Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Control Release ; 368: 42-51, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38365180

RESUMEN

Protein corona has long been a source of concern, as it might impair the targeting efficacy of targeted drug delivery systems. However, engineered up-regulating the adsorption of certain functional serum proteins could provide nanoparticles with specific targeting drug delivery capacity. Herein, apolipoprotein A-I absorption increased nanoparticles (SPC-PLGA NPs), composed with the Food and Drug Administration approved intravenously injectable soybean phosphatidylcholine (SPC) and poly (DL-lactide-co-glycolide) (PLGA), were fabricated for enhanced glioma targeting. Due to the high affinity of SPC and apolipoprotein A-I, the percentage of apolipoprotein A-I in the protein corona of SPC-PLGA NPs was 2.19-fold higher than that of nanoparticles without SPC, which made SPC-PLGA NPs have superior glioma targeting ability through binding to scavenger receptor class BI on blood-brain barrier and glioma cells both in vitro and in vivo. SPC-PLGA NPs loaded with paclitaxel could effectively reduce glioma invasion and prolong the survival time of glioma-bearing mice. In conclusion, we provided a good example of the direction of achieving targeting drug delivery based on protein corona regulation.


Asunto(s)
Glioma , Nanopartículas , Corona de Proteínas , Ratones , Animales , Apolipoproteína A-I , Línea Celular Tumoral , Glioma/tratamiento farmacológico , Glioma/metabolismo , Paclitaxel/uso terapéutico , Sistemas de Liberación de Medicamentos , Portadores de Fármacos/uso terapéutico
2.
Int J Pharm ; 630: 122436, 2023 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-36436742

RESUMEN

Ibrutinib (IBR) is an oral covalent inhibitor of Bruton's tyrosine kinase (BTK) that has been approved for the treatment of hematological malignancies. It was reported that IBR exhibited great therapeutic potential for glioma. However, the poor water solubility and high hepatic first-pass effect restrict its anti-glioma application. Meanwhile, IBR induces cytoprotective autophagy through Akt/mTOR signaling pathway, thus leading to a compromised antitumor effect. Herein, we aimed to develop a human serum albumin (HSA) based co-delivery system (IBR&HCQ HSA NPs) encapsulating IBR and hydroxychloroquine (HCQ). The bioavailability of IBR was largely improved, and enhanced sensitivity of glioma to IBR was achieved due to inhibition effect of HCQ on IBR-induced pro-survival autophagy. The physicochemical properties of IBR&HCQ HSA NPs were characterized to optimize the formulation. Biodistribution investigation revealed that HSA NPs (20 mg/kg, i.v.) dramatically increased the accumulation of IBR in glioma, which was 5.59 times higher than that of free IBR (100 mg/kg, i.g.). CCK-8 and apoptosis assays demonstrated that IBR&HCQ HSA NPs showed maximal cytotoxicity to C6 cells. In vivo studies indicated that the survival time was significantly prolonged in IBR&HCQ HSA NPs treated mice compared to those treated with IBR HSA NPs. Taken together, the HSA-based drug delivery system of IBR and HCQ opens a new avenue for efficient treatment of glioma.


Asunto(s)
Glioma , Nanopartículas , Humanos , Ratones , Animales , Hidroxicloroquina/farmacología , Distribución Tisular , Glioma/tratamiento farmacológico , Nanopartículas/química , Albúmina Sérica Humana , Línea Celular Tumoral
3.
Acta Pharm Sin B ; 12(4): 1987-1999, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35847512

RESUMEN

The treatment of Alzheimer's disease (AD) is one of the most difficult challenges in neurodegenerative diseases due to the insufficient blood‒brain barrier (BBB) permeability and unsatisfactory intra-brain distribution of drugs. Therefore, we established an ibuprofen and FK506 encapsulated drug co-delivery system (Ibu&FK@RNPs), which can target the receptor of advanced glycation endproducts (RAGE) and response to the high level of reactive oxygen species (ROS) in AD. RAGE is highly and specifically expressed on the lesion neurovascular unit of AD, this property helps to improve targeting specificity of the system and reduce unselective distribution in normal brain. Meanwhile, these two drugs can be specifically released in astrocytes of AD lesion in response to high levels of ROS. As a result, the cognition of AD mice was significantly improved and the quantity of Aß plaques was decreased. Neurotoxicity was also alleviated with structural regeneration and functional recovery of neurons. Besides, the neuroinflammation dominated by NF-κB pathway was significantly inhibited with decreased NF-κB and IL-1ß in the brain. Overall, Ibu&FK@RNPs can efficiently and successively target diseased BBB and astrocytes in AD lesion. Thus it significantly enhances intracephalic accumulation of drugs and efficiently treats AD by anti-neuroinflammation and neuroprotection.

5.
Small ; 18(30): e2203182, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35771092

RESUMEN

Alzheimer's disease (AD), as a progressive and irreversible brain disorder, remains the most universal neurodegenerative disease. No effective therapeutic methods are established yet due to the hindrance of the blood-brain barrier (BBB) and the complex pathological condition of AD.  Therefore, a multifunctional nanocarrier (Rapa@DAK/siRNA) for AD treatment is constructed to achieve small interfering RNA of ß-site precursor protein (APP) cleaving enzyme-1 (BACE1 siRNA) and rapamycin co-delivery into the brain, based on Aleuria aurantia lectin (AAL) and ß-amyploid (Aß)-binding peptides (KLVFF) modified PEGylated dendrigraft poly-l-lysines (DGLs) via intranasal administration. Nasal administration provides an effective way to deliver drugs directly into the brain through the nose-to-brain pathway. AAL, specifically binding to L-fucose located in the olfactory epithelium, endows Rapa@DAK/siRNA with high brain entry efficiency through intranasal administration. KLVFF peptide as an Aß targeting ligand and aggregation inhibitor enables nanoparticles to bind with Aß, inhibit Aß aggregation, and reduce toxicity. Meanwhile, the release of BACE1 siRNA and rapamycin is confirmed to reduce BACE1 expression, promote autophagy, and reduce Aß deposition. Rapa@DAK/siRNA is verified to improve the cognition of transgenic AD mice after intranasal administration. Collectively, the multifunctional nanocarrier provides an effective and potential intranasal avenue for combination therapy of AD.


Asunto(s)
Enfermedad de Alzheimer , Nanopartículas , Enfermedades Neurodegenerativas , Administración Intranasal , Enfermedad de Alzheimer/tratamiento farmacológico , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Secretasas de la Proteína Precursora del Amiloide/farmacología , Péptidos beta-Amiloides/metabolismo , Animales , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Ácido Aspártico Endopeptidasas/farmacología , Encéfalo/metabolismo , Ratones , Ratones Transgénicos , ARN Interferente Pequeño , Sirolimus/farmacología
6.
Adv Sci (Weinh) ; 9(14): e2104286, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35285177

RESUMEN

The treatment of autism spectrum disorder (ASD) is one of the most difficult challenges in neurodevelopmental diseases, because of the unclear pathogenesis research and low brain-lesion targeting efficiency. Besides, maternal immune activation has been reported as the most mature and widely used model of ASD and aspirin-triggered lipoxin A4 is a potent anti-inflammatory mediator being involved in the resolution of neuroinflammation in ASD. Therefore, an aspirin encapsulated cascade drug delivery system (Asp@TMNPs) is established, which can successively target the blood-brain barrier (BBB) and microglial cells and response to the acid microenvironment in lysosome. As a result, the mitochondrial oxidative stress, DNA damage, and inflammation of microglial cells are prominently alleviated. After the treatment of Asp@TMNPs, the social interaction, stereotype behavior, and anxious condition of ASD mice are notably improved and the activation of microglial cells is inhibited. Overall, this system successively penetrates the BBB and targets microglial cells, therefore, it significantly enhances the intracephalic drug accumulation and improves anti-neuroinflammatory efficacy of aspirin, providing a promising strategy for ASD treatment.


Asunto(s)
Trastorno del Espectro Autista , Nanopartículas , Animales , Aspirina/uso terapéutico , Trastorno del Espectro Autista/tratamiento farmacológico , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Ratones , Microglía/patología , Fenotipo
7.
Front Pharmacol ; 12: 719905, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34630098

RESUMEN

It's of great significance to develop insulin-loaded dissolving microneedles (MNs) which are fabricated with various methods and materials for transdermal delivery of insulin to effectively and efficiently treat diabetes. In this work, we present a kind of FITC-insulin tip-loaded dissolving MNs fabricated with the mixture of polyvinyl alcohol (PVA) and sucrose using homemade PDMS MNs mold under vacuum conditions. The uniform appearance of MN arrays contributes to controlling the drug dosage well as required. Sufficient mechanical strength for penetrating tough stratum corneum can be obtained by vacuum frozen-drying for at least 6 h after peeling MNs off the mold. About 90% of the FITC-insulin is localized in the conical MN tips and can be released into the skin within 2 min after insertion. The in vivo insulin absorption study and hypoglycemic effect in diabetic mice demonstrate that the proposed insulin-loaded MNs can efficiently deliver the insulin to the systemic circulation and exhibit a similar effect to hypodermic injection on hypoglycemic administration. Together these results suggested that the efficient MN fabrication process proposed in this work shows great potential for mass production and practical application of drug-loaded dissolving MNs in the future.

8.
Acta Pharm Sin B ; 11(12): 4032-4044, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35024324

RESUMEN

Insurmountable blood‒brain barrier (BBB) and complex pathological features are the key factors affecting the treatment of Alzheimer's disease (AD). Poor accumulation of drugs in lesion sites and undesired effectiveness of simply reducing Aß deposition or TAU protein need to be resolved urgently. Herein, a nanocleaner is designed with a rapamycin-loaded ROS-responsive PLGA core and surface modification with KLVFF peptide and acid-cleavable DAG peptide [R@(ox-PLGA)-KcD]. DAG can enhance the targeting and internalization effect of nanocleaner towards neurovascular unit endothelial cells in AD lesions, and subsequently detach from nanocleaner in response to acidic microenvironment of endosomes to promote the transcytosis of nanocleaner from endothelial cells into brain parenchyma. Then exposed KLVFF can capture and carry Aß to microglia, attenuating Aß-induced neurotoxicity. Strikingly, rapamycin, an autophagy promoter, is rapidly liberated from nanocleaner in the high ROS level of lesions to improve Aß degradation and normalize inflammatory condition. This design altogether accelerates Aß degradation and alleviates oxidative stress and excessive inflammatory response. Collectively, our finding offers a strategy to target the AD lesions precisely and multi-pronged therapies for clearing the toxic proteins and modulating lesion microenvironment, to achieve efficient AD therapy.

9.
Acta Pharm Sin B ; 10(12): 2348-2361, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33354506

RESUMEN

Accurate tumor targeting, deep penetration and superb retention are still the main pursuit of developing excellent nanomedicine. To achieve these requirements, a stepwise stimuli-responsive strategy was developed through co-administration tumor penetration peptide iRGD with shape-transformable and GSH-responsive SN38-dimer (d-SN38)-loaded nanoparticles (d-SN38@NPs/iRGD). Upon intravenous injection, d-SN38@NPs with high drug loading efficiency (33.92 ± 1.33%) could effectively accumulate and penetrate into the deep region of tumor sites with the assistance of iRGD. The gathered nanoparticles simultaneously transformed into nanofibers upon 650 nm laser irradiation at tumor sites so as to promote their retention in the tumor and burst release of reactive oxygen species for photodynamic therapy. The loaded d-SN38 with disulfide bond responded to the high level of GSH in tumor cytoplasm, which consequently resulted in SN38 release and excellent chemo-photodynamic effect on tumor. In vitro, co-administering iRGD with d-SN38@NPs+laser showed higher cellular uptake, apoptosis ratio and multicellular spheroid penetration. In vivo, d-SN38@NPs/iRGD+laser displayed advanced penetration and accumulation in tumor, leading to 60.89% of tumor suppression in 4T1 tumor-bearing mouse model with a favorable toxicity profile. Our new strategy combining iRGD with structural transformable nanoparticles greatly improves tumor targeting, penetrating and retention, and empowers anticancer efficacy.

10.
Sci Adv ; 6(36)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32917616

RESUMEN

Application of cancer vaccines is limited due to their systemic immunotoxicity and inability to satisfy all the steps, including loading of tumor antigens, draining of antigens to lymph nodes (LNs), internalization of antigens by dendritic cells (DCs), DC maturation, and cross-presentation of antigens for T cell activation. Here, we present a combinatorial therapy, based on a α-cyclodextrin (CD)-based gel system, DOX/ICG/CpG-P-ss-M/CD, fabricated by encapsulating doxorubicin (DOX) and the photothermal agent indocyanine green (ICG). Upon irradiation, the gel system exhibited heat-responsive release of DOX and vaccine-like nanoparticles, CpG-P-ss-M, along with chemotherapy- and phototherapy-generated abundant tumor-specific antigen storage in situ. The released CpG-P-ss-M acted as a carrier adsorbed and delivered antigens to LNs, promoting the uptake of antigens by DCs and DC maturation. Notably, combined with PD-L1 blocking, the therapy effectively inhibited primary tumor growth and induced tumor-specific immune response against tumor recurrence and metastasis.

11.
Drug Deliv ; 27(1): 1156-1164, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32755291

RESUMEN

In this work, we prepared a stimuli-responsive system for drug delivery and controlled release by engineering the bovine serum albumin (BSA). The doxorubicin (DOX)-loaded BSA nanoparticles (NPs) were conveniently prepared using desolvation method, followed by crosslinking through Schiff base bonds, leading to pH-sensitive DOX-loaded system (DOXs@BSA NPs). The resulted DOXs@BSA NPs showed high drug loading capacity (21.4%), and the particle size was about 130 nm with narrow polydispersity and high negative surface charge (-20.5 mV). The pH-sensitivity of DOXs@BSA NPs was evidenced by the size changes and charge reversal after incubation at different pH values. The DOXs@BSA NPs showed high serum stability which indicated the prolonged circulation time. The in vitro drug release experiment showed that the release of DOX was obviously accelerated by acidity because of disassembly of NPs induced by cleavage of Schiff base bonds. The drug release mechanism was thoroughly studied using a semi-empirical model, further confirming the pH played an important role in drug controlled release process. The results of cytotoxicity assay revealed that DOXs@BSA NPs exhibited much higher toxic effects for tumor cells in comparison to the free DOX control. Collectively, these results demonstrated that DOXs@BSA NPs might be potential application for drug delivery and controlled release in cancer chemotherapy. Moreover, this work also showed that preparation of stimuli-responsive drug delivery system by engineering the commercial biomaterials could be a promising method to develop multi-functional nanomedicine.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Nanopartículas/química , Albúmina Sérica Bovina/química , Animales , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Química Farmacéutica , Preparaciones de Acción Retardada , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Estabilidad de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Ratones , Tamaño de la Partícula , Propiedades de Superficie
12.
Biomaterials ; 217: 119309, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31271855

RESUMEN

Recently, photodynamic therapy (PDT) emerges as a promising way to initiate immune response and being used in combination with chemotherapy. However, the antitumor effect is restricted due to the poor tumor penetration and retention, premature drug release and immunosuppressive environment of tumor sites. And as the size of nanoparticles plays a key role in drug delivery, series of hyaluronidase-responsive size-reducible biomimetic nanoparticles (mCAuNCs@HA) with different initial sizes are synthesized, and the optimal size of 150 nm is screened out because of the best blood circulation, tumor penetration and retention. Then the photosensitizer pheophorbide A and ROS-responsive paclitaxel dimer prodrug (PXTK) are co-loaded to facilitate on-demand drug release. The hydrolysis byproduct cinnamaldehyde in turn stimulates the ROS production by mitochondria, which compensates for the ROS consumed in the hydrolysis process. Anti-PD-L1 peptide (dPPA) is furthered loaded to alleviate the immunosuppressive environment of tumor and enhance the function of cytotoxic T lymphocytes activated by PDT-induced immunogenic cell death. The combination therapy activates CD4+, CD8+ T cells and NK cells and enhances secretion of cytokines (TNF-α and IL-12) with tumor inhibition rate increased to 84.2% and no metastasis is observed, providing a viable combination therapy for better anti-tumor and anti-metastasis efficacy.


Asunto(s)
Materiales Biomiméticos/química , Neoplasias de la Mama/terapia , Inmunoterapia , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Nanopartículas del Metal/química , Tamaño de la Partícula , Animales , Antígeno B7-H1/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Cationes , Muerte Celular , Línea Celular Tumoral , Endocitosis/efectos de los fármacos , Femenino , Oro/química , Humanos , Hialuronoglucosaminidasa/metabolismo , Neoplasias Pulmonares/inmunología , Nanopartículas del Metal/ultraestructura , Ratones , Ratones Endogámicos BALB C , Paclitaxel/farmacocinética , Paclitaxel/uso terapéutico , Péptidos/química , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Distribución Tisular/efectos de los fármacos
13.
Front Physiol ; 9: 473, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29867533

RESUMEN

Monocyte/macrophage recruitment is closely associated with the degree of hypertensive renal injury. We investigated the direct role of macrophages using liposome-encapsulated clodronate (LEC) to deplete monocytes/macrophages in hypertensive renal injury. C57BL/6 mice were treated with a pressor dose of angiotensin (Ang, 1.4 mg/kg/day) II plus LEC or the PBS-liposome for 2 weeks. Ang II mice developed hypertension, albuminuria, glomerulosclerosis, and renal fibrosis. LEC treatment reduced systolic blood pressure (SBP), albuminuria, and protected against renal structural injury in Ang II mice. Ang II significantly increased renal macrophage infiltration (MOMA2+ cells) and the expression of renal tumor necrosis factor α and interleukin ß1, which were significantly reduced in Ang II/LEC mice. Ang II increased renal oxidative stress and the expression of profibrotic factors transforming growth factor (TGF) ß1 and fibronectin. Ang II also inhibited the phosphorylation of endothelial nitric oxide synthase [phospho-endothelial nitric oxide synthesis (eNOS), ser1177]. LEC treatment reduced renal oxidative stress and TGFß1 and fibronectin expressions, and increased phospho-eNOS expression in the Ang II mice. In Dahl rats of salt-sensitive hypertension, LEC treatment for 4 weeks significantly attenuated the elevation of SBP induced by high salt intake and protected against renal injury and fibrosis. Our results demonstrate that renal macrophages play a critical role in the development of hypertension and hypertensive renal injury and fibrosis; the underlying mechanisms may be involved in the reduction in macrophage-driven renal inflammation and restoration of the balance between renal oxidative stress and eNOS. Therefore, macrophages should be considered as a potential therapeutic target to reduce the adverse consequences of hypertensive renal diseases.

14.
Neurochem Res ; 42(12): 3439-3445, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28852940

RESUMEN

Silibinin has been widely used to treat liver diseases due to its antioxidant activity. However, the effects of silibinin on the central nervous system have not been thoroughly investigated. The pathological hallmarks of Alzheimer's disease are the accumulation of amyloid ß protein, development of neurofibrillary tangles and increased oxidative stress, which ultimately lead to irreversible neuronal loss and cognitive impairment. Our findings show that silibinin ameliorated memory impairments in APP/PS1 mice in the Morris water maze via suppression of oxidative stress and inhibition of apoptosis. Treatment with silibinin reduced malondialdehyde content level and increased glutathione and superoxide dismutase activity in APP/PS1 mice. A terminal deoxynucleotidyl transferase dUTP nick end labeling assay revealed an anti-apoptotic effect of silibinin. Silibinin suppressed the activation of caspase-3 by inhibiting Jun N-terminal kinase phosphorylation and the downstream hippocampal Bax/Bcl-2 ratio. Silibinin treatment significantly increased levels of synaptophysin and PSD95 in APP/PS1 transgenic mice. These results suggest that silibinin could be a potential therapeutic agent for the treatment of Alzheimer's disease.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Antioxidantes/farmacología , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Silimarina/farmacología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones Transgénicos , Estrés Oxidativo , Silibina
15.
Neurosci Lett ; 629: 256-261, 2016 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-27276653

RESUMEN

Silibinin was reported to be effective in reversing the learning and memory deficits of several AD animal models. These improvements are thought to be regulated by various factors, including antioxidative stress, inhibition of acetylcholinesterase activity and Aß aggregation. However, there are still no reports that demonstrate the effect of silibinin on microglia activation in vivo. Thus, in this study, we used the senescence-accelerated mouse (SAMP8) strain to test the effects of silibinin on behavioral impairments and microglia activation-induced neuroinflammation. Silibinin treatment significantly rescued memory deficits in novel object recognition test and Morris water maze test. Silibinin treatment significantly attenuated microglial activation; down-regulated the level of the proinflammatory cytokine IL-6, anti-inflammatory cytokine IL-4, and inflammation-associated proteins, iNOS and COX-2; and further modulated MAPK to protect neural cells. These results suggest that silibinin could be a potential candidate for the therapy of neurodegenerative disorders.


Asunto(s)
Encefalitis/metabolismo , Aprendizaje/efectos de los fármacos , Trastornos de la Memoria/metabolismo , Memoria/efectos de los fármacos , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Silimarina/administración & dosificación , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Ciclooxigenasa 2/metabolismo , Encefalitis/complicaciones , Encefalitis/prevención & control , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Interleucina-4/metabolismo , Interleucina-6/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/prevención & control , Ratones , Microglía/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Silibina , Memoria Espacial/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
J Neurosci Res ; 92(6): 703-13, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24523141

RESUMEN

This study aims to determine the effects of vascular endothelial growth factor (VEGF), papaverine (PA), and the combination of VEGF and PA on the permeability of the blood-tumor barrier (BTB) and to determine possible molecular mechanisms contributing to the effects. In the rat C6 glioma model, the extravasation of Evans blue (EB) through the BTB was increased significantly by VEGF and PA. VEGF-induced and PA-induced increase of EB extravasation was further increased after combining VEGF with PA infusion. Transmission electron microscopy (TEM) showed that the combination of VEGF and PA not only opened tight junctions (TJ) dramatically but increased the presence of pinocytotic vesicles of brain microvascular endothelial cells (BMECs) significantly. Meanwhile, the downregulation of the TJ-associated proteins occludin and claudin-5 and the upregulation of the caveolae structure proteins caveolin-1 and caveolin-2 caused by the combination of VEGF and PA were observed by Western blot and immunohistochemistry, which were more remarkable than those by the two strategies separately. In addition, after VEGF and PA infusion, the results of radioimmunoassay, Western blot, and enzyme-linked immunosorbent assay (ELISA) revealed a significant increase in expression levels of cGMP and protein kinase G-1 (PKG-1) and the activation of nuclear factor-κB (NF-κB) p65. This study demonstrates that combination of VEGF and PA can increase the permeability of the BTB by a paracellular pathway (downregulation of occludin and claudin-5) and a transcellular pathway (upregulation of caveolin-1 and caveolin-2) and that the cGMP/PKG/NF-κB signal pathway might be involved in the modulation process.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/patología , Papaverina/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Western Blotting , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Femenino , Glioma/irrigación sanguínea , Glioma/patología , Inmunohistoquímica , Microscopía Electrónica de Transmisión , Inhibidores de Fosfodiesterasa/farmacología , Pinocitosis/efectos de los fármacos , Radioinmunoensayo , Ratas , Ratas Wistar , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/ultraestructura
17.
J Mol Neurosci ; 44(2): 122-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21193965

RESUMEN

The first goal of this study was to determine the effect of vascular endothelial growth factor (VEGF) on permeability of the blood-tumor barrier (BTB). The second goal was to determine possible cellular mechanisms by which VEGF increases permeability of the BTB. In the rat C6 glioma model, the permeability of the BTB was significantly increased after VEGF injection at dose of 0.05 ng/g and reached its peak at 45 min. Meanwhile, we observed that the density of pinocytotic vesicles of brain microvascular endothelial cells (BMECs) in the BTB increased dramatically by transmission electron microscopy. The immunohistochemistry and western blot analysis revealed that the expression level of caveolae structure proteins caveolin-1 and caveolin-2 in BMECs was increased after VEGF injection, peaked at 45 min, and then decreased to the untreated level. The time peak of expression level of caveolin-1 and caveolin-2 was identical with the peak time of permeability of the BTB and the density of pinocytotic vesicles. All of these results strongly indicated that VEGF increased permeability of the BTB caused by enhancement of the density of pinocytotic vesicles, and the molecular mechanism might be associated with upregulated expression of caveolin-1 and caveolin-2.


Asunto(s)
Neoplasias Encefálicas/patología , Caveolas/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Glioma/patología , Transcitosis/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiología , Caveolas/ultraestructura , Caveolina 1/metabolismo , Caveolina 2/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Permeabilidad , Distribución Aleatoria , Ratas , Ratas Wistar , Transcitosis/fisiología
18.
Br J Neurosurg ; 19(4): 322-6, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16455538

RESUMEN

In clinical practice there is a difference in response of the blood-tumour barrier (BTB) permeability induced by bradykinin in brain tumours with the same pathology. The variability in response of tumours to bradykinin is likely to be related to the expression level of bradykinin B(2) receptor. This study used fresh human glioma samples to determine the expression level of bradykinin B(2) receptor on gliomas with different pathological grades. The grade of tumour was classified using the WHO classification. To determine the bradykinin B(2) receptor expression level in gliomas, Immunohistochemistry and Western blot methods were used. In 24 cases of gliomas there were eight cases of WHO I glioma, eight cases of WHO II glioma and eight cases of WHO III glioma. Both Western blot and immunohistochemistry showed bradykinin B(2) receptors localized on tumour cells, whilst brain cells at the edge of the glioma hardly expressed B(2) receptor. There were significant differences of bradykinin B(2) receptor expression level among different pathological grades of glioma. The expression of B(2) receptor in the three grades of glioma was in the order of WHO I < WHO II < WHO III. Determination of bradykinin B(2) receptor expression level in human glioma may be useful in screening glioma patients to predict whether they will be suitable for opening of the blood - tumour barrier with bradykinin or its analogue.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Receptor de Bradiquinina B2/metabolismo , Adolescente , Adulto , Anciano , Western Blotting , Neoplasias Encefálicas/patología , Niño , Preescolar , Femenino , Glioma/patología , Humanos , Técnicas para Inmunoenzimas , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...