Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Am J Cancer Res ; 12(7): 3303-3317, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35968331

RESUMEN

Epithelial ovarian cancer (EOC), a common tumor of the female reproductive system, ranks first in fatalities among gynecological malignancies. Most patients find tumors at late stage and have extremely poor prognoses, which necessitates improvements in early detection. This study applied bioinformatic methods to identify potential biomarkers of EOC. First, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed on differentially expressed genes (DEGs) and hub genes, and a protein-protein interaction (PPI) network was constructed. The network of hub genes was analyzed using GeneMANIA, and an analysis of biological processes was constructed with BINGO. Lastly, hub genes were analyzed for EOC-related oncology using the Oncomine and TCGA databases, and the cBioPortal online platform. Overall, cell division cycle 20 (CDC20) was identified as a key gene in EOC. Short hairpin RNA (shRNA) was used to silence CDC20 to explore its effects on EOC cell proliferation, apoptosis and SRY-related HMG-box 2 (SOX2) expression. DEGs were enriched in pathways related to cell cycle signaling, cancer, progesterone-mediated oocyte maturation, Wnt signaling and P53 signaling. Analysis revealed high expression of CDC20 in EOC tissues and a correlation with histology and tumor grade. CDC20 levels are highest in serous adenocarcinoma, when compared to ovarian clear cell carcinoma, ovarian endometrioid carcinoma and mucinous adenocarcinoma. High CDC20 expression within the tumor is associated with poor EOC prognosis. After silencing CDC20, EOC cell proliferation and migration decreased, apoptosis increased, and SOX2 expression decreased. In conclusion, CDC20 is likely a key biomarker of EOC and may act as an upstream regulator of SOX2 to mediate the SOX2 signaling in the progression of EOC. Future application of CDC20 analysis to early detection may improve prognosis, and it has the potential to be a therapeutic target.

2.
Nanotechnology ; 33(34)2022 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-35580558

RESUMEN

Vaccines have become one of the most effective strategies to deal with various infectious diseases and chronic noninfectious diseases, such as SARS virus, Novel Coronavirus, cancer, etc. However, recent studies have found that the neutralizing antibody titers induced by vaccines would drop to half level or even lower after vaccination. In this study, we designed a novel small-sized positively charged nanofiber-1 (PEI-CNF-1) as a vaccine carrier, which can induce a high long-term humoral immune response by controlled release of antigen. Further studies showed that PEI-CNF-1 could significantly induce the release of immune response factor IL-1ßand bone marrow-derived cell (BMDC) maturation. Moreover, compare to other cellulose nanofibers (CNFs), PEI-CNF-1 combined antigen (ovalbumin, OVA) induced and maintained the highest and longest antibody titers after vaccination. Interestingly, the antibody titers have no significant difference between at 21 and 90 d. Mechanically, we found that PEI-NCF-1 not only could control the slow-release of antigen, but also could be more easily swallowed by macrophages and metabolized by the bodies, thus presenting antigen more effectively. In conclusion, we believe that PEI-CNF-1 have a very high application prospect in inducing long-term humoral immune response, so as to achieve efficient prevention effect to epidemic viruses.


Asunto(s)
COVID-19 , Nanofibras , Vacunas , Adyuvantes Inmunológicos/farmacología , Animales , Antígenos , Celulosa , Inmunidad Humoral , Ratones
3.
Hepatology ; 73(3): 952-967, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32418220

RESUMEN

BACKGROUND AND AIMS: Chronic alcohol consumption is accompanied by intestinal inflammation. However, little is known about how alterations to the intestinal immune system and sphingolipids contribute to the pathogenesis of alcohol-associated liver disease (ALD). APPROACH AND RESULTS: We used wild-type mice, retinoid-related orphan receptor gamma t (RORγt)-deficient mice, sphingosine kinase-deficient mice, and local gut anti-inflammatory, 5-aminosalicyclic acid-treated mice in a chronic-binge ethanol feeding model. Targeted lipidomics assessed the sphingolipids in gut and liver samples. Gut immune cell populations, the amounts of sphingolipids, and the level of liver injury were examined. Alcohol intake induces a pro-inflammatory shift in immune cell populations in the gut, including an increase in Th17 cells. Using RORγt-deficient mice, we found that Th17 cells are required for alcohol-associated gut inflammation and the development of ALD. Treatment with 5-aminosalicyclic acid decreases alcohol-induced liver injury and reverses gut inflammation by the suppression of CD4+ /RORγt+ /interleukin-17A+ cells. Increased Th17 cells were due to up-regulation of sphingosine kinase 1 activity and RORγt activation. We found that S1P/S1PR1 signaling is required for the development of Th17 cell-mediated ALD. Importantly, in vivo intervention blocking of S1P/S1PR1 signaling markedly attenuated alcohol-induced liver inflammation, steatosis, and damage. CONCLUSIONS: Gut inflammation is a functional alteration of immune cells in ALD. Reducing gut Th17 cells leads to reduced liver damage. S1P signaling was crucial in the pathogenesis of ALD in a Th17 cell-dependent manner. Furthermore, our findings suggest that compounds that reduce gut inflammation locally may represent a unique targeted approach in the treatment of ALD.


Asunto(s)
Etanol/efectos adversos , Hígado Graso Alcohólico/prevención & control , Lisofosfolípidos/farmacología , Esfingosina/análogos & derivados , Células Th17/fisiología , Animales , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Modelos Animales de Enfermedad , Hígado Graso Alcohólico/etiología , Femenino , Intestinos/citología , Intestinos/efectos de los fármacos , Intestinos/inmunología , Masculino , Mesalamina/farmacología , Mesalamina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esfingosina/farmacología , Células Th17/efectos de los fármacos
4.
Mol Cancer ; 19(1): 168, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33261601

RESUMEN

Hypoxic stress plays a pivotal role in cancer progression; however, how hypoxia drives tumors to become more aggressive or metastatic and adaptive to adverse environmental stress is still poorly understood. In this study, we revealed that CSN8 might be a key regulatory switch controlling hypoxia-induced malignant tumor progression. We demonstrated that the expression of CSN8 increased significantly in colorectal cancerous tissues, which was correlated with lymph node metastasis and predicted poor patient survival. CSN8 overexpression induces the epithelial-mesenchymal transition (EMT) process in colorectal cancer cells, increasing migration and invasion. CSN8 overexpression arrested cell proliferation, upregulated key dormancy marker (NR2F1, DEC2, p27) and hypoxia response genes (HIF-1α, GLUT1), and dramatically enhanced survival under hypoxia, serum deprivation, or chemo-drug 5-fluorouracil treatment conditions. In particular, silenced CSN8 blocks the EMT and dormancy processes induced by the hypoxia of 1% O2 in vitro and undermines the adaptive capacity of colorectal cancer cells in vivo. The further study showed that CSN8 regulated EMT and dormancy partly by activating the HIF-1α signaling pathway, which increased HIF-1α mRNA expression by activating NF-κB and stabilized the HIF-1α protein via HIF-1α de-ubiquitination. Taken together, CSN8 endows primary colorectal cancer cells with highly aggressive/metastatic and adaptive capacities through regulating both EMT and dormancy induced by hypoxia. CSN8 could serve as a novel prognostic biomarker for colorectal cancer and would be an ideal target of disseminated dormant cell elimination and tumor metastasis, recurrence, and chemoresistance prevention.


Asunto(s)
Complejo del Señalosoma COP9/metabolismo , Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal , Proteínas/metabolismo , Hipoxia Tumoral , Complejo del Señalosoma COP9/genética , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas/genética , Estrés Fisiológico/genética , Hipoxia Tumoral/genética , Regulación hacia Arriba/genética
5.
BMC Pregnancy Childbirth ; 20(1): 284, 2020 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-32393255

RESUMEN

BACKGROUND: Recent studies suggest that there is a link between the gut microbiota and glucose metabolism. This study aimed to compare the gut microbiota during early pregnancy of women with hyperglycymia to those with normal blood glucose. METHODS: Gut microbial composition was analysed in 22 women with hyperglycaemia and 28 age-matched healthy controls during their first prenatal visits (< 20 weeks) using high throughput sequencing of the V3-V4 region of the 16S ribosomal RNA gene. Hyperglycemia was diagnosed based on the criteria recommended by the International Association of Diabetes and Pregnancy Study Groups in 2010. RESULTS: Women with hyperglycemia in pregnancy (HIP) had significantly lower microbial richness and diversity compared with healthy pregnant women. The proportions of the Firmicutes and Bacteroidetes phyla and the ratio of Firmicutes:Bacteroidetes were not different between the two groups. We observed that individuals with HIP had an increased abundance of Nocardiaceae, Fusobacteriaceae, etc., whereas healthy controls had significantly higher levels of Christensenellaceae, Clostridiales_vadinBB60_group, Coriobacteriaceae, etc. Similarly, levels of the members of the Ruminococcaceae family, including Ruminococcaceae_UCG-014, Ruminococcaceae_UCG-003, and Ruminococcaceae_UCG-002, were significantly reduced in the HIP group and were negatively correlated with HbA1c. HbA1c levels were positively correlated with Bacteroidaceae and Enterobacteriaceae and negatively correlated with Christensenellaceae, etc. CRP was positively correlated with the Bacteroidaceae and Fusobacteriaceae families and the Fusobacterium genus. CONCLUSIONS: Our study revealed that individuals with HIP have gut microbial dysbiosis and that certain bacterial groups are associated with glucose metabolism during pregnancy. Further study is needed to provide new ideas to control glucose by modifying the gut microbiota.


Asunto(s)
Glucemia/metabolismo , Microbioma Gastrointestinal , Hiperglucemia/metabolismo , Adulto , Estudios de Casos y Controles , Disbiosis , Heces/microbiología , Femenino , Humanos , Hiperglucemia/microbiología , Embarazo , ARN Ribosómico 16S
6.
Cell Cycle ; 19(5): 577-591, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31992123

RESUMEN

Anti-tumor immune response and the prognosis of tumor are the results of competition between stimulatory and inhibitory checkpoints. Except for upregulating inhibitory checkpoints, lowering some immune accelerating molecules to convert an immunostimulatory microenvironment into an immunodormant one through "decelerating the accelerator" might be another effective immune escape pattern. 4-1BBL is a classical transmembrane costimulatory molecule involving in antitumor immune responses. In contrast, we demonstrated that 4-1BBL is predominantly localized in the nuclei of cancer cells in colon cancer specimens and is positively correlated with tumor size, lymph node metastasis, and a lower survival ratio. Furthermore, the nuclear localization of 4-1BBL was also ascertained in vitro. 4-1BBL knockout (KO) arrests the proliferation and impaired the migration and invasion ability of colon cancer cells in vitro and retarded tumor growth in vivo. 4-1BBL KO increased the accumulation of Gsk3ß in the nuclei of colon cancer cells and consequently decreased the expression of Wnt pathway target genes and thus alter tumor biological behavior. We hypothesized that unlike membrane-expressed 4-1BBL, which stimulates the 4-1BB signaling of antitumor cytotoxic T cells, the nuclear-localized 4-1BBL could facilitate the malignant behavior of colon cancer cells by circumventing antitumor signaling and driving some key oncotropic signal pathway in the nucleus. Nuclear-localized 4-1BBL might be an indicator of colon cancer malignancy and serve as a promising target of immunotherapy.


Asunto(s)
Ligando 4-1BB/metabolismo , Movimiento Celular , Núcleo Celular/metabolismo , Neoplasias del Colon/diagnóstico , Neoplasias del Colon/patología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Ligando 4-1BB/biosíntesis , Ligando 4-1BB/deficiencia , Ligando 4-1BB/genética , Animales , Secuencia de Bases , Línea Celular Tumoral , Núcleo Celular/enzimología , Proliferación Celular , Estudios de Cohortes , Neoplasias del Colon/enzimología , Neoplasias del Colon/metabolismo , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Células HCT116 , Humanos , Ratones , Trasplante de Neoplasias , Pronóstico , Transporte de Proteínas , Análisis de Supervivencia
7.
Cancer Lett ; 443: 67-79, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30502359

RESUMEN

Multiple studies have revealed that long non-coding RNAs (lncRNAs) extensively participate in human cancer malignant progression. The long intergenic non-protein coding RNA 707 (LINC00707), 3087 bp in length, was recently reported to be an essential oncogene in promoting lung adenocarcinoma cell proliferation and metastasis. However, its role in gastric cancer (GC) remains unclear. In this study, we identified that LINC00707 was excessively expressed in GC tissues and correlated with advanced stage, larger tumor size, lymph node metastasis and poorer prognosis in GC patients. In vitro and in vivo assays showed that LINC00707 promote GC cell proliferation and metastasis. Mechanistically, LINC00707 could abundantly interact with mRNA stabilizing protein HuR; "LINC00707-HuR" coalition ulteriorly combined with VAV3/F11R mRNAs and increased their stability. Taken together, our findings prove that LINC00707 may act as an oncogene in GC by regulating mRNA stability and serve as a potential target for GC diagnosis and prognosis.


Asunto(s)
Proteína 1 Similar a ELAV/genética , ARN Largo no Codificante/genética , ARN Mensajero/química , Neoplasias Gástricas/patología , Regulación hacia Arriba , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Proteína 1 Similar a ELAV/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis Linfática , Masculino , Ratones , Estadificación de Neoplasias , Trasplante de Neoplasias , Pronóstico , Estabilidad del ARN , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo
8.
Cell Cycle ; 17(1): 53-63, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28976231

RESUMEN

Inflammatory bowel disease (IBD) results from a chronic intestinal inflammation and tissue destruction via an aberrant immune-driven inflammatory response towards an altered gut microbiota. Dietary intervention is becoming an attractive avenue for the therapy of colitis because diet is a key determinant of the mucosal immune response. Quercetin (QCN) is the most common in nature and the major representative of dietary antioxidant flavonoids, which has been demonstrated to influence the progression of colitis. However, the underlying mechanism of QCN on intestinal immunomodulation remains unclear. Here, our study demonstrated dietary QCN could ameliorate experimental colitis in part by modulating the anti-inflammatory effects and bactericidal capacity of macrophages via Heme oxygenase-1 (Hmox1, HO-1) dependent pathway. It suggested that QCN might restore the proper intestinal host-microbe relationship to ameliorate the colitis via rebalancing the pro-inflammatory, anti-inflammatory and bactericidal function of enteric macrophages. Hence, modulating the function of intestinal macrophages with dietary administration of QCN to restore the immunological hemostasis and rebalance the enteric commensal flora is a potential and promising strategy for IBD therapy.


Asunto(s)
Colitis/tratamiento farmacológico , Colitis/patología , Colon/patología , Dieta , Hemo-Oxigenasa 1/metabolismo , Macrófagos/patología , Quercetina/uso terapéutico , Transducción de Señal , Animales , Colitis/inducido químicamente , Colitis/inmunología , Sulfato de Dextran , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/efectos de los fármacos , Homeostasis , Inflamación/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , Quercetina/administración & dosificación , Quercetina/farmacología , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
9.
Biomark Med ; 11(9): 741-749, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28891298

RESUMEN

AIM: Accumulating evidence suggested that challenge of the maternal-fetal interaction during pregnancy might cause the impairment of immunological hemostasis and lead to gestational diabetes mellitus (GDM) pathological process. Immune checkpoint molecule PD-1 is one of the critical molecule balancing immune response and immunological tolerance. METHODS: PD-1 expressions on T-cell subsets of GDM patients and control groups were measured via flow cytometric analysis and followed up. RESULTS: Downregulation of PD-1 acted as an indicator for GDM occurrence in the third trimester of pregnancy. With the recovery of GDM, PD-1 expression restored to normal level. CONCLUSION: PD-1 expression on T-cell subsets is a novel biomarker for the occurrence and recovery of GDM.


Asunto(s)
Biomarcadores/metabolismo , Diabetes Gestacional/diagnóstico , Receptor de Muerte Celular Programada 1/metabolismo , Adulto , Glucemia/análisis , Estudios de Casos y Controles , Diabetes Gestacional/inmunología , Regulación hacia Abajo , Femenino , Humanos , Embarazo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo
10.
Cytokine ; 95: 27-34, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28214675

RESUMEN

Radiation-induced intestinal injury (RIII) commonly occurs in patients who received radiotherapy for pelvic or abdominal cancer, or who suffered from whole-body irradiation during a nuclear accident. RIII can lead to intestinal disorders and even death given its integrity damage that results from intestinal stem cell (ISC) loss. Recovery from RIII relies on the intensity of supportive treatment, which can attenuate lethal infection and give surviving stem cells an opportunity to regenerate. It has been reported that RSPO1 is a cytokine with potent and specific proliferative effects on intestinal crypt cells. MSCs have multiple RIII-healing effects, including anti-inflammatory and anti-irradiation injury properties, due to its negative immune regulation and its homing ability to the damaged intestinal epithelia. To combine the comprehensive anti-injury potential of MSCs, and the potent ability of RSPO1 as a mitogenic factor for ISCs, we constructed RSPO1-modified C3H10 T1/2 cells and expected that RSPO1, the ISC-proliferative cytokine, could be delivered to the site of injury in a targeted manner. In this study, we transferred C3H10/RSPO1 intravenously via the retro-orbital sinus into mice suffering from abdominal irradiation at lethal dosages. Our findings demonstrated that C3H10/RSPO1 cells are able to directionally migrate to the injury site; enhance ISC survival, proliferation, and differentiation; and effectively repair the radiation-damaged intestinal epithelial cells. This study suggests that the directional delivery of RSPO1 by MSCs is a promising strategy to ameliorate, and even cure, RIII.


Asunto(s)
Mucosa Intestinal/citología , Trasplante de Células Madre Mesenquimatosas , Traumatismos Experimentales por Radiación/terapia , Trombospondinas/genética , Traslado Adoptivo , Animales , Línea Celular , Movimiento Celular , Humanos , Mucosa Intestinal/fisiología , Células Madre Mesenquimatosas/fisiología , Ratones , Traumatismos Experimentales por Radiación/inmunología , Regeneración , Células Madre/fisiología , Transfección
12.
Int J Clin Exp Med ; 8(12): 22529-42, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26885239

RESUMEN

This review aimed to summarize the epidemiology (incidence, prevalence and morality) and risk factors of inflammatory bowel disease (IBD). IBD is a chronic, relapsing, inflammatory disorder of the gastrointestinal tract and includes Crohn's Disease (CD) and ulcerative colitis (UC). IBD has increasing incidence and prevalence in most of countries and becomes a global emerging disease. A westernized lifestyle or habits and some environmental factors have been found to contribute to the pathogenesis of IBD. The relevant risk factors include Smoking, hygiene hypothesis, microorganisms, appendectomy, medication, nutrition, and stress have all been found to be associated with the modality of IBD, but results are inconsistent on this issue in available studies. Therefore, more studies are required to identify and understand the environmental determinants of IBD.

13.
Mol Ther ; 21(7): 1345-57, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23752315

RESUMEN

Food-derived exosome-like nanoparticles pass through the intestinal tract throughout our lives, but little is known about their impact or function. Here, as a proof of concept, we show that the cells targeted by grape exosome-like nanoparticles (GELNs) are intestinal stem cells whose responses underlie the GELN-mediated intestinal tissue remodeling and protection against dextran sulfate sodium (DSS)-induced colitis. This finding is further supported by the fact that coculturing of crypt or sorted Lgr5⁺ stem cells with GELNs markedly improved organoid formation. GELN lipids play a role in induction of Lgr5⁺ stem cells, and the liposome-like nanoparticles (LLNs) assembled with lipids from GELNs are required for in vivo targeting of intestinal stem cells. Blocking ß-catenin-mediated signaling pathways of GELN recipient cells attenuates the production of Lgr5⁺ stem cells. Thus, GELNs not only modulate intestinal tissue renewal processes, but can participate in the remodeling of it in response to pathological triggers.


Asunto(s)
Colitis/inducido químicamente , Colitis/prevención & control , Sulfato de Dextran/toxicidad , Intestinos/citología , Nanopartículas/uso terapéutico , Células Madre/citología , Vitis/química , Animales , Masculino , Ratones
15.
Monoclon Antib Immunodiagn Immunother ; 32(1): 16-20, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23600500

RESUMEN

CD13 (CD13/aminopeptidase N, APN, or CD13/APN) is a widely expressed type II membrane-bound metalloprotease. It is often overexpressed on cancer cells and expressed on CD4(+)CD25(hi) Treg cell subpopulation with higher suppressive ability. It has been determined to be a promising target in cancer diagnosis and therapy. In this study, a functional anti-human CD13 monoclonal antibody, MAb 9E4, was obtained and the specificity of this MAb was verified by flow cytometry. This MAb effectively recognized the CD13 molecule expressed on a series of malignant cell lines. Furthermore, we demonstrated that MAb 9E4 suppresses the suppressive function of Treg cells. This functional anti-human CD13 MAb provides a valuable tool for further study targeting the CD13 positive Treg cells.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos CD13/inmunología , Regulación de la Expresión Génica/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos BALB C , Factor de Crecimiento Transformador beta/metabolismo
16.
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
17.
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
18.
PLoS One ; 7(12): e50781, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23284647

RESUMEN

Until now the essential transcription factor that determines the epithelial phenotype of breast cancer has not been identified and its role in epithelial-to-mesenchymal transition (EMT) and tumor progression remain unclear. Here, by analyzing large expression profiles of human breast cancer cells, we found an extraordinary correlation between the expression of Grainyhead transcription factor Grhl2 and epithelial marker E-cadherin. Knockdown of Grhl2 expression by shRNA in human mammary epithelial cell MCF10A leads to down-regulation of E-cadherin and EMT. Grhl2 is down-regulated in disseminated cancer cells that have undergone EMT, and over-expression of Grhl2 is sufficient to induce epithelial gene expression. Large clinical datasets reveal that expression of Grhl2 is significantly associated with poor relapse free survival and increased risk of metastasis in breast cancer patients. In mouse models, over-expression of Grhl2 significantly promotes tumor growth and metastasis. Further testing of several Grhl2 regulated genes leads to the same conclusions that the tumorigenic and metastatic potentials of tumor cells are linked to epithelial phenotype but not mesenchymal phenotype. In conclusion, our findings indicate that Grhl2 plays an essential role in the determination of epithelial phenotype of breast cancers, EMT and tumor progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Glándulas Mamarias Humanas/patología , Fenotipo , Factores de Transcripción/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Cadherinas/metabolismo , Línea Celular Tumoral , Supervivencia sin Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Glándulas Mamarias Humanas/metabolismo , Ratones , Metástasis de la Neoplasia , Especificidad de Órganos , Isoformas de Proteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Wnt/metabolismo
19.
Am J Pathol ; 180(1): 390-8, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22067905

RESUMEN

Exosomes participate in intercellular communication, but most data published are based on exosomes released from in vitro cultured cells that do not communicate with neighboring cells located in the same microenvironment as the exosomal-producing cells in vivo. In this study, our data show that co-culture of leukocytes isolated from breast tumor tissue leads to uptake of fibronectin (FN) on or in the tumor exosomes (Exo(fib+)). The induction of FN and exosomal uptake is tumor tissue derived and leukocyte specific, because leukocytes isolated from the peripheral blood of naïve mice failed to induce FN uptake by tumor exosomes. Furthermore, depletion of both CD25(+) cells and Gr-1(+) cells from tumor-associated leukocytes causes a reduction of Exo(fib+), suggesting that tumor-associated CD25(+) cells and Gr-1(+) cells participate in FN production and uptake by tumor exosomes, resulting in Exo(fib+). As a result of tumor cells absorbing Exo(fib+), two major events are induced: focal adhesion kinase/Src-dependent signaling pathways are activated, and the production of proinflammatory cytokines and metalloproteinase 9 is enhanced in response to absorbing exosomes. This, in turn, enhances tumor cell invasion in vitro and in vivo. Collectively, our findings provide evidence that exosomes released from freshly excised tumor tissue cells that have communicated/interacted with immune cells gain new immune evasion capacity.


Asunto(s)
Comunicación Celular/fisiología , Neoplasias del Colon/fisiopatología , Exosomas/fisiología , Fibronectinas/metabolismo , Leucocitos/fisiología , Animales , Neoplasias de la Mama/fisiopatología , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Neoplasias Pulmonares/fisiopatología , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica/fisiopatología , Metástasis de la Neoplasia , Trasplante de Neoplasias , Microambiente Tumoral/fisiología
20.
Mol Ther ; 19(10): 1769-79, 2011 10.
Artículo en Inglés | MEDLINE | ID: mdl-21915101

RESUMEN

In this study, exosomes used to encapsulate curcumin (Exo-cur) or a signal transducer and activator of transcription 3 (Stat3) inhibitor, i.e., JSI124 (Exo-JSI124) were delivered noninvasively to microglia cells via an intranasal route. The results generated from three inflammation-mediated disease models, i.e., a lipopolysaccharide (LPS)-induced brain inflammation model, experimental autoimmune encephalitis and a GL26 brain tumor model, showed that mice treated intranasally with Exo-cur or Exo-JSI124 are protected from LPS-induced brain inflammation, the progression of myelin oligodendrocyte glycoprotein (MOG) peptide induced experimental autoimmune encephalomyelitis (EAE), and had significantly delayed brain tumor growth in the GL26 tumor model. Intranasal administration of Exo-cur or Exo-JSI124 led to rapid delivery of exosome encapsulated drug to the brain that was selectively taken up by microglial cells, and subsequently induced apoptosis of microglial cells. Our results demonstrate that this strategy may provide a noninvasive and novel therapeutic approach for treating brain inflammatory-related diseases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Portadores de Fármacos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Exosomas , Administración Intranasal , Animales , Antiinflamatorios/administración & dosificación , Neoplasias Encefálicas/patología , Humanos , Ratones , Microglía/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA