Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Adv Sci (Weinh) ; 11(15): e2305326, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38342616

RESUMEN

Botulinum neurotoxin serotype A (BoNT/A) is widely used in therapeutics and cosmetics. The effects of multi-dosed BoNT/A treatment are well documented on the peripheral nervous system (PNS), but much less is known on the central nervous system (CNS). Here, the mechanism of multi-dosed BoNT/A leading to CNS neurodegeneration is explored by using the 3D human neuron-glia model. BoNT/A treatment reduces acetylcholine, triggers astrocytic transforming growth factor beta, and upregulates C1q, C3, and C5 expression, inducing microglial proinflammation. The disintegration of the neuronal microtubules is escorted by microglial nitric oxide, interleukin 1ß, tumor necrosis factor α, and interleukin 8. The microglial proinflammation eventually causes synaptic impairment, phosphorylated tau (pTau) aggregation, and the loss of the BoNT/A-treated neurons. Taking a more holistic approach, the model will allow to assess therapeutics for the CNS neurodegeneration under the prolonged use of BoNT/A.


Asunto(s)
Microglía , Neuronas , Humanos , Microglía/metabolismo , Astrocitos
2.
Sci Rep ; 14(1): 744, 2024 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-38185738

RESUMEN

Aluminum oxide nanoparticle (AlNP), a ubiquitous neurotoxin highly enriched in air pollution, is often produced as an inevitable byproduct in the manufacturing of industrial products such as cosmetics and metal materials. Meanwhile, ALNP has emerged as a significant public health concern due to its potential association with neurological diseases. However, the studies about the neurotoxic effects of AlNP are limited, partially due to the lack of physiologically relevant human neurovascular unit with innate immunity (hNVUI). Here, we employed our AlNP-treated hNVUI model to investigate the underlying mechanism of AlNP-driven neurodegeneration. First, we validated the penetration of AlNP across a blood-brain barrier (BBB) compartment and found AlNP-derived endothelial cellular senescence through the p16 and p53/p21 pathways. Our study showed that BBB-penetrating AlNP promoted reactive astrocytes, which produced a significant level of reactive oxygen species (ROS). The astrocytic neurotoxic factors caused neuronal damage, including the synaptic impairment, the accumulation of phosphoric-tau proteins, and even neuronal death. Our study suggests that AlNP could be a potential environmental risk factor of neurological disorders mediated by neuroinflammation.


Asunto(s)
Contaminación del Aire , Síndromes de Neurotoxicidad , Humanos , Óxido de Aluminio/toxicidad , Barrera Hematoencefálica , Muerte Celular , Senescencia Celular
3.
Nat Protoc ; 18(9): 2838-2867, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37542184

RESUMEN

Neuroinflammation has either beneficial or detrimental effects, depending on risk factors and neuron-glia interactions in neurological disorders. However, studying neuroinflammation has been challenging due to the complexity of cell-cell interactions and lack of physio-pathologically relevant neuroinflammatory models. Here, we describe our three-dimensional microfluidic multicellular human neural culture model, referred to as a 'brain-on-a-chip' (BoC). This elucidates neuron-glia interactions in a controlled manner and recapitulates pathological signatures of the major neurological disorders: dementia, brain tumor and brain edema. This platform includes a chemotaxis module offering a week-long, stable chemo-gradient compared with the few hours in other chemotaxis models. Additionally, compared with conventional brain models cultured with mixed phenotypes of microglia, our BoC can separate the disease-associated microglia out of heterogeneous population and allow selective neuro-glial engagement in three dimensions. This provides benefits of interpreting the neuro-glia interactions while revealing that the prominent activation of innate immune cells is the risk factor leading to synaptic impairment and neuronal loss, validated in our BoC models of disorders. This protocol describes how to fabricate and implement our human BoC, manipulate in real time and perform end-point analyses. It takes 2 d to set up the device and cell preparations, 1-9 weeks to develop brain models under disease conditions and 2-3 d to carry out analyses. This protocol requires at least 1 month training for researchers with basic molecular biology techniques. Taken together, our human BoCs serve as reliable and valuable platforms to investigate pathological mechanisms involving neuroinflammation and to assess therapeutic strategies modulating neuroinflammation in neurological disorders.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedades Neuroinflamatorias , Humanos , Dispositivos Laboratorio en un Chip , Enfermedades Neuroinflamatorias/patología , Técnicas de Cultivo de Célula , Enfermedades Neurodegenerativas/patología
4.
Biomater Res ; 27(1): 71, 2023 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-37468961

RESUMEN

BACKGROUND: Glial scar formation is a reactive glial response confining injured regions in a central nervous system. However, it remains challenging to identify key factors formulating glial scar in response to glioblastoma (GBM) due to complex glia-GBM crosstalk. METHODS: Here, we constructed an astrocytic scar enclosing GBM in a human assembloid and a mouse xenograft model. GBM spheroids were preformed and then co-cultured with microglia and astrocytes in 3D Matrigel. For the xenograft model, U87-MG cells were subcutaneously injected to the Balb/C nude female mice. RESULTS: Additional glutamate was released from GBM-microglia assembloid by 3.2-folds compared to GBM alone. The glutamate upregulated astrocytic monoamine oxidase-B (MAO-B) activity and chondroitin sulfate proteoglycans (CSPGs) deposition, forming the astrocytic scar and restricting GBM growth. Attenuating scar formation by the glutamate-MAO-B inhibition increased drug penetration into GBM assembloid, while reducing GBM confinement. CONCLUSIONS: Taken together, our study suggests that astrocytic scar could be a critical modulator in GBM therapeutics.

5.
Int J Mol Sci ; 23(20)2022 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-36293070

RESUMEN

Degeneration of the intervertebral disc (IVD) is a major contributor to low back pain (LBP). IVD degeneration is characterized by abnormal production of inflammatory cytokines secreted by IVD cells. Although the underlying molecular mechanisms of LBP have not been elucidated, increasing evidence suggests that LBP is associated particularly with microglia in IVD tissues and the peridiscal space, aggravating the cascade of degenerative events. In this study, we implemented our microfluidic chemotaxis platform to investigate microglial inflammation in response to our reconstituted degenerative IVD models. The IVD models were constructed by stimulating human nucleus pulposus (NP) cells with interleukin-1ß and producing interleukin-6 (129.93 folds), interleukin-8 (18.31 folds), C-C motif chemokine ligand-2 (CCL-2) (6.12 folds), and CCL-5 (5.68 folds). We measured microglial chemotaxis (p < 0.05) toward the conditioned media of the IVD models. In addition, we observed considerable activation of neurodegenerative and deactivation of protective microglia via upregulated expression of CD11b (p < 0.001) and down-regulation of CD206 protein (p < 0.001) by soluble factors from IVD models. This, in turn, enhances the inflammatory milieu in IVD tissues, causing matrix degradation and cellular damage. Our findings indicate that degenerative IVD may induce degenerative microglial proinflammation, leading to LBP development.


Asunto(s)
Degeneración del Disco Intervertebral , Disco Intervertebral , Dolor de la Región Lumbar , Humanos , Microglía/metabolismo , Interleucina-1beta/metabolismo , Degeneración del Disco Intervertebral/metabolismo , Interleucina-8/metabolismo , Medios de Cultivo Condicionados/metabolismo , Interleucina-6/metabolismo , Ligandos , Disco Intervertebral/metabolismo , Citocinas/metabolismo
6.
Elife ; 112022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35191835

RESUMEN

The membrane protein TREM2 (Triggering Receptor Expressed on Myeloid cells 2) regulates key microglial functions including phagocytosis and chemotaxis. Loss-of-function variants of TREM2 are associated with increased risk of Alzheimer's disease (AD). Because abnormalities in Ca2+ signaling have been observed in several AD models, we investigated TREM2 regulation of Ca2+ signaling in human induced pluripotent stem cell-derived microglia (iPSC-microglia) with genetic deletion of TREM2. We found that iPSC-microglia lacking TREM2 (TREM2 KO) show exaggerated Ca2+ signals in response to purinergic agonists, such as ADP, that shape microglial injury responses. This ADP hypersensitivity, driven by increased expression of P2Y12 and P2Y13 receptors, results in greater release of Ca2+ from the endoplasmic reticulum stores, which triggers sustained Ca2+ influx through Orai channels and alters cell motility in TREM2 KO microglia. Using iPSC-microglia expressing the genetically encoded Ca2+ probe, Salsa6f, we found that cytosolic Ca2+ tunes motility to a greater extent in TREM2 KO microglia. Despite showing greater overall displacement, TREM2 KO microglia exhibit reduced directional chemotaxis along ADP gradients. Accordingly, the chemotactic defect in TREM2 KO microglia was rescued by reducing cytosolic Ca2+ using a P2Y12 receptor antagonist. Our results show that loss of TREM2 confers a defect in microglial Ca2+ response to purinergic signals, suggesting a window of Ca2+ signaling for optimal microglial motility.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Adenosina Difosfato/metabolismo , Enfermedad de Alzheimer/metabolismo , Calcio/metabolismo , Señalización del Calcio , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Microglía/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Purinérgicos/metabolismo
7.
Adv Healthc Mater ; 10(2): e2001261, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33191612

RESUMEN

The limited effectiveness of current therapeutics against malignant brain gliomas has led to an urgent need for development of new formulations against these tumors. Chelator Dp44mT (di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone) presents a promising candidate to defeat gliomas due to its exceptional anti-tumor activity and its unique ability to overcome multidrug resistance. The goal of this study is to develop a targeted nano-carrier for Dp44mT delivery to glioma tumors and to assess its therapeutic efficacy in vitro and in vivo. Dp44mT is loaded into poly(ethylene glycol) (PEG)ylated poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) decorated with glioma-targeting ligand Interlukin 13 (IL13). IL13-conjugation enhanced the NP uptake by glioma cells and also improved their transport across an in vitro blood-brain-barrier (BBB) model. This targeted formulation showed an outstanding toxicity towards glioma cell lines and patient-derived stem cells in vitro, with IC50 values less than 125 nM, and caused no significant death in healthy brain microvascular endothelial cells. In vivo, when tested on a xenograft mouse model, IL13-conjugated Dp44mT-NPs reduced the glioma tumor growth by ≈62% while their untargeted counterparts reduced the tumor growth by only ≈16%. Notably, this formulation does not cause any significant weight loss or kidney/liver toxicity in mice, demonstrating its great therapeutic potential.


Asunto(s)
Neoplasias Encefálicas , Glioma , Nanopartículas , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Resistencia a Múltiples Medicamentos , Células Endoteliales , Glioma/tratamiento farmacológico , Humanos , Ratones
8.
Polymers (Basel) ; 12(6)2020 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-32580368

RESUMEN

Esthetic dental computer-aided design/computer-aided manufacturing (CAD/CAM) polymers such as disperse-filled composites (DFC) and polymer-infiltrated ceramic networks (PICN) should be subjected to surface treatment before bonding. However, such treatment can lead to defect formation and a decrease in strength. Therefore, in this study, we compared the flexural strengths of DFC and PICN materials air-abraded with alumina particles of different sizes at different pressures. In addition to Weibull analysis, the samples (untreated and treated) were characterized by scanning electron microscopy and atomic force microscopy. Both DFC and PICN exhibited the lowest flexural strength at large particle sizes and high pressures. Therefore, we optimized the air abrasion parameters to maintain the flexural strength and significantly increase surface roughness. In the case of DFC, the optimal particle size and pressure conditions were 50 µm at 2 bar and 110 µm at 1 bar, while for PICN, the best performance was obtained using Al2O3 particles with a size of 50 µm at 1 bar. This study reveals that optimization of the surface treatment process is crucial in the fabrication of high-performance clinical materials for dental restorations.

9.
Colloids Surf B Biointerfaces ; 184: 110481, 2019 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-31539751

RESUMEN

This study aims to develop a new anti-cancer formulation based on the chelator Dp44mT (Di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone). Dp44mT has outstanding anti-tumor activity and the unique ability to overcome multidrug-resistance in cancer cells. This highly toxic compound has thus far only been applied in free form, limiting its therapeutic effectiveness. To reach its full therapeutic potential, however, Dp44mT should be encapsulated in a nano-carrier that would enable its selective and controlled delivery to malignant cells. As the first step towards this goal, here we encapsulate Dp44mT in nanoparticles (NPs) of poly(lactic-co-glycolic acid) (PLGA), characterize this nano-formulation, and evaluate its therapeutic potential against cancer cells in vitro. Our results showed that the Dp44mT-loaded NPs were homogenous in shape and size, and had good colloidal stability. These PLGA NPs also showed high encapsulation efficiency and loading capacity for Dp44mT and enabled the sustained and tunable release of this chelator. Dp44mT-NPs were uptaken by cancer cells, showed a strong and dose-dependent cytotoxicity towards these cells, and significantly increased apoptotic cell death, in both monolayer and spheroid tumor models. This formulation had a low-level of toxicity towards healthy control cells, indicating an inherent selectivity towards malignant cells. These results demonstrate the great potential of this novel Dp44mT-based nano-formulation for the use in cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Materiales Biocompatibles/farmacología , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Tiosemicarbazonas/farmacología , Antineoplásicos/química , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacocinética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Quelantes/química , Quelantes/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Tamaño de la Partícula , Tiosemicarbazonas/química , Tiosemicarbazonas/farmacocinética
10.
PLoS One ; 14(1): e0211078, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30677075

RESUMEN

Treatment of glioblastoma, the most common and aggressive type of primary brain tumors, is a major medical challenge and the development of new alternatives requires simple yet realistic models for these tumors. In vitro spheroid models offer attractive platforms to mimic the tumor behavior in vivo and have thus, been increasingly applied for assessment of drug efficacy in various tumors. The aim of this study was to produce and characterize size-controlled U251 glioma spheroids towards application in glioma drug evaluation studies. To this end, we fabricated agarose hydrogel microwells with cylindrical shape and diameters of 70-700 µm and applied these wells without any surface modification for glioma spheroid formation. The resultant spheroids were homogeneous in size and shape, exhibited high cell viability (> 90%), and had a similar growth rate to that of natural brain tumors. The final size of spheroids depended on cell seeding density and microwell size. The spheroids' volume increased linearly with the cell seeding density and the rate of this change increased with the well size. Lastly, we tested the therapeutic effect of an anti-cancer drug, Di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT) on the resultant glioma spheroids and demonstrated the applicability of this spheroid model for drug efficacy studies.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Glioma/metabolismo , Hidrogeles/química , Sefarosa/química , Esferoides Celulares/metabolismo , Línea Celular Tumoral , Glioma/patología , Humanos , Esferoides Celulares/patología
11.
Nano Converg ; 5(1): 35, 2018 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-30499047

RESUMEN

The major neurological disorders found in a central nervous system (CNS), such as brain tumors, Alzheimer's diseases, Parkinson's diseases, and Huntington's disease, have led to devastating outcomes on the human public health. Of these disorders, early diagnostics remains poor, and no treatment has been successfully discovered; therefore, they become the most life-threatening medical burdens worldwide compared to other major diseases. The major obstacles for the drug discovery are the presence of a restrictive blood-brain barrier (BBB), limiting drug entry into brains and undesired neuroimmune activities caused by untargeted drugs, leading to irreversible neuronal damages. Recent advances in nanotechnology have contributed to the development of novel nanoplatforms and effective delivering strategies to improve the CNS disorder treatment while less disturbing brain systems. The nanoscale drug carriers, including liposomes, dendrimers, viral capsids, polymeric nanoparticles, silicon nanoparticles, and magnetic/metallic nanoparticles, enable the effective drug delivery penetrating across the BBB, the aforementioned challenges in the CNS. Moreover, drugs encapsulated by the nanocarriers can reach further deeper into targeting regions while preventing the degradation. In this review, we classify novel disease hallmarks incorporated with emerging nanoplatforms, describe promising approaches for improving drug delivery to the disordered CNS, and discuss their implications for clinical practice.

12.
Proc Natl Acad Sci U S A ; 112(32): 9810-5, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26216964

RESUMEN

Bioinspired artificial water channels aim to combine the high permeability and selectivity of biological aquaporin (AQP) water channels with chemical stability. Here, we carefully characterized a class of artificial water channels, peptide-appended pillar[5]arenes (PAPs). The average single-channel osmotic water permeability for PAPs is 1.0(± 0.3) × 10(-14) cm(3)/s or 3.5(± 1.0) × 10(8) water molecules per s, which is in the range of AQPs (3.4 ∼ 40.3 × 10(8) water molecules per s) and their current synthetic analogs, carbon nanotubes (CNTs, 9.0 × 10(8) water molecules per s). This permeability is an order of magnitude higher than first-generation artificial water channels (20 to ∼ 10(7) water molecules per s). Furthermore, within lipid bilayers, PAP channels can self-assemble into 2D arrays. Relevant to permeable membrane design, the pore density of PAP channel arrays (∼ 2.6 × 10(5) pores per µm(2)) is two orders of magnitude higher than that of CNT membranes (0.1 ∼ 2.5 × 10(3) pores per µm(2)). PAP channels thus combine the advantages of biological channels and CNTs and improve upon them through their relatively simple synthesis, chemical stability, and propensity to form arrays.


Asunto(s)
Canales Iónicos/química , Agua/química , Acuaporinas/química , Iones , Modelos Moleculares , Simulación de Dinámica Molecular , Nanotubos de Carbono , Péptidos/química , Permeabilidad , Liposomas Unilamelares/química
13.
Carcinogenesis ; 34(8): 1918-28, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23615398

RESUMEN

Tumor necrosis factor-related apoptosis-induced ligand (TRAIL) is preferentially cytotoxic to cancer cells over normal cells. However, many cancer cells, including malignant glioma cells, tend to be resistant to TRAIL. Monensin (a polyether ionophore antibiotic that is widely used in veterinary medicine) and salinomycin (a compound that is structurally related to monensin and shows cancer stem cell-inhibiting activity) are currently recognized as anticancer drug candidates. In this study, we show that monensin effectively sensitizes various glioma cells, but not normal astrocytes, to TRAIL-mediated apoptosis; this occurs at least partly via monensin-induced endoplasmic reticulum (ER) stress, CHOP-mediated DR5 upregulation and proteasome-mediated downregulation of c-FLIP. Interestingly, other polyether antibiotics, such as salinomycin, nigericin, narasin and lasalocid A, also stimulated TRAIL-mediated apoptosis in glioma cells via ER stress, CHOP-mediated DR5 upregulation and c-FLIP downregulation. Taken together, these results suggest that combined treatment of glioma cells with TRAIL and polyether ionophore antibiotics may offer an effective therapeutic strategy.


Asunto(s)
Antibacterianos/farmacología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glioma/tratamiento farmacológico , Monensina/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Glioma/genética , Glioma/metabolismo , Humanos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteínas Recombinantes/farmacología , Regulación hacia Arriba/efectos de los fármacos
14.
Carcinogenesis ; 33(3): 492-500, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22159225

RESUMEN

Tumor necrosis factor-related apoptosis-induced ligand (TRAIL) induces apoptosis selectively in cancer cells while sparing normal cells. However, many cancer cells are resistant to TRAIL-induced cell death. In this study, we examined whether Aurora B, which is frequently overexpressed in cancer cells, is associated with TRAIL resistance. The protein levels of Aurora B were higher in TRAIL-resistant cancer cell lines than in TRAIL-sensitive cancer cell lines. Exogenously expressed Aurora B attenuated TRAIL-induced apoptosis in the tested TRAIL-sensitive cancer cell lines, whereas the small interfering RNA-mediated suppression of Aurora B expression stimulated TRAIL-mediated apoptosis in the tested TRAIL-resistant cancer cell lines. Furthermore, combined treatment with TRAIL and ZM447439, a specific inhibitor of Aurora B, synergistically induced apoptosis in various TRAIL-resistant cancer cells, suggesting that this combined regimen may represent an attractive strategy for effectively treating TRAIL-resistant malignant cancers. Mechanistically, the inhibition of Aurora B activity in various cancer cells commonly downregulated survivin protein levels and potentiated the activation of caspase-3. In addition, Aurora B inhibition induced mitotic catastrophe, which also contributed to the sensitization of cells to TRAIL-mediated apoptosis. Interestingly, forced overexpression of Aurora B increased the protein levels of survivin, but not those of a non-phosphorylatable survivin mutant in which threonine 117 was replaced by alanine, indicating that phosphorylation of survivin is required for this effect. Furthermore, TRAIL-induced apoptosis in MDA-MB-435S cells was attenuated by wild-type survivin but not by the non-phosphorylatable survivin mutant. Collectively, our results demonstrate that Aurora B confers TRAIL resistance to cancer cells via phosphorylation of survivin.


Asunto(s)
Apoptosis , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Aurora Quinasa B , Aurora Quinasas , Benzamidas/farmacología , Caspasa 3/biosíntesis , Caspasa 3/metabolismo , Línea Celular Tumoral , Células Hep G2 , Humanos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Neoplasias , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Quinazolinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño , Survivin , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
15.
Neuro Oncol ; 13(3): 267-79, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21292685

RESUMEN

Amiodarone is a widely used anti-arrhythmic drug that inhibits diverse ion channels, including the Na(+)/Ca(2+) exchanger (NCX), L-type Ca(2+) channels, and Na(+) channels. Here, we report that subtoxic doses of amiodarone and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) synergistically induced apoptosis of various glioma cells. Treatment of U251MG glioma cells with amiodarone increased intracellular Ca(2+) levels and enhanced the expression of the endoplasmic reticulum (ER) stress-inducible transcription factor C/EBP homologous protein (CHOP). This upregulation of CHOP was followed by marked upregulation of the TRAIL receptor, DR5. Suppression of DR5 expression by small interfering (si) RNAs almost completely blocked amiodarone/TRAIL-induced apoptosis in U251MG glioma cells, demonstrating that DR5 is critical to this cell death. siRNA-mediated CHOP suppression reduced amiodarone-induced DR5 upregulation and attenuated the cell death induced by amiodarone plus TRAIL. In addition, omitting Ca(2+) from the external medium using ethylene glycol tetraacetic acid markedly inhibited this cell death, reducing the protein levels of CHOP and DR5. These results suggest that amiodarone-induced influx of Ca(2+) plays an important role in sensitizing U251MG cells to TRAIL-mediated apoptosis through CHOP-mediated DR5 upregulation. Furthermore, subtoxic doses of bepridil and cibenzoline, two other anti-arrhythmic drugs with NCX-inhibitor activity, also sensitized glioma cells to TRAIL-mediated apoptosis, via the upregulation of both CHOP and DR5. Notably, amiodarone/TRAIL cotreatment did not induce cell death in astrocytes, nor did it affect the expression of CHOP or DR5 in these cells. These results collectively suggest that a combined regimen of amiodarone plus TRAIL may offer an effective therapeutic strategy for safely and selectively treating resistant gliomas.


Asunto(s)
Amiodarona/farmacología , Apoptosis/efectos de los fármacos , Astrocitos/patología , Glioma/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Factor de Transcripción CHOP/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Glioma/tratamiento farmacológico , Glioma/metabolismo , Humanos , Luciferasas/metabolismo , ARN Mensajero/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción CHOP/genética , Vasodilatadores/farmacología
16.
Exp Mol Med ; 43(1): 24-34, 2011 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-21150246

RESUMEN

Tumor necrosis factor-related apoptosis-induced ligand (TRAIL) induces apoptosis selectively in cancer cells while sparing normal cells. However, many cancer cells are resistant to TRAIL-induced cell death. Here, we report that paxilline, an indole alkaloid from Penicillium paxilli, can sensitize various glioma cells to TRAIL-mediated apoptosis. While treatment with TRAIL alone caused partial processing of caspase-3 to its p20 intermediate in TRAIL-resistant glioma cell lines, co-treatment with TRAIL and subtoxic doses of paxilline caused complete processing of caspase-3 into its active subunits. Paxilline treatment markedly upregulated DR5, a receptor of TRAIL, through a CHOP/GADD153-mediated process. In addition, paxilline treatment markedly downregulated the protein levels of the short form of the cellular FLICE-inhibitory protein (c-FLIPs) and the caspase inhibitor, survivin, through proteasome-mediated degradation. Taken together, these results show that paxilline effectively sensitizes glioma cells to TRAIL-mediated apoptosis by modulating multiple components of the death receptor-mediated apoptotic pathway. Interestingly, paxilline/TRAIL co-treatment did not induce apoptosis in normal astrocytes, nor did it affect the protein levels of CHOP, DR5 or survivin in these cells. Thus, combined treatment regimens involving paxilline and TRAIL may offer an attractive strategy for safely treating resistant gliomas.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Glioma/metabolismo , Indoles/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Astrocitos/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Descubrimiento de Drogas , Citometría de Flujo , Glioma/patología , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , ARN Interferente Pequeño , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Survivin , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Factor de Transcripción CHOP/análisis
17.
Anticancer Res ; 23(6C): 4709-13, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14981917

RESUMEN

BACKGROUND: Normal cells lose their ability to divide after a finite number of cell divisions. Under the influence of Simian virus 40 (SV 40) large T-antigen, which interacts with the cell cycle regulators p53 and pRb, cells enter a phase of an extended pre-immortalized cells. Immortalization-up-regulated protein 1 (IMUP-1) and immortalization-up-regulated protein 2 (IMUP-2) genes have been recently cloned and are known to be involved in SV40-mediated immortalization. However, the roles of IMUP-1 and IMUP-2 are not known in human ovarian epithelial tumors. PATIENTS AND METHODS: This study was performed to determine mRNA expression and intracellular localization of the IMUP-1 and IMUP-2 in ovarian epithelial tumors by RT-PCR and immunohistochemical staining using human IMUP-1 and IMUP-2 monoclonal antibodies. RESULTS: IMUP-1 (4.0-fold) and IMUP-2 (2.4-fold) mRNA expression in ovarian epithelial tumors were significantly higher than in normal ovarian tissues (p < 0.05). The mRNAs of IMUP-1 and IMUP-2 in the ovarian cancer cell lines were about 4.9- and 2.9-fold compared to the normal ovarian cell line, respectively. The subcellular expression of these two proteins in immunohistochemical stains was detected mainly in the nucleus of tumor cells, whereas adjacent normal ovarian stromal cells were faintly or negatively stained with these proteins. However, the staining intensity of IMUP-1 and IMUP-2 in ovarian epithelial tumors were not different between histological types or grades. CONCLUSION: These results showed the up-regulation of IMUP-1 and IMUP-2 in human ovarian epithelial tumors and suggested that the altered mRNA level of these molecules is possibly associated with ovarian tumorigenesis.


Asunto(s)
Carcinoma/genética , Regulación Neoplásica de la Expresión Génica/genética , Proteínas Nucleares/genética , Neoplasias Ováricas/genética , Factores de Transcripción/genética , Femenino , Humanos , Inmunohistoquímica , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA