Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 79(10): 537, 2022 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-36183290

RESUMEN

Contact-based pericellular interactions play important roles in cancer progression via juxtacrine signaling pathways. The present study revealed that hypoxia-inducible factor-1α (HIF-1α), induced even in non-hypoxic conditions by cell-to-cell contact, was a critical cue responsible for the malignant characteristics of glioblastoma multiforme (GBM) cells through Notch1 signaling. Densely cultured GBM cells showed enhanced viability and resistance to temozolomide (TMZ) compared to GBM cells at a low density. Ablating Notch1 signaling by a γ-secretase inhibitor or siRNA transfection resensitized resistant GBM cells to TMZ treatment and decreased their viability under dense culture conditions. The expression of HIF-1α was significantly elevated in highly dense GBM cells even under non-hypoxic conditions. Atypical HIF-1α expression was associated with the Notch1 signaling pathway in both GBM and glioblastoma stem cells (GSC). Proteasomal degradation of HIF-1α was prevented by binding with Notch1 intracellular domain (NICD), which translocated to the nuclei of GBM cells. Silencing Notch1 signaling using a doxycycline-inducible Notch1 RNA-interfering system or treatment with chetomin, a HIF pathway inhibitor, retarded tumor development with a significant anti-cancer effect in a murine U251-xenograft model. Using GBM patient tissue microarray analysis, a significant increase in HIF-1α expression was identified in the group with Notch1 expression compared to the group without Notch1 expression among those with positive HIF-1α expression. Collectively, these findings highlight the critical role of cell-to-cell contact-dependent signaling in GBM progression. They provide a rationale for targeting HIF-1α signaling even in a non-hypoxic microenvironment.


Asunto(s)
Glioblastoma , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Línea Celular Tumoral , Doxiciclina , Glioblastoma/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , ARN Interferente Pequeño/genética , Receptor Notch1/genética , Transducción de Señal , Temozolomida , Microambiente Tumoral
2.
Am J Obstet Gynecol ; 221(5): 502.e1-502.e12, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31207235

RESUMEN

BACKGROUND: During pregnancy, feto-maternal communication can be mediated through extracellular vesicles, specifically exosomes, 30- to 150-nm particles released from each cell. Exosomes carry cellular signals, and traffic between fetal and maternal tissues to produce functional changes in recipient cells. Exosomes may function as a biomarker indicative of the physiologic status of their tissue of origin. These properties of exosomes during pregnancy are not well studied. OBJECTIVE: To test exosome trafficking and function, we used a transgenic mouse model containing membrane-targeted, red fluorescent protein tdTomato and enhanced green fluorescent protein cyclic recombinase-reporter construct expressed only in fetal tissues. This model allows fetal tissues and their exosomes to express tdTomato under normal conditions or green fluorescent protein if fetal tissues are exposed to cyclic recombinase that will excise tdTomato. As maternal tissue remains negative for this construct, tdTomato/green fluorescent protein expression and their switching can be used to determine fetal-specific cell and exosome trafficking. MATERIALS AND METHODS: tdTomato/green fluorescent protein-homozygous male mice were mated with wild-type females to have all fetal tissues express the tdTomato/green fluorescent protein allele. Red fluorescence due to tdTomato expression of the tdTomato/green fluorescent protein allele in fetal tissues (placenta, fetal membranes) was confirmed by confocal microscopy on embryonic day 16. Localization of fetal exosomes in maternal uterine tissues were performed by immunostaining for exosome marker CD81 and tdTomato expression followed by confocal microscopy. Fetal exosomes (tdTomato-positive) in maternal plasma were immunoprecipitated using anti-red fluorescent protein tdTomato, followed by confirmation with flow cytometry. To further illustrate the fidelity of fetal exosomes in maternal samples, exosomes bioengineered to contain cyclic recombinase (1.0 × 1010 exosomes) were injected intraperitoneally on embryonic day 13. On embryonic day 16, fetal (placenta and fetal membranes) tissues were imaged to show tdTomato-to-green fluorescent protein transition. The green fluorescent protein-expressing exomes were localized in maternal tissues (confocal microscopy) and plasma (flow cytometry). RESULTS: Mating between a male with the tdTomato/green fluorescent protein construct and a null female resulted in fetal tissues and their exosomes expressing tdTomato positivity. Total fetal exosomes in maternal plasma was about 35%. tdTomato-positive exosomes were isolated from maternal plasma and immunostaining localized tdTomato-positive exosomes in maternal uterine tissues. Maternal intraperitoneal injection of cyclic recombinase-enriched exosomes crossed placenta, excised tdTomato from the tdTomato/green fluorescent protein construct in the fetal tissues, and caused green fluorescent protein expression in fetal cells. Furthermore, green fluorescent protein-positive exosomes released from fetal cells were isolated from maternal blood. CONCLUSION: In this pilot study, we report feto-maternal and maternal-fetal trafficking of exosomes indicative of paracrine signaling during pregnancy. Exosomes from the maternal side can produce functional changes in fetal tissues. Trafficking of exosomes suggests their potential role in pregnancy as biomarkers of fetal functions and usefulness as a carrier of drugs and other cargo to the fetal side during pregnancy. Isolation and characterization of fetal exosomes can advance fetal research without performing invasive procedures.


Asunto(s)
Exosomas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Luminiscentes/metabolismo , Comunicación Paracrina/fisiología , Animales , Biomarcadores/metabolismo , Membranas Extraembrionarias/metabolismo , Femenino , Citometría de Flujo , Masculino , Ratones Transgénicos , Microscopía Confocal , Modelos Animales , Proyectos Piloto , Placenta/metabolismo , Embarazo , Útero/metabolismo , Proteína Fluorescente Roja
3.
Cell Physiol Biochem ; 48(3): 1332-1346, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30048972

RESUMEN

BACKGROUND/AIMS: Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. The defining characteristics of GBM are diffuse infiltration of tumor cells into normal brain parenchyma, rapid growth, a high degree of infiltration of microglia and macrophages, and the presence of necrosis. Microglia/macrophages are frequently found in gliomas and they extensively infiltrate GBM tissue, up to 30% of total tumor mass. However, little is known about the effect of necrotic cells (NCs) on microglia infiltration in GBM and the tumor-infiltrating microglia-induced factors in GBMs. METHODS: In this study, to address whether necrosis or necrosis-exposed GBM cells affect the degree of microglia/macrophage infiltration, migration and invasion/infiltration assays were performed. Culture supernatants and nuclear extracts of CRT-MG cells treated or untreated with necrotic cells were analyzed using a chemokine array and electrophoretic mobility shift assay, respectively. RESULTS: The presence of NCs promoted the migration/infiltration of microglia, and GBM cell line CRT-MG cells exposed to NCs further enhanced the migration and infiltration of HMO6 microglial cells. Treatment with NCs induced mRNA and protein expression of chemokines such as Monocyte Chemoattractant Protein-1 (CCL2/MCP-1) and Macrophage Inflammatory Protein-3α (CCL20/MIP-3α) in CRT-MG cells. In particular, CCL2/MCP-1 and CCL20/MIP-3α were significantly increased in NC-treated CRT-MG cells. NCs induced DNA binding of the transcription factors Nuclear Factor (NF)-κB and Activator Protein 1 (AP-1) to the CCL2/MCP-1 and CCL20/MIP-3α promoters, leading to increased CCL2/MCP-1 and CCL20/MIP-3α mRNA and protein expression in CRT-MG cells. CONCLUSION: These results provide evidence that NCs induce the expression of CCL2/MCP-1 and CCL20/MIP-3α in glioblastoma cells through activation of NF-κB and AP-1 and facilitate the infiltration of microglia into tumor tissues.


Asunto(s)
Neoplasias Encefálicas/patología , Quimiocina CCL20/metabolismo , Quimiocina CCL2/metabolismo , Glioblastoma/patología , Microglía/patología , Necrosis/patología , Invasividad Neoplásica/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Quimiocina CCL2/análisis , Quimiocina CCL2/genética , Quimiocina CCL20/análisis , Quimiocina CCL20/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Microglía/metabolismo , Necrosis/genética , Necrosis/metabolismo , Invasividad Neoplásica/genética , ARN Mensajero/análisis , ARN Mensajero/genética
4.
Biochem Biophys Res Commun ; 470(4): 924-9, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26820528

RESUMEN

Ginsenosides, the major bio-active ingredients included in Panax ginseng, have been known for the hair growth activity and used to treat patients who suffer from hair loss; however, the detailed mechanisms of this action are still largely unknown. This study was conducted to investigate the molecular and cellular mechanisms responsible for hair growth promoting effect of ginsenoside Re (GRe) in vitro and in vivo. Different doses of minoxidil and GRe were administered topically to the back regions of nude mice for up to 45 days, and hair shaft length and hair cycles were determined for hair promoting activities. Topical treatment of GRe significantly increased the hair shaft length and hair existent time, which was comparable to the action of minoxidil. We also demonstrated that GRe stimulated hair shaft elongation in the ex vivo cultures of vibrissa hair follicles isolated from C57BL/6 mouse. Systemic transcriptome analysis by next generation sequencing demonstrated that TGF-ß-pathway related genes were selectively down-regulated by treatment of GRe in vivo, and the same treatment suppressed TGF-ß-induced phosphorylation of ERK in HeLa cells. The results clearly indicated that GRe is the effective constituent in the ginseng on hair promotion via selective inhibition of the hair growth phase transition related signaling pathways, TGF-ß signaling cascades.


Asunto(s)
Ginsenósidos/administración & dosificación , Folículo Piloso/efectos de los fármacos , Folículo Piloso/crecimiento & desarrollo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Triterpenos/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Células HeLa , Humanos , Ratones , Ratones Desnudos , Transducción de Señal/efectos de los fármacos , Proteína smad3/metabolismo
5.
J Neurooncol ; 128(1): 29-34, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26951556

RESUMEN

Glioblastoma multiforme (GBM) is the most common and aggressive type of primary brain tumor with grave prognosis. Despite the growing understanding of the complex signaling networks responsible for the initiation and progression of GBM, many experimental therapies have fallen short of their treatment goals. In the present study, we investigated the novel molecular mechanisms responsible for synergistic action of temozolomide (TMZ) and anti-VEGF therapy in GBM cells. We tested the combined effects of TMZ and VEGF blockade in four human GBM cell lines: TMZ-sensitive U251-MG and U373-MG cells, and TMZ-resistant CRT-MG and LN215-MG cells, which correlated with MGMT promoter methylation status. Treatment of TMZ along with a sublethal dosage range of SU1498, a chemical inhibitor of the VEGF receptor signaling, induced significant cell death in both TMZ-sensitive and TMZ-resistant GBM cells without changing the status of the MGMT promoter methylation. Treatment with TMZ specifically reduced the expression of NRP-1, a coreceptor of VEGF but not those of VEGF-R1 and VEGF-R2. We further confirmed the key role of NRP-1 by showing that the reduction of NRP-1 by siRNA also increased the SU1498-induced cytotoxicity of LN215-MG. These results collectively indicate that combined treatment of TMZ can sensitize GBM cells to blockade of autocrine VEGF signaling through specific down-regulation of NRP-1, which provide a rationale for further evaluation and a potential clinical trial of combinatorial therapy of TMZ and SU1498 or other VEGF inhibitors for intractable brain tumors.


Asunto(s)
Antineoplásicos/farmacología , Cinamatos/farmacología , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Neuropilina-1/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Metilación de ADN , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Dacarbazina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Neuropilina-1/genética , Neuropilina-1/metabolismo , Regiones Promotoras Genéticas , ARN Interferente Pequeño , Temozolomida , Proteínas Supresoras de Tumor/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
6.
Hepatology ; 56(3): 831-40, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22430873

RESUMEN

UNLABELLED: Hepatitis C virus (HCV) infection results in liver injury and long-term complications, such as liver cirrhosis and hepatocellular carcinoma. Liver injury in HCV infection is believed to be caused by host immune responses, not by viral cytopathic effects. Tumor necrosis factor-alpha (TNF-α) plays a pivotal role in the inflammatory processes of hepatitis C. TNF-α induces cell death that can be ameliorated by nuclear factor kappaB (NF-κB) activation. We investigated the regulation of TNF-α signal transduction in HCV-infected cells and identified HCV proteins responsible for sensitization to TNF-α-induced cell death. We studied the effect of HCV infection on TNF-α signal transduction using an in vitro HCV infection model (JFH-1, genotype 2a) with Huh-7 and Huh-7.5 cells. We found that TNF-α-induced cell death significantly increased in HCV-infected cells. HCV infection diminished TNF-α-induced phosphorylation of IκB kinase (IKK) and inhibitor of NF-κB (IκB), which are upstream regulators of NF-κB activation. HCV infection also inhibited nuclear translocation of NF-κB and expression of NF-κB-dependent anti-apoptotic proteins, such as B-cell lymphoma--extra large (Bcl-xL), X-linked inhibitor of apoptosis protein (XIAP), and the long form of cellular-FLICE inhibitory protein (c-FLIP). Decreased levels of Bcl-xL, XIAP, and c-FLIP messenger RNA and protein were also observed in livers with chronic hepatitis C. Transfection with plasmids encoding each HCV protein revealed that core, nonstructural protein (NS)4B, and NS5B attenuated TNF-α-induced NF-κB activation and enhanced TNF-α-induced cell death. CONCLUSION: HCV infection enhances TNF-α-induced cell death by suppressing NF-κB activation through the action of core, NS4B, and NS5B. This mechanism may contribute to immune-mediated liver injury in HCV infection.


Asunto(s)
Muerte Celular/fisiología , Hepatitis C/inmunología , FN-kappa B/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Células Cultivadas , Humanos
7.
J Cell Physiol ; 227(8): 3027-35, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22020994

RESUMEN

Bcl-2/adenovirus E1B 19-kDa interacting protein 1 (BNIP1), which is predominantly localized to the endoplasmic reticulum (ER), is a pro-apoptotic Bcl-2 homology domain 3 (BH3)-only protein. Here, we show that the expression of BNIP1 induced not only a highly interconnected ER network but also mitochondrial fragmentation in a BH3 domain-dependent manner. Functional analysis demonstrated that BNIP1 expression increased dynamin-related protein 1 (Drp1) expression followed by the mitochondrial translocation of Drp1 and subsequent mitochondrial fission. Both BNIP1-induced mitochondrial fission and the translocation of Drp1 were abrogated by Bcl-2 overexpression. These results collectively indicate that ER-specific BNIP1 plays an important role in mitochondrial dynamics by modulating the mitochondrial fission protein Drp1 in a BH3 domain-dependent fashion.


Asunto(s)
Retículo Endoplásmico/metabolismo , GTP Fosfohidrolasas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Apoptosis , Células COS , Chlorocebus aethiops , Dinaminas , GTP Fosfohidrolasas/genética , Regulación de la Expresión Génica , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias/fisiología , Proteínas Mitocondriales/genética , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/genética , Especies Reactivas de Oxígeno/metabolismo
8.
Mol Vis ; 18: 1010-20, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22550395

RESUMEN

PURPOSE: Transforming growth factor-ß (TGF-ß) plays a key role in transforming retinal pigment epithelial (RPE) cells into mesenchymal fibroblastic cells, which are implicated in proliferative vitreoretinopathy. Herein, we tested the effect of pirfenidone, a novel antifibrotic agent, on TGF-ß1-mediated fibrogenesis in the human RPE cell line ARPE-19. METHODS: The effect of pirfenidone on the TGF-ß1-induced phenotype in ARPE-19 cells was measured with immunocytochemistry as the change in F-actin. Fibronectin and collagen production was measured with enzyme-linked immunosorbent assay, and cell migration activity was investigated using a scratch assay. Immunoblot analyses of cofilin, sma and mad protein (smad) 2/3, p38 mitogen-activated protein kinase, c-Jun N-terminal kinase, and extracellular signal-related kinase expression were conducted to elucidate the cell signaling networks that contribute to the antifibrotic effect of pirfenidone. RESULTS: Treatment with TGF-ß1 induced typical phenotypic changes such as formation of stress fiber running parallel to the long axis of cells and enhanced migration and production of extracellular matrix components such as collagen type I and fibronectin. This fibroblast-like phenotype induced by TGF-ß1 was significantly inhibited by pretreatment with pirfenidone in a dose-dependent manner. We also elucidated the TGF-ß signaling pathways as the target of the inhibitory effect of pirfenidone. Pirfenidone inhibited TGF-ß signaling by preventing nuclear accumulation of active Smad2/3 complexes rather than phosphorylation of Smad2/3. CONCLUSIONS: These results collectively provide a rational background for future evaluation of pirfenidone as a potential antifibrotic agent for treating proliferative vitreoretinopathy and other fibrotic retinal disorders.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Células Epiteliales/efectos de los fármacos , Piridonas/farmacología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Actinas/genética , Actinas/metabolismo , Diferenciación Celular , Línea Celular , Movimiento Celular , Colágeno/genética , Colágeno/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrosis/prevención & control , Expresión Génica/efectos de los fármacos , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Transporte de Proteínas/efectos de los fármacos , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Smad/genética , Factor de Crecimiento Transformador beta1/farmacología , Vitreorretinopatía Proliferativa/tratamiento farmacológico
9.
Biomed Microdevices ; 14(6): 1141-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22821236

RESUMEN

Since most of the bioavailable drugs are impermeable through the blood-brain barrier (BBB), development of a rapid and reliable permeability assay system has been a challenge in drug discovery targeting central nervous system (CNS). Here, we designed a microfluidic device to monitor the drug permeability into the CNS. Human umbilical vein endothelial cells (HUVECs) were shortly (2 ~ 3 h) incubated with astrocyte-conditioned medium after being trapped on microholes in the microfluidic device and tested for chip-based permeability measurement of drugs. The measured permeability values were highly correlated with those measured by conventional in vitro methods and the brain uptake index representing the quantity of transported substances across the in vivo BBB of rats. Using the microfluidic device, we could easily monitor the effect of hydrogen peroxide on the trans-endothelial permeability, which are consistent with the finding that the same treatment disrupted the formation of tight junctions between endothelial cells. Considering relatively short period of time needed for endothelial cell culture and ability to monitor the BBB physiology continuously, we propose that this novel system can be used as an invaluable first-line tool for CNS-related drug development.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/irrigación sanguínea , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Técnicas Analíticas Microfluídicas/instrumentación , Animales , Astrocitos/citología , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/citología , Células Cultivadas , Cromatografía Líquida de Alta Presión , Medios de Cultivo Condicionados , Diseño de Equipo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Técnicas Analíticas Microfluídicas/métodos , Permeabilidad , Preparaciones Farmacéuticas , Farmacocinética , Ratas , Reproducibilidad de los Resultados , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo
10.
Pharmaceutics ; 14(3)2022 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-35336049

RESUMEN

Delivering therapeutics to the central nervous system (CNS) is difficult because of the blood-brain barrier (BBB). Therapeutic delivery across the tight junctions of the BBB can be achieved through various endogenous transportation mechanisms. Receptor-mediated transcytosis (RMT) is one of the most widely investigated and used methods. Drugs can hijack RMT by expressing specific ligands that bind to receptors mediating transcytosis, such as the transferrin receptor (TfR), low-density lipoprotein receptor (LDLR), and insulin receptor (INSR). Cell-penetrating peptides and viral components originating from neurotropic viruses can also be utilized for the efficient BBB crossing of therapeutics. Exosomes, or small extracellular vesicles, have gained attention as natural nanoparticles for treating CNS diseases, owing to their potential for natural BBB crossing and broad surface engineering capability. RMT-mediated transport of exosomes expressing ligands such as LDLR-targeting apolipoprotein B has shown promising results. Although surface-modified exosomes possessing brain targetability have shown enhanced CNS delivery in preclinical studies, the successful development of clinically approved exosome therapeutics for CNS diseases requires the establishment of quantitative and qualitative methods for monitoring exosomal delivery to the brain parenchyma in vivo as well as elucidation of the mechanisms underlying the BBB crossing of surface-modified exosomes.

11.
Int J Oncol ; 61(4)2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36043525

RESUMEN

Despite the high expression of neuropilin­1 (NRP­1) in human glioblastoma (GB), the understanding of its function as a co­receptor of vascular endothelial growth factor receptors (VEGFRs) in angiogenesis is currently limited. Therefore, the aim of the present study was to elucidate the non­classical function of NRP­1 expression in human GB. Expression patterns of NRP­1 and VEGF­A were determined by sandwich ELISA, western blot analysis, or immunohistochemistry. Differential dependency of GB cells following ablation of VEGF­A signaling was validated in vitro and in vivo. Cellular mechanism responsible for distinct response to VEGF­A signaling was evaluated by western blotting and immunoprecipitation analysis. Prognostic implications were assessed using IHC analysis. GB cells exhibited differing sensitivity to silencing of vascular endothelial growth factor (VEGF)­A signaling, which resulted in a distinct expression pattern of wild­type or chondroitin­sulfated NRP­1. VEGF­A­sensitive GB exhibited the physical interaction between wild­type NRP­1 and FMS related receptor tyrosine kinase 1 (Flt­1) whereas VEGF­A­resistant GB exhibited chondroitin­sulfated NRP­1 without interaction with Flt­1. Eliminating the chondroitin sulfate modification in NRP­1 led to re­sensitization to VEGF­A signaling, and chondroitin sulfate modification was found to be associated with an adverse prognosis in patients with GB. The present study identified the distinct functions of NRP­1 in VEGF­A signaling in accordance with its unique expression type and interaction with Flt­1. The present research is expected to provide a strong basis for targeting VEGF­A signaling in patients with GB, with variable responses.


Asunto(s)
Glioblastoma , Neuropilina-1 , Factor A de Crecimiento Endotelial Vascular , Sulfatos de Condroitina , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Neuropilina-1/genética , Neuropilina-1/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Microvasc Res ; 82(2): 141-6, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21570413

RESUMEN

Because of high spatial resolution and superior tissue penetration, a femtosecond laser of the near-infrared spectrum has great potential to improve the efficacy of conventional photodynamic therapy; however, the lack of suitable photosensitizers has so far limited its bedside applications. Recently, our group reported that a brief irradiation by femtosecond lasers in the absence of exogenous probes can modulate various cellular behaviors in vitro and in vivo. Here, we demonstrate that targeted irradiation by a femtosecond laser disrupted tumor-associated blood vessels, and the inhibition of vascular endothelial growth factor signaling augmented the efficacy of laser-induced angiolysis. Further, we show that reactive oxygen species (ROS) are generated in response to laser irradiation, and reducing the intracellular levels of ROS rendered endothelial cells resistant to laser-induced cytotoxicity. Collectively, these results indicate that a femtosecond laser can be used as a vascular-disrupting therapeutic modality for cancer treatment, especially when used in combination with conventional anti-angiogenic therapies.


Asunto(s)
Neoplasias/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Vasos Sanguíneos/patología , Línea Celular Tumoral , Humanos , Rayos Láser , Masculino , Ratones , Ratones Desnudos , Modelos Biológicos , Neoplasias/terapia , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/química , Especies Reactivas de Oxígeno , Venas Umbilicales/citología
13.
Sci Adv ; 7(4)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33523942

RESUMEN

Accumulation of immune cells and activation of the pro-inflammatory transcription factor NF-κB in feto-maternal uterine tissues is a key feature of preterm birth (PTB) pathophysiology. Reduction of the fetal inflammatory response and NF-κB activation are key strategies to minimize infection-associated PTB. Therefore, we engineered extracellular vesicles (exosomes) to contain an NF-κB inhibitor, termed super-repressor (SR) IκBα. Treatment with SR exosomes (1 × 1010 per intraperitoneal injection) after lipopolysaccharide (LPS) challenge on gestation day 15 (E15) prolonged gestation by over 24 hours (PTB ≤ E18.5) and reduced maternal inflammation (n ≥ 4). Furthermore, using a transgenic model in which fetal tissues express the red fluorescent protein tdTomato while maternal tissues do not, we report that LPS-induced PTB in mice is associated with influx of fetal innate immune cells, not maternal, into feto-maternal uterine tissues. SR packaged in exosomes provides a stable and specific intervention for reducing the inflammatory response associated with PTB.


Asunto(s)
Lipopolisacáridos , Nacimiento Prematuro , Animales , Modelos Animales de Enfermedad , Femenino , Feto , Humanos , Recién Nacido , Inflamación/metabolismo , Lipopolisacáridos/efectos adversos , Ratones , FN-kappa B/metabolismo , Embarazo , Nacimiento Prematuro/metabolismo , Útero/metabolismo
14.
Biochem Biophys Res Commun ; 391(1): 33-7, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19895789

RESUMEN

Mutant ubiquitin found in neurodegenerative diseases has been thought to hamper activation of transcription factor nuclear factor-kappa B (NF-kappaB) by inhibiting ubiquitin-proteasome system (UPS). It has been reported that ubiquitin also is involved in signal transduction in an UPS-independent manner. We used a modeling and simulation approach to delineate the roles of ubiquitin on NF-kappaB activation. Inhibition of proteasome complex increased maximal activation of IKK mainly by decreasing the UPS efficiency. On the contrary, mutant ubiquitin decreased maximal activity of IKK. Computational modeling showed that the inhibition effect of mutant ubiquitin is mainly attributed to decreased activity of UPS-independent function of ubiquitin. Collectively, our results suggest that mutant ubiquitin affects NF-kappaB activation in an UPS-independent manner.


Asunto(s)
Simulación por Computador , Modelos Biológicos , FN-kappa B/metabolismo , Ubiquitina/metabolismo , Ubiquitinación , Activación Enzimática , Humanos , Quinasa I-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Ubiquitina/genética
15.
Sci Adv ; 6(15): eaaz6980, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32285005

RESUMEN

As extracellular vesicles that play an active role in intercellular communication by transferring cellular materials to recipient cells, exosomes offer great potential as a natural therapeutic drug delivery vehicle. The inflammatory responses in various disease models can be attenuated through introduction of super-repressor IκB (srIκB), which is the dominant active form of IκBα and can inhibit translocation of nuclear factor κB into the nucleus. An optogenetically engineered exosome system (EXPLOR) that we previously developed was implemented for loading a large amount of srIκB into exosomes. We showed that intraperitoneal injection of purified srIκB-loaded exosomes (Exo-srIκBs) attenuates mortality and systemic inflammation in septic mouse models. In a biodistribution study, Exo-srIκBs were observed mainly in the neutrophils, and in monocytes to a lesser extent, in the spleens and livers of mice. Moreover, we found that Exo-srIκB alleviates inflammatory responses in monocytic THP-1 cells and human umbilical vein endothelial cells.


Asunto(s)
Exosomas/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , Sepsis/metabolismo , Sepsis/patología , Animales , Modelos Animales de Enfermedad , Exosomas/ultraestructura , Lipopolisacáridos/efectos adversos , Ratones , Mortalidad , Inhibidor NF-kappaB alfa/administración & dosificación , FN-kappa B/metabolismo , Sustancias Protectoras/administración & dosificación , Sepsis/tratamiento farmacológico , Sepsis/etiología , Transducción de Señal , Distribución Tisular
16.
Macromol Rapid Commun ; 30(14): 1232-7, 2009 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-21638375

RESUMEN

A new method to tune both phosphorescence emission intensity and spectroscopic colors based on the alternatively structured host/guest multilayer thin films prepared by the spin-assisted LbL deposition is presented. Their emission characteristics are demonstrated with pairs of positively charged Ir-PEI complexes as guests and negatively charged CBZ-PAA as hosts. The phosphorescent emission of Ir-PEI complexes is enhanced by the energy transfer from the host to the guest and, additionally, this energy transfer can be finely tuned by the insertion of spacer layers between the phosphorescent donor and acceptor layers to vary the emission intensity as well as to render different emission colors.

17.
J Neurochem ; 105(2): 557-64, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18088368

RESUMEN

Caffeic acid phenethyl ester (CAPE), an active component of propolis extracts, has been known for its specific inhibition of nuclear factor kappaB (NF-kappaB) and subsequent anti-inflammatory activity. In this study, we report that (i) CAPE exerts its anti-inflammatory action (inhibition of tumor necrosis factor-induced expression of intercellular adhesion molecule-1 and CC chemokine ligand-2) via NF-kappaB inhibition by two distinct molecular mechanisms in a cell-specific manner: CAPE inhibited downstream pathways of inhibitor kappaB (IkappaB) degradation in monocytic cells, while activation of upstream IkappaB kinase was suppressed by CAPE pre-treatment in astroglial cells; and (ii) CAPE paradoxically activates the c-Jun N-terminal kinase (JNK) pathway, which might be responsible for its pro-apoptotic action and divergent regulation of proinflammatory mediators such as CXC chemokine ligand-8.


Asunto(s)
Astrocitos/efectos de los fármacos , Ácidos Cafeicos/farmacología , Inhibidores Enzimáticos/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Monocitos/efectos de los fármacos , FN-kappa B/metabolismo , Alcohol Feniletílico/análogos & derivados , Transducción de Señal/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Activación Enzimática/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática/métodos , Feto , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Modelos Biológicos , Alcohol Feniletílico/farmacología , Factor de Necrosis Tumoral alfa/farmacología
18.
Biochem Biophys Res Commun ; 372(4): 870-4, 2008 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-18534188

RESUMEN

The central nervous system (CNS) is particularly vulnerable to reactive oxygen species (ROS), which have been implicated in the pathogenesis of various neurological disorders. The TNF superfamily of cytokines, especially tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), induces caspase-dependent cell death and is also implicated in various neurodegenerative diseases. In this study, we investigated the relationship between ROS and TRAIL-induced cell death. Exposure to hydrogen peroxide (H(2)O(2)) (100 microM) sensitized human astrocytic cells to TRAIL-induced cell death (up to 7-fold induction). To delineate the molecular mechanisms responsible for H(2)O(2)-induced sensitization, we examined expression of various genes (Caspase-8, Fas, FasL, DR4, DR5, DcR1, DcR2, TRAIL, TNFRp55) related to TRAIL-induced cell death. Treatment with H(2)O(2) significantly increased DR5 mRNA and protein expression in a time- and dose-dependent manner. H(2)O(2)-mediated cell death was blocked upon treatment with DR5:Fc protein, a TRAIL-specific antagonistic protein. These findings collectively suggest that oxidative stress sensitizes human astroglial cells to TRAIL-induced cell death through up-regulation of DR5 expression.


Asunto(s)
Apoptosis , Astrocitos/metabolismo , Estrés Oxidativo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Apoptosis/genética , Astrocitos/citología , Astrocitos/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/toxicidad , ARN Mensajero/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Regulación hacia Arriba
19.
Immunol Lett ; 117(2): 168-73, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18339430

RESUMEN

Among tumor necrosis factor (TNF) superfamily, TNF-related apoptosis inducing ligand (TRAIL) along with TNF-alpha and FasL is known as death ligand due to its selective cytotoxicity against transformed tumor cells. TRAIL can also induce alternative angiogenic and/or proinflammatory signals other than apoptosis, however, the molecular mechanisms responsible for the alternative signals have not been detailed yet. Intercellular adhesion molecule-1 (ICAM-1) is thought to be involved in the processes of metastasis and angiogenesis in various tumors. We investigated the molecular mechanisms responsible for ICAM-1 expression by death ligands in human astroglial cells to delineate the alternative signals of these ligands. Here, we demonstrate that (1) death ligands induced expression of ICAM-1 at the mRNA and protein levels in human astroglial cells; (2) pre-treatment of z-VAD-fmk and/or SB202190 suppressed death ligand-induced ICAM-1 expression and subsequent adhesion of activated monocytic cells; and (3) inhibition of caspase suppressed death ligand-induced phosphorylation of p38 MAPK and IKK. These findings suggest biological function of death receptors other than apoptosis in human astroglial cells, and the involvement of caspase and/or p38 MAPK in alternative signaling through death receptors.


Asunto(s)
Astrocitos/metabolismo , Caspasas/metabolismo , Molécula 1 de Adhesión Intercelular/biosíntesis , Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Astrocitos/citología , Astrocitos/inmunología , Inhibidores de Caspasas , Caspasas/inmunología , Adhesión Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica , Humanos , Imidazoles/farmacología , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Monocitos/citología , Monocitos/inmunología , Monocitos/metabolismo , Piridinas/farmacología , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transfección , Células U937 , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
20.
FEBS Lett ; 581(24): 4691-6, 2007 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-17854800

RESUMEN

Impaired functioning of the proteasome pathway is one of the molecular mechanism underlying neurodegenerative changes in Alzheimer's disease. In this study, we report that dysfunction of the proteasome pathway in astroglial cells leads to decreased survival and dysregulation of chemokines by differential regulation of the nuclear factor kappa B and c-jun N-terminal kinase (JNK) pathways. We further demonstrated that proteasome inhibition augmented interleukin-1 beta- and tumor necrosis factor-alpha-induced activation of the IkappaBalpha kinase and MKK4/JNK/c-Jun pathway along with TAK1 activation. These results suggest that impaired function of the proteasome pathway may potentiate the immuno-pathologic role of secondarily activated astrocytes in the brain.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Interleucina-1beta/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Apoptosis/efectos de los fármacos , Astrocitos/citología , Caspasas/metabolismo , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Regulación de la Expresión Génica , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , FN-kappa B/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA