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26810222

RESUMEN

Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of immature myeloid cells that promote tumor progression. In this study, we demonstrated that activation of a C-type lectin receptor, dectin-1, in MDSC differentially modulates the function of different MDSC subsets. Yeast-derived whole ß-glucan particles (WGP; a ligand to engage and activate dectin-1, oral treatment in vivo) significantly decreased tumor weight and splenomegaly in tumor-bearing mice with reduced accumulation of polymorphonuclear MDSC but not monocytic MDSC (M-MDSC), and decreased polymorphonuclear MDSC suppression in vitro through the induction of respiratory burst and apoptosis. On a different axis, WGP-treated M-MDSC differentiated into F4/80(+)CD11c(+) cells in vitro that served as potent APC to induce Ag-specific CD4(+) and CD8(+) T cell responses in a dectin-1-dependent manner. Additionally, Erk1/2 phosphorylation was required for the acquisition of APC properties in M-MDSC. Moreover, WGP-treated M-MDSC differentiated into CD11c(+) cells in vivo with high MHC class II expression and induced decreased tumor burden when inoculated s.c. with Lewis lung carcinoma cells. This effect was dependent on the dectin-1 receptor. Strikingly, patients with non-small cell lung carcinoma that had received WGP treatment for 10-14 d prior to any other treatment had a decreased frequency of CD14(-)HLA-DR(-)CD11b(+)CD33(+) MDSC in the peripheral blood. Overall, these data indicate that WGP may be a potent immune modulator of MDSC suppressive function and differentiation in cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Monocitos/inmunología , Neutrófilos/inmunología , beta-Glucanos/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Animales , Células Presentadoras de Antígenos/inmunología , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Western Blotting , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Diferenciación Celular/inmunología , Separación Celular , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Humanos , Lectinas Tipo C/metabolismo , Neoplasias Pulmonares/inmunología , Prueba de Cultivo Mixto de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Monocitos/citología , Células Mieloides/citología , Células Mieloides/inmunología , Neutrófilos/citología , Reacción en Cadena en Tiempo Real de la Polimerasa , Levaduras , beta-Glucanos/inmunología
10.
J Immunol ; 196(11): 4477-86, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27183592

RESUMEN

Ab maturation as well as memory B and plasma cell differentiation occur primarily in the germinal centers (GCs). Systemic lupus erythematosus (SLE) may develop as a result of enhanced GC activity. Previous studies have shown that the dysregulated STAT3 pathway is linked to lupus pathogenesis. However, the exact role of STAT3 in regulating SLE disease progression has not been fully understood. In this study, we demonstrated that STAT3 signaling in B cells is essential for GC formation and maintenance as well as Ab response. Increased cell apoptosis and downregulated Bcl-xL and Mcl-1 antiapoptotic gene expression were found in STAT3-deficient GC B cells. The follicular helper T cell response positively correlated with GC B cells and was significantly decreased in immunized B cell STAT3-deficient mice. STAT3 deficiency also led to the defect of plasma cell differentiation. Furthermore, STAT3 deficiency in autoreactive B cells resulted in decreased autoantibody production. Results obtained from B cell STAT3-deficient B6.MRL/lpr mice suggest that STAT3 signaling significantly contributes to SLE pathogenesis by regulation of GC reactivity, autoantibody production, and kidney pathology. Our findings provide new insights into the role of STAT3 signaling in the maintenance of GC formation and GC B cell differentiation and identify STAT3 as a novel target for treatment of SLE.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Centro Germinal/citología , Centro Germinal/inmunología , Lupus Eritematoso Sistémico/inmunología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT3/deficiencia
11.
Mol Ther ; 25(7): 1641-1654, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28274798

RESUMEN

The intestinal immune system is continuously exposed to massive amounts of nanoparticles derived from food. Whether nanoparticles from plants we eat daily have a role in maintaining intestinal immune homeostasis is poorly defined. Here, we present evidence supporting our hypothesis that edible nanoparticles regulate intestinal immune homeostasis by targeting dendritic cells (DCs). Using three mouse colitis models, our data show that orally given nanoparticles isolated from broccoli extracts protect mice against colitis. Broccoli-derived nanoparticle (BDN)-mediated activation of adenosine monophosphate-activated protein kinase (AMPK) in DCs plays a role in not only prevention of DC activation but also induction of tolerant DCs. Adoptively transferring DCs pre-pulsed with total BDN lipids, but not sulforaphane (SFN)-depleted BDN lipids, prevented DSS-induced colitis in C57BL/6 (B6) mice, supporting the role of BDN SFN in the induction of DC tolerance. Adoptively transferring AMPK+/+, but not AMPK-/-, DCs pre-pulsed with SFN prevented DSS-induced colitis in B6 mice, further supporting the DC AMPK role in SFN-mediated prevention of DSS-induced colitis. This finding could open new preventive or therapeutic avenues to address intestinal-related inflammatory diseases via activating AMPK.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Antiinflamatorios/farmacología , Brassica/química , Colitis Ulcerosa/prevención & control , Células Dendríticas/efectos de los fármacos , Nanopartículas/química , Proteínas Quinasas Activadas por AMP/metabolismo , Administración Oral , Traslado Adoptivo , Animales , Antiinflamatorios/química , Antiinflamatorios/metabolismo , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Células Dendríticas/inmunología , Células Dendríticas/patología , Células Dendríticas/trasplante , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Expresión Génica , Humanos , Tolerancia Inmunológica , Isotiocianatos/química , Lípidos/aislamiento & purificación , Lípidos/farmacología , Ratones , Ratones Endogámicos C57BL , Nanopartículas/administración & dosificación , Extractos Vegetales/química , Dodecil Sulfato de Sodio , Sulfóxidos
12.
J Immunol ; 195(10): 5055-65, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26453753

RESUMEN

Tumor-associated macrophages (TAM) with an alternatively activated phenotype have been linked to tumor-elicited inflammation, immunosuppression, and resistance to chemotherapies in cancer, thus representing an attractive target for an effective cancer immunotherapy. In this study, we demonstrate that particulate yeast-derived ß-glucan, a natural polysaccharide compound, converts polarized alternatively activated macrophages or immunosuppressive TAM into a classically activated phenotype with potent immunostimulating activity. This process is associated with macrophage metabolic reprograming with enhanced glycolysis, Krebs cycle, and glutamine utilization. In addition, particulate ß-glucan converts immunosuppressive TAM via the C-type lectin receptor dectin-1-induced spleen tyrosine kinase-Card9-Erk pathway. Further in vivo studies show that oral particulate ß-glucan treatment significantly delays tumor growth, which is associated with in vivo TAM phenotype conversion and enhanced effector T cell activation. Mice injected with particulate ß-glucan-treated TAM mixed with tumor cells have significantly reduced tumor burden with less blood vascular vessels compared with those with TAM plus tumor cell injection. In addition, macrophage depletion significantly reduced the therapeutic efficacy of particulate ß-glucan in tumor-bearing mice. These findings have established a new paradigm for macrophage polarization and immunosuppressive TAM conversion and shed light on the action mode of ß-glucan treatment in cancer.


Asunto(s)
Polisacáridos Fúngicos/farmacología , Lectinas Tipo C/inmunología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/inmunología , Neoplasias Experimentales/tratamiento farmacológico , Saccharomyces cerevisiae/química , beta-Glucanos/farmacología , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/inmunología , Línea Celular Tumoral , Polisacáridos Fúngicos/química , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , beta-Glucanos/química
13.
Mol Ther ; 24(1): 96-105, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26444082

RESUMEN

The lack of access to the brain is a major obstacle for central nervous system drug development. In this study, we demonstrate the capability of a grapefruit-derived nanovector (GNV) to carry miR17 for therapeutic treatment of mouse brain tumor. We show that GNVs coated with folic acid (FA-GNVs) are enhanced for targeting the GNVs to a folate receptor-positive GL-26 brain tumor. Additionally, FA-GNV-coated polyethylenimine (FA-pGNVs) not only enhance the capacity to carry RNA, but the toxicity of the polyethylenimine is eliminated by the GNVs. Intranasal administration of miR17 carried by FA-pGNVs led to rapid delivery of miR17 to the brain that was selectively taken up by GL-26 tumor cells. Mice treated intranasally with FA-pGNV/miR17 had delayed brain tumor growth. Our results demonstrate that this strategy may provide a noninvasive therapeutic approach for treating brain-related disease through intranasal delivery.


Asunto(s)
Neoplasias Encefálicas/terapia , Citrus paradisi/química , Terapia Genética/métodos , MicroARNs/administración & dosificación , MicroARNs/genética , Nanopartículas/química , Administración Intranasal , Animales , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Ácido Fólico/uso terapéutico , Ratones , Nanopartículas/administración & dosificación , Especificidad de Órganos , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Polietileneimina/química , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Am J Pathol ; 184(1): 28-41, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24269592

RESUMEN

Normal and diseased cells release bilayered membrane-bound nanovesicles into interstitial spaces and into bodily fluids. A subgroup of such microvesicles is called exosomes and is described in blood as 30 to 100 nm in diameter and as spherical to cup-shaped nanoparticles with specific surface molecular characteristics (eg, expression of the tetraspanins CD9, CD81, and CD63). Extracellular microvesicles provide local signals (eg, autocrine and paracrine) and distant endocrine signals to cells via the transfer of their contents, which include signal proteins, lipids, miRNAs, and functional mRNAs. Exosomes and related microvesicles also aid cells in exporting less-needed molecules and potentially harmful molecules, including drugs; in the case of neoplasia, the export of chemotherapeutic drugs may facilitate cellular chemoresistance. Cancers have adapted the exosome and related microvesicles as a pathway by which neoplastic cells communicate with each other (autocrine) and with nonneoplastic cells (paracrine and endocrine); via this pathway, cancer suppresses the immune system and establishes a fertile local and distant environment to support neoplastic growth, invasion, and metastases. Because exosomes mirror and bind to the cells from which they arise, they can be used for delivery of drugs, vaccines, and gene therapy, as biomarkers and targets. We review how exosomes and related extracellular microvesicles facilitate the progression and metastases of cancers and describe how these microvesicles may affect clinical care.


Asunto(s)
Exoma/fisiología , Neoplasias/metabolismo , Neoplasias/patología , Animales , Humanos
15.
Am J Pathol ; 184(5): 1355-68, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24681249

RESUMEN

The miRNA cluster miR-17~92 targets mRNAs involved in distinct pathways that either promote or inhibit tumor progression. However, the cellular and molecular mechanisms underlying miR-17~92 cluster-mediated protumorigenic or anti-tumorigenic effects have not been studied. Herein, we determined that inhibition of colon cancer progression is dictated by quantitatively controlling expression of the miR-17~92 cluster. miR-19 in the context of the miR-17~92 cluster at medium levels promoted tumor metastasis through induction of Wnt/ß-catenin-mediated epithelial-mesenchymal transition by targeting to the tumor-suppressor gene, PTEN. However, higher levels of the miR-17~92 cluster switched from PTEN to oncogenes, including Ctnnb1 (ß-catenin) via miR-18a, which resulted in inhibition of tumor growth and metastasis. However, overexpression of Ctnnb1in tumor cells with high-level miR-17~92 did not lead to an increase in the levels of ß-catenin protein, suggesting that other factors regulated by higher levels of miR-17~92 might also contribute to inhibition of tumor growth and metastasis. Those unidentified factors may negatively regulate the production of ß-catenin protein. Collectively, the data presented in this study revealed that higher levels of miR-17~92 were a critical negative regulator for activation of the Wnt/ß-catenin pathway and could have a potential therapeutic application.


Asunto(s)
Neoplasias del Colon/genética , Neoplasias del Colon/patología , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Animales , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/genética , Femenino , Marcación de Gen , Genoma , Humanos , Ratones Endogámicos BALB C , MicroARNs/metabolismo , Datos de Secuencia Molecular , Metástasis de la Neoplasia , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
16.
J Immunol ; 190(11): 5588-99, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23630363

RESUMEN

Ab therapy against surface Ags on tumor cells has demonstrated significant efficacy for some cancers. However, it is costly and patients frequently develop acquired resistance over time. In cases of Ab therapy resistance, T cell responses have been shown to be essential in controlling disease progression. Thus, vaccination that generates a sustained Ab response as well as a T cell response may be more effective and economical. In this article, we have developed a vaccination strategy by targeting protein Ags to B cells via a CD19 single-chain variable fragment miniAb. Using the tumor-associated Ag her-2/neu extracellular domain, we showed that the coengagement of CD19 and BCR induced full B cell activation to produce a high titer of Abs and enhanced CD4 Th2 response and CD8 T cell activation and differentiation. These Abs competitively inhibited humanized her-2/neu Ab binding and were capable of activating the complement and inhibiting human breast cancer growth in vitro. Therapeutic efficacy was demonstrated in vivo using murine mammary carcinoma models. Furthermore, four different extracellular domains of her-2/neu could be targeted to B cells to generate Abs against particular domains with different antitumor properties. This approach may offer a new avenue for vaccine development with significantly lower cost, which may be of use not only for cancer therapy but also for infectious agents.


Asunto(s)
Antígenos CD19/inmunología , Antígenos de Neoplasias/inmunología , Linfocitos B/inmunología , Vacunas contra el Cáncer/inmunología , Neoplasias/inmunología , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD19/metabolismo , Antígenos de Neoplasias/metabolismo , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Citocinas/biosíntesis , Epítopos/inmunología , Humanos , Mediadores de Inflamación/metabolismo , Activación de Linfocitos , Ratones , Ratones Transgénicos , Neoplasias/mortalidad , Neoplasias/patología , Neoplasias/terapia , Unión Proteica/inmunología , Receptor ErbB-2/inmunología , Proteínas Recombinantes de Fusión/inmunología , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/metabolismo , Células Th2/inmunología , Trastuzumab , Carga Tumoral/inmunología
17.
J Immunol ; 190(7): 3579-89, 2013 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-23467936

RESUMEN

Regulation and induction of anergy in NKT cells of the liver can inhibit autoimmune and antitumor responses by mechanisms that are poorly understood. We investigated the effects of PGE2, delivered by intestinal, mucus-derived, exosome-like nanoparticles (IDENs), on NKT cells in mice. In this study, we demonstrate that IDENs migrate to the liver where they induce NKT cell anergy. These effects were mediated by an IDENs' PGE2. Blocking PGE2 synthesis attenuated IDENs inhibition of induction of IFN-γ and IL-4 by α-galactosylceramide (α-GalCer)-stimulated liver NKT cells in a PGE2 E-type prostanoid 2/E-type prostanoid 4 receptor-mediated manner. Proinflammatory conditions enhanced the migration of IDENs to the liver where α-GalCer and PGE2 induced NKT anergy in response to subsequent α-GalCer stimulation. These findings demonstrate that IDENs carrying PGE2 can be transferred from the intestine to the liver, where they act as immune modulators, inducing an anergic-like state of NKT cells. These reagents might be developed as therapeutics for autoimmune liver diseases.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Dinoprostona/metabolismo , Exosomas/metabolismo , Mucosa Intestinal/metabolismo , Hígado/inmunología , Activación de Linfocitos/inmunología , Células T Asesinas Naturales/inmunología , Animales , Anergia Clonal/inmunología , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Exosomas/inmunología , Galactosilceramidas/inmunología , Hepatitis Autoinmune/inmunología , Hepatitis Autoinmune/metabolismo , Humanos , Mucosa Intestinal/inmunología , Hígado/metabolismo , Masculino , Ratones , Transducción de Señal
18.
Mol Ther ; 22(3): 522-534, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23939022

RESUMEN

The gut mucosal immune system is considered to play an important role in counteracting potential adverse effects of food-derived antigens including nanovesicles. Whether nanovesicles naturally released from edible fruit work in a coordinated manner with gut immune cells to maintain the gut in a noninflammatory status is not known. Here, as proof of concept, we demonstrate that grapefruit-derived nanovesicles (GDNs) are selectively taken up by intestinal macrophages and ameliorate dextran sulfate sodium (DSS)-induced mouse colitis. These effects were mediated by upregulating the expression of heme oxygenase-1 (HO-1) and inhibiting the production of IL-1ß and TNF-α in intestinal macrophages. The inherent biocompatibility and biodegradability, stability at wide ranges of pH values, and targeting of intestinal macrophages led us to further develop a novel GDN-based oral delivery system. Incorporating methotrexate (MTX), an anti-inflammatory drug, into GDNs and delivering the MTX-GDNs to mice significantly lowered the MTX toxicity when compared with free MTX, and remarkably increased its therapeutic effects in DSS-induced mouse colitis. These findings demonstrate that GDNs can serve as immune modulators in the intestine, maintain intestinal macrophage homeostasis, and can be developed for oral delivery of small molecule drugs to attenuate inflammatory responses in human disease.


Asunto(s)
Antiinflamatorios/administración & dosificación , Citrus paradisi/química , Colitis/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Mucosa Intestinal/metabolismo , Metotrexato/administración & dosificación , Nanoestructuras/administración & dosificación , Extractos Vegetales/administración & dosificación , Animales , Colitis/inducido químicamente , Sulfato de Dextran , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/metabolismo , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/inmunología , Interleucina-1beta/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Factor de Necrosis Tumoral alfa/metabolismo
19.
J Biol Chem ; 288(39): 28058-67, 2013 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-23926109

RESUMEN

Exosomes released from different types of host cells have different biological effects. We report that exosomes released from retinal astroglial cells (RACs) suppress retinal vessel leakage and inhibit choroidal neovascularization (CNV) in a laser-induced CNV model, whereas exosomes released from retinal pigmental epithelium do not. RAC exosomes inhibit the migration of macrophages and the tubule forming of mouse retinal microvascular endothelial cells. Further, we analyzed antiangiogenic components in RAC exosomes using an angiogenesis array kit and detected several endogenous inhibitors of angiogenesis exclusively present in RAC exosomes, such as endostatin. Moreover, blockade of matrix metalloproteinases in the cleavage of collagen XVIII to form endostatin using FN-439 reverses RAC exosome-mediated retinal vessel leakage. This study demonstrates that exosomes released from retinal tissue cells have different angiogenic effects, with exosomes from RACs containing antiangiogenic components that might protect the eye from angiogenesis and maintain its functional integrity. In addition, by identifying additional components and their functions of RAC exosomes, we might improve the antiangiogenic therapy for CNV in age-related macular degeneration and diabetic retinopathy.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Astrocitos/metabolismo , Neovascularización Coroidal/metabolismo , Exosomas/metabolismo , Retina/citología , Animales , Células de la Médula Ósea/citología , Movimiento Celular , Células Cultivadas , Quimiotaxis , Femenino , Inflamación , Rayos Láser , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Epitelio Pigmentado de la Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Hepatology ; 57(3): 1250-61, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22991247

RESUMEN

UNLABELLED: The Wnt/ß-catenin pathway has been known to play a role in induction of immune tolerance, but its role in the induction and maintenance of natural killer T (NKT) cell anergy is unknown. We found that activation of the Wnt pathways in the liver microenvironment is important for induction of NKT cell anergy. We identified a number of stimuli triggering Wnt/ß-catenin pathway activation, including exogenous NKT cell activator, glycolipid α-GalCer, and endogenous prostaglandin E2 (PGE2). Glycolipid α-GalCer treatment of mice induced the expression of wnt3a and wnt5a in the liver and subsequently resulted in a liver microenvironment that induced NKT cell anergy to α-GalCer restimulation. We also found that circulating PGE2 carried by nanoparticles is stable, and that these nanoparticles are A33(+) . A33(+) is a marker of intestinal epithelial cells, which suggests that the nanoparticles are derived from the intestine. Mice treated with PGE2 associated with intestinal mucus-derived exosome-like nanoparticles (IDENs) induced NKT cell anergy. PGE2 treatment leads to activation of the Wnt/ß-catenin pathway by inactivation of glycogen synthase kinase 3ß of NKT cells. IDEN-associated PGE2 also induces NKT cell anergy through modification of the ability of dendritic cells to induce interleukin-12 and interferon-ß in the context of both glycolipid presentation and Toll-like receptor-mediated pathways. CONCLUSION: These findings demonstrate that IDEN-associated PGE2 serves as an endogenous immune modulator between the liver and intestines and maintains liver NKT cell homeostasis. This finding has implications for development of NKT cell-based immunotherapies. (HEPATOLOGY 2013).


Asunto(s)
Mucosa Intestinal/inmunología , Células Asesinas Naturales/inmunología , Hígado/inmunología , Proteínas Wnt/metabolismo , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo , Animales , Microambiente Celular/inmunología , Anergia Clonal/efectos de los fármacos , Anergia Clonal/inmunología , Dinoprostona/inmunología , Dinoprostona/metabolismo , Galactosilceramidas/farmacología , Tolerancia Inmunológica/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Células Asesinas Naturales/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Moco/inmunología , Moco/metabolismo , Nanopartículas , Proteínas Wnt/inmunología , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/inmunología , Proteína Wnt-5a , Proteína Wnt3A/inmunología , beta Catenina/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA