Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(13)2023 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-37445896

RESUMEN

Periodontitis is an inflammatory disease caused by microorganisms that induce the destruction of periodontal tissue. Inflamed and damaged tissue produces various inflammatory cytokines, which activate osteoclasts and induce alveolar bone loss and, eventually, tooth loss. Sirt6 expression suppresses inflammation and bone resorption; however, its role in periodontitis remains unclear. We hypothesized that Sirt6 has a protective role in periodontitis. To understand the role of Sirt6 in periodontitis, we compared periodontitis with ligature placement around the maxillary left second molar in 8-week-old control (C57BL/6J) male mice to Sirt6-overexpressing Tg (Sirt6Tg) mice, and we observed the resulting phenotypes using micro-CT. MDL801, a Sirt6 activator, was used as a therapy for periodontitis through oral gavage. Pro-inflammatory cytokines and increased osteoclast numbers were observed in alveolar bone tissue under periodontitis surgery. In the same condition, interestingly, protein levels from Sirt6 were the most downregulated among sirtuins in alveolar bone tissue. Based on micro-CT and CEJ-ABC distance, Sirt6Tg was observed to resist bone loss against ligature-induced periodontitis. Furthermore, the number of osteoclasts was significantly reduced in Sirt6Tg-ligated mice compared with control-ligated mice, although systemic inflammatory cytokines did not change. Consistent with this observation, we confirmed that bone loss was significantly reduced when MDL801, a Sirt6 activator, was included in the ligation mouse model. Our findings demonstrate that Sirt6 activation prevents bone loss against ligature-induced periodontitis. Thus, a Sirt6 activator may provide a new therapeutic approach for periodontitis.


Asunto(s)
Pérdida de Hueso Alveolar , Periodontitis , Sirtuinas , Ratones , Masculino , Animales , Ratones Endogámicos C57BL , Periodontitis/metabolismo , Inflamación/complicaciones , Pérdida de Hueso Alveolar/etiología , Pérdida de Hueso Alveolar/prevención & control , Osteoclastos/metabolismo , Modelos Animales de Enfermedad , Citocinas/metabolismo , Sirtuinas/genética
2.
Biochem Biophys Res Commun ; 533(4): 1198-1203, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33059920

RESUMEN

The small GTPase Rheb binds and activates mTORC1, which plays a pivotal role in diverse cellular physiologies. To increase our understanding of how Rheb regulates mTORC1 signaling, we set out to identify Rheb binding proteins using shotgun proteomics approaches. In this study, we characterized HSP70, one of the identified proteins, as a new Rheb binding protein. The present study showed that Rheb forms a complex with HSP70 in intact cells. Interestingly, the binding of Rheb to mTORC1 was abolished by HSP70. Furthermore, the stability of Rheb is dramatically decreased by HSP70, and this degradation is proteasome-dependent. As a result, Rheb-dependent mTORC1 activation was decreased by HSP70. Taken together, HSP70 dissociates Rheb from mTORC1 and induces proteasome-dependent degradation, leading to the inhibition of mTORC1 signaling. Our findings suggest that HSP70 is a negative regulator of mTORC1 signaling via interaction with Rheb.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Células HEK293 , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Estabilidad Proteica , Transducción de Señal
3.
J Cell Physiol ; 232(12): 3444-3453, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28098340

RESUMEN

The aim of this study was to determine the effects and molecular mechanism of blue light emitting diode (LED) in tumor cells. A migration and invasion assay for the metastatic behavior of mouse colon cancer CT-26 and human fibrosarcoma HT-1080 cells was performed. Cancer cell migration-related proteins were identified by obtaining a 2-dimensional gel electrophoresis (2-DE) in total cellular protein profile of blue LED-irradiated cancer cells, followed by matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) analysis of proteins. Protein levels were examined by immunoblotting. Irradiation with blue LED inhibited CT-26 and HT-1080 cell migration and invasion. The anti-metastatic effects of blue LED irradiation were associated with inhibition of matrix metalloproteinase (MMP)-2 and MMP-9 expression. P38 MAPK phosphorylation was increased in blue LED-irradiated CT-26 and HT-1080 cells, but was inhibited after pretreatment with SB203580, a specific inhibitor of p38 MAPK. Inhibition of p38 MAPK phosphorylation by SB203580 treatment increased number of migratory cancer cells in CT-26 and HT-1080 cells, indicating that blue LED irradiation inhibited cancer cell migration via phosphorylation of p38 MAPK. Additionally blue LED irradiation of mice injected with CT-26 cells expressing luciferase decreased early stage lung metastasis compared to untreated control mice. These results indicate that blue LED irradiation inhibits cancer cell migration and invasion in vitro and in vivo.


Asunto(s)
Movimiento Celular/efectos de la radiación , Neoplasias del Colon/terapia , Fibrosarcoma/terapia , Luz , Fototerapia/métodos , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Electroforesis en Gel Bidimensional , Femenino , Fibrosarcoma/enzimología , Fibrosarcoma/patología , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteómica/métodos , Transducción de Señal/efectos de la radiación , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
J Nanosci Nanotechnol ; 15(1): 125-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26328315

RESUMEN

To investigate the possibility of drug targeting via the transferrin receptor-mediated pathway, iron-saturated transferrin was conjugated with chitosan (Tr-chitosan) and complexed with doxorubicin-conjugated methoxy poly(ethylene glycol)-b-dextran succinate (DEX-DOX). DEX-DOX nanoparticles have spherical morphologies with less than 150 nm particle sizes. When Tr-chitosan was complexed with DEX-DOX nanoparticles (TR nanoparticle), particle sizes were increased to higher than 200 nm. Viability of 9L cells with treatment of doxorubicin (DOX) or DEX-DOX nanoparticle was dose-dependently decreased regardless of transferrin receptor blocking. However, cytotoxicity of TR nanoparticles was reduced by blocking of transferrin receptor. Flow cytometric analysis and confocal microscopic observation showed that fluorescence intensity of tumor cells with treatment of TR nanoparticles was significantly decreased by blocking of transferring receptor while DEX-DOX nanoparticles were not affected by blocking of transferring receptor. These results indicated that TR nanoparticles are promising candidates for brain tumor drug delivery.


Asunto(s)
Dextranos/química , Sistemas de Liberación de Medicamentos , Nanopartículas/química , Transferrina/farmacocinética , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quitosano/química , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Endocitosis , Nanopartículas/toxicidad , Tamaño de la Partícula , Ratas , Receptores de Transferrina/metabolismo , Transferrina/química
5.
Childs Nerv Syst ; 31(11): 2063-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26293677

RESUMEN

PURPOSE: Pilocytic astrocytoma (PA) is a World Health Organization grade I neoplasm that generally follows a benign course. However, in some patients, PA exhibits an aggressive clinical course. Here, we examined the clinical course of pediatric and adult PAs with progression at a single institution. METHODS: Between 1995 and 2013, 39 patients with PA were treated. Nineteen were pediatric patients (mean age, 12 years; range, 1-17 years) with a male-to-female patient ratio of 10:9, while 20 were adults (mean age, 36.4 years; range, 19-65 years) with a male-to-female ratio of 9:11. We analyzed and compared tumor location, extent of tumor resection, adjuvant treatment, and clinical course in all patients. RESULTS: In the 19 pediatric patients, tumors were located in the cerebellar vermis, cerebellar hemisphere, optic pathways plus hypothalamus, hypothalamus, brainstem, and the temporal lobe in 6 (31.6%), 5 (26.3%), 3 (15.8%), 2 (10.5%), and 2 (10.5%) patients and 1 (5.3%) patient, respectively. The mass was totally, subtotally, or partially resected in 11 (57.9%), 2 (10.5%), and 4 (21.1%) patients, respectively; biopsies were performed in 2 (10.5%) patients. Immediate postoperative adjuvant treatment was carried out in 6 patients. Tumor progression was detected in 3 patients at 3.0, 4.6, and 5.2 years after treatment, respectively, without significant symptoms. In the 20 adult patients, tumors were located in the cerebellar hemisphere, cerebellar vermis, hypothalamus, brainstem, cerebral hemisphere, and lateral ventricle in 5 (25%), 4 (20%), 3 (15%), 3 (15%), 3 (15%), and 2 (10%) patients, respectively. The mass was totally, subtotally, or partially resected in 11 (55%) and 6 (30%) patients and 1 (5%) patient, respectively; biopsies were performed in 2 patients. Immediate adjuvant treatment was carried out in 2 patients. Progression was detected in 3 patients at 0.3, 0.9, and 2.5 years after treatment, respectively, with progressive neurologic symptoms. There was one case of disease-related mortality during follow-up among the adult patients. CONCLUSION: Most of the PA cases evaluated in this study were benign. However, tumor progression in adult PAs followed a more aggressive clinical course than those in pediatric PAs.


Asunto(s)
Envejecimiento , Astrocitoma/diagnóstico , Astrocitoma/terapia , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/terapia , Adolescente , Adulto , Anciano , Niño , Preescolar , Manejo de la Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
6.
Acta Neurochir (Wien) ; 156(12): 2263-73, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25238986

RESUMEN

OBJECTIVE: The dural tail sign was first described as a thin, tapering rim of dural enhancement, in continuity with meningiomas on enhanced T1-weighted magnetic resonance (MR) images. However, the exact nature of the dural tail is still unclear. This study investigated the immunohistochemical (IHC) characteristics of the dural tail in intracranial meningiomas and the correlation between clinicopathological profiles and tumor invasion of the dural tail. METHODS: The study group consisted of 36 patients of meningioma with the dural tail noted on MR imaging and in pathological findings, and 18 patients of meningioma without the dural tail as the control group. IHC staining of tumor masses and dural tails for vascular endothelial growth factor (VEGF), epithelial membrane antigen, CD34, Ki-67, and vimentin were performed. RESULTS: The data showed that 61.1 % (22/36) of cases in the study group revealed tumor invasion of dural tail, and 55.6 % (30/54) of all the cases demonstrated dura mater invasion in all the samples. The dura mater invasion was significantly positively related to invasion of the dural tail in the study group (p = 0.009). IHC staining detected higher expression of VEGF and CD34 in the dural tail than in the main tumor mass. CONCLUSIONS: Considering the high proportion of patients with tumor invasion into the dural tail, we tried to perform wide resection of the dural tail during intracranial meningioma surgery. Furthermore, VEGF was strongly expressed in tumor cells that invaded into the dural tail, and hence VEGF can be used as a marker to differentiate tumor cells from normal meningeal cells in the dural tail.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Duramadre/metabolismo , Neoplasias Meníngeas/diagnóstico , Meningioma/diagnóstico , Adulto , Anciano , Estudios de Casos y Controles , Duramadre/patología , Femenino , Humanos , Inmunohistoquímica , Imagen por Resonancia Magnética , Masculino , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/cirugía , Meningioma/patología , Meningioma/cirugía , Persona de Mediana Edad , Mucina-1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Vimentina/metabolismo
7.
IEEE Trans Nanobioscience ; 22(4): 845-854, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37022021

RESUMEN

BACKGROUND: Radiosurgery has been recognized as a reasonable treatment for metastatic brain tumors. Increasing the radiosensitivity and synergistic effects are possible ways to improve the therapeutic efficacy of specific regions of tumors. c-Jun-N-terminal kinase (JNK) signaling regulates H2AX phosphorylation to repair radiation-induced DNA breakage. We previously showed that blocking JNK signaling influenced radiosensitivity in vitro and in an in vivo mouse tumor model. Drugs can be incorporated into nanoparticles to produce a slow-release effect. This study assessed JNK radiosensitivity following the slow release of the JNK inhibitor SP600125 from a poly (DL-lactide-co-glycolide) (LGEsese) block copolymer in a brain tumor model. MATERIALS AND METHODS: A LGEsese block copolymer was synthesized to fabricate SP600125-incorporated nanoparticles by nanoprecipitation and dialysis methods. The chemical structure of the LGEsese block copolymer was confirmed by 1H nuclear magnetic resonance (NMR) spectroscopy. The physicochemical and morphological properties were observed by transmission electron microscopy (TEM) imaging and measured with particle size analyzer. The blood-brain barrier (BBB) permeability to the JNK inhibitor was estimated by BBBflammaTM 440-dye-labeled SP600125. The effects of the JNK inhibitor were investigated using SP600125-incorporated nanoparticles and by optical bioluminescence, magnetic resonance imaging (MRI), and a survival assay in a mouse brain tumor model for Lewis lung cancer (LLC)-Fluc cells. DNA damage was estimated by histone γ H2AX expression and apoptosis was assessed by the immunohistochemical examination of cleaved caspase 3. RESULTS: The SP600125-incorporated nanoparticles of the LGEsese block copolymer were spherical and released SP600125 continuously for 24h. The use of BBBflammaTM 440-dye-labeled SP600125 demonstrated the ability of SP600125 to cross the BBB. The blockade of JNK signaling with SP600125-incorporated nanoparticles significantly delayed mouse brain tumor growth and prolonged mouse survival after radiotherapy. γ H2AX, which mediates DNA repair protein, was reduced and the apoptotic protein cleaved-caspase 3 was increased by the combination of radiation and SP600125-incorporated nanoparticles.


Asunto(s)
Neoplasias Encefálicas , Carcinoma Pulmonar de Lewis , Nanopartículas , Ratones , Animales , Carcinoma Pulmonar de Lewis/terapia , Caspasa 3/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Apoptosis
8.
Acta Neurochir (Wien) ; 154(4): 611-20; discussion 620, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22327326

RESUMEN

OBJECT: Although bone invasion and hyperostosis are common phenomena in patients with intracranial meningiomas, the basic pathomechanism is not fully understood. Based on an immunohistochemical study of surgically resected samples with hyperostosis, we postulate a possible mechanism of hyperostosis in patients with intracranial meningiomas. MATERIALS AND METHODS: Forty-six meningiomas were evaluated in this study. Twenty-six meningiomas associated with hyperostosis specimens served as the study group, and 20 meningiomas without any bony changes served as controls. An immunohistochemical staining technique was used to detect the expression of matrix metalloproteinase (MMP)-2, -9, and -13, membrane type (MT)1-MMP, estrogen receptor (ER), and progesterone receptor (PR) in the main tumor and hyperostotic portions of the studied samples. RESULTS: In the non-hyperostosis group, expression of MMP-13, MT1-MMP, and ER was significantly less than in the main tumor portion of hyperostotic meningiomas, while there was no difference in the expression of MMP-2 and -9 and PR in the main tumor between the two groups. In the hyperostosis group, the immunoreactivity of MMP-2 in the hyperostotic portion revealed a higher pattern of expression than the main tumor (p < 0.002). The expression of MMP-9, MT1-MMP, ER, and PR had relatively positive immunoreactivity in the main tumor portion (P < 0.05). CONCLUSIONS: Increased expression of MMP-13 and MT1-MMP in the tumor portion of hyperostosis of meningiomas might contribute to the initiation of osteolysis. Activated MMP-2 in hyperostotic lesions may change the physiological metabolism of the skull bone, thus playing an important role in hyperostosis formation.


Asunto(s)
Hiperostosis/enzimología , Metaloproteinasas de la Matriz/fisiología , Neoplasias Meníngeas/enzimología , Meningioma/enzimología , Cráneo/enzimología , Biomarcadores de Tumor/fisiología , Femenino , Humanos , Hiperostosis/patología , Hiperostosis/fisiopatología , Masculino , Metaloproteinasa 13 de la Matriz/fisiología , Metaloproteinasa 14 de la Matriz/fisiología , Metaloproteinasa 2 de la Matriz/fisiología , Metaloproteinasa 9 de la Matriz/fisiología , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/fisiopatología , Meningioma/patología , Meningioma/fisiopatología , Invasividad Neoplásica/patología , Invasividad Neoplásica/fisiopatología , Osteólisis/enzimología , Osteólisis/patología , Osteólisis/fisiopatología , Cráneo/patología , Cráneo/fisiopatología
9.
Artículo en Inglés | MEDLINE | ID: mdl-32419816

RESUMEN

METHODS: A cytotoxicity assay for BHD was performed using the MTT assay. Following treatment with BHD, mBHD-1, and mBHD-2 in the presence of lipopolysaccharide (LPS), nitric oxide (NO) secretion was detected in cell supernatants using a NO detection kit. The expression of proinflammatory mediators was detected using RT-PCR and western blotting. To verify the mechanism of mBHD, specific inhibitors of JNK (SP600125) or p38 (SB203580) were used for co-treatment with mBHD, and then the changes in NO and nitric oxide synthase (iNOS) were measured. RESULTS: Both mBHD-1 and mBHD-2 showed greater anti-inflammatory effects than BHD. Both mBHD-1 and mBHD-2 inhibited NO secretion and decreased the expression of IL-1ß, IL-6, TNF-α, and iNOS. Treatment with a p38 inhibitor and a JNK inhibitor in mBHD-1- and mBHD-2-treated cells resulted in inhibition of NO and iNOS. CONCLUSION: We provided the first experimental evidence that mBHD may be a more useful anti-inflammatory than BHD. High concentrations or long-term use of BHD may be harmful to inflammatory status. Therefore, the length of treatment and concentration should be considered depending on the targeted disease.

10.
J Neurosurg ; 109(2): 273-84, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18671640

RESUMEN

OBJECT: Galectin-1 is highly expressed in motile cell lines. The authors investigated whether galectin-1 actually modulates the migration and invasion of human glioblastoma multiforme (GBM) cell lines, and whether its expression with respect to invasion and prognosis is attributable to certain glioma subgroups. METHODS: In the human GBM cell lines U343MG-A, U87MG, and U87MG-10', the RNA differential display was evaluated using Genefishing technology. The results were validated by reverse transcription polymerase chain reaction and Northern blot analysis to detect possible genetic changes as the determining factors for the motility of the malignant glioma. The migration and invasion abilities were investigated in human GBM cell lines and galectin-1 transfectant using an in vitro brain slice invasion model and a simple scratch technique. The morphological and cytoskeletal (such as the development of actin and vimentin) changes were examined under light and confocal microscopy. Galectin-1 expression was assessed on immunohistochemical tests and Western blot analysis. RESULTS: Endogenous galectin-1 expression in the human GBM cell lines was statistically correlated with migratory abilities and invasiveness. The U87-G-AS cells became more round than the U87MG cells and lacked lamellipodia. On immunohistochemical staining, galectin-1 expression was increased in higher-grade glioma subgroups (p = 0.027). CONCLUSIONS: Diffuse gliomas demonstrated higher expression levels than pilocytic astrocytoma in the Western blot. Galectin-1 appears to modulate migration and invasion in human glioma cell lines and may play a role in tumor progression and invasiveness in human gliomas.


Asunto(s)
Neoplasias Encefálicas/patología , Neoplasias Encefálicas/fisiopatología , Galectina 1/genética , Glioblastoma/patología , Glioblastoma/fisiopatología , Astrocitoma/metabolismo , Astrocitoma/patología , Astrocitoma/fisiopatología , Northern Blotting , Western Blotting , Neoplasias Encefálicas/metabolismo , División Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Galectina 1/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Humanos , Invasividad Neoplásica , Pronóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
11.
J Pharm Sci ; 97(3): 1268-76, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17674407

RESUMEN

The aim of this study is to prepare cisplatin-incorporated nanoparticles based on ion complex formation between hyaluronic acid (HA) and cisplatin for antitumor drug delivery. To prepare nanoparticles using HA, bulk HA was degraded by hyaluronidases (HAses). Cisplatin-incorporated HA nanoparticles were prepared by mixing cisplatin with an aqueous solution of HA and then the nanoparticle solution was dialyzed to remove trace elements. Since glioma tumor cell lines are able to secrete HAse, extracts from U343MG and U87MG cell lines were used to test the release of cisplatin from the nanoparticles. The morphological observation of the cisplatin-incorporated nanoparticles showed that they had spherical shapes with a particle size around 100-200 nm. The loading efficiency of cisplatin in the nanoparticles was about 67-81% (w/w) and cisplatin was continuously released from the nanoparticles for 4 days. Especially, the release rate of cisplatin from the nanoparticles increased when HAse was added to the release medium. In the results of the HA zymography, the U343MG cell line secreted HAse, while the U87MG cell line did not. When the extracts from U343MG were added to the release medium, the release rate of cisplatin was slightly increased, while the extracts from U87MG did not significantly affect the release rate of cisplatin. In conclusion, cisplatin-incorporated nanoparticles have sufficiently small particle sizes to use as a drug targeting system. The release of cisplatin from the nanoparticles was responsive to the secretion of HAse. These nanoparticles are suitable vehicles for an antitumor drug targeting system.


Asunto(s)
Cisplatino/química , Ácido Hialurónico/química , Nanopartículas , Microscopía Electrónica de Transmisión , Tamaño de la Partícula , Difracción de Rayos X
12.
J Neurosurg ; 106(5): 866-71, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17542531

RESUMEN

OBJECT: The authors evaluated the clinical manifestations and surgical results in patients with cystic vestibular schwannoma (VS), and investigated the matrix metalloproteinase (MMP) expression of the cyst fluid and wall in an attempt to elucidate the pathogenesis and characteristics of this disease. METHODS: The clinical and neuroimaging features, perioperative findings, and surgical outcomes in 24 cases of cystic VS and 82 cases of solid VS, all of which were treated using the suboccipital approach, were retrospectively compared. To evaluate the role of MMP in cystic VS, gelatin zymography and immunohistochemical studies of the cyst fluid, wall, and solid portion were performed in nine cases of this disease. The mean duration of symptoms was shorter (14.0 months compared with 26.1 months; p = 0.04) and the mean size of the tumor was larger (43.8 mm compared with 34.2 mm; p = 0.048) in the cystic than the solid VS group. Although gross-total resection was easier to accomplish in this group (100% compared with 84.1%), adhesion to the facial nerve was more frequent (62.5% compared with 48.8%; p = 0.042). On gelatin zymography studies, MMP-2 expression was ubiquitously observed in all cyst fluids. Immunohistochemical analysis of the cyst wall showed that MMP-2 was apparently localized to the tumor cells on the luminal inner surface, adjacent to the cyst cavity. CONCLUSIONS: Resection of cystic VS is complicated by severe adhesion of the tumor capsule to the facial nerve and the large size of the lesion. The authors believe that MMP-2 may be involved in the pathogenesis of cyst formation or in its enlargement and may aggravate adhesion to the facial nerve, either by promoting the enlargement of the tumor or engendering the degradation of the tumor-nerve barrier proteolytically.


Asunto(s)
Quistes del Sistema Nervioso Central/patología , Quistes del Sistema Nervioso Central/cirugía , Metaloproteinasa 2 de la Matriz/análisis , Metaloproteinasa 9 de la Matriz/análisis , Neuroma Acústico/patología , Neuroma Acústico/cirugía , Adolescente , Adulto , Anciano , Quistes del Sistema Nervioso Central/diagnóstico , Diagnóstico Diferencial , Nervio Facial/patología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Neuroma Acústico/diagnóstico , Estudios Retrospectivos , Adherencias Tisulares/cirugía , Resultado del Tratamiento
13.
J Clin Neurosci ; 14(12): 1192-8, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17964788

RESUMEN

Although several types of brain tumors are commonly associated with cyst formation, the pathogenesis of tumor-associated cysts (TAC) is unknown. We investigated the matrix metalloproteinase (MMP) expression of cyst fluids to elucidate the pathogenesis of TAC in brain tumors. We also examined the relationship between the severity of peritumoral edema and the expression of intracystic MMP. We collected 40 cyst fluid samples from 34 patients with TAC and studied the expression of MMP-2 and -9 in the cyst fluid using gelatin zymography. Radiological studies were used to estimate the severity of the peritumoral edema and to determine the presence of TAC. Although gelatin zymography of the cyst fluid showed high levels of MMPs, there was no correlation between the expression of MMPs in the cyst fluid and that in the tumor tissue. The level of MMP expression in the cyst fluid did not reflect the pathologic grade of the individual tumors. However, the total and activated MMP-9 levels were significantly associated with the severity of the peritumoral edema (p<0.05). These results suggest that MMPs may be partly involved in the pathogenesis of TAC and peritumoral edema in brain tumors.


Asunto(s)
Edema Encefálico/enzimología , Edema Encefálico/patología , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Quistes del Sistema Nervioso Central/enzimología , Quistes del Sistema Nervioso Central/patología , Metaloproteinasas de la Matriz/biosíntesis , Edema Encefálico/etiología , Neoplasias Encefálicas/complicaciones , Quistes del Sistema Nervioso Central/etiología , Humanos , Isoenzimas/biosíntesis , Isoenzimas/líquido cefalorraquídeo , Imagen por Resonancia Magnética , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/líquido cefalorraquídeo , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/líquido cefalorraquídeo , Metaloproteinasas de la Matriz/líquido cefalorraquídeo
14.
J Neurosurg ; 126(6): 1829-1838, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27540907

RESUMEN

OBJECTIVE In patients with glioblastoma, local invasion of tumor cells causes recurrence and shortens survival. The goal of this study was to determine whether protein disulfide isomerase (PDI) A6 regulates migration and invasion of glioblastoma cells and the associated factors. METHODS U87MG cells were treated with either PDIA6 or ADAM17 small interfering RNA (siRNA) fragments or with both types of siRNA fragments, and expression was confirmed by reverse transcription-polymerase chain reaction and Western blot. Migration and invasion were assessed using a wound-healing assay, a Matrigel assay, and an organotypic culture system. After the U87MG cells were treated with siRNAs and epidermal growth factor receptor (EGFR) inhibitors, the expression of matrix metalloproteinase-2 (MMP-2), membrane Type 1-matrix metalloproteinase (MT1-MMP), integrin, phosphorylated focal adhesion kinase (pFAK), and phosphorylated EGFR (pEGFR) was detected by Western blotting and zymography. RESULTS U87MG cell migration and invasion increased significantly after inhibition of PDIA6. The MMP-2 activation ratio and ADAM17 activity (as a sheddase of the proligand) increased, and expression of pEGFR, pFAK, integrin α5ß3, and MT1-MMP was induced, compared with control levels. Furthermore, heparin-binding epidermal growth factor (EGFR signaling ligand) was highly expressed in PDIA6-knockdown cells. After siPDIA6-transfected U87MG cells were treated with EGFR signaling inhibitors, expression of pFAK, MMP-2, and MT1-MMP decreased and invasion decreased significantly. Simultaneous double-knockdown of PDIA6 and ADAM17 reduced pEGFR and pFAK expression, compared with control levels. CONCLUSIONS The authors propose that inhibiting PDIA6 could transduce EGFR signaling by activating and inducing ADAM17 during migration and invasion of U87MG glioblastoma cells. The results of this study suggest that PDIA6 is an important component of EGFR-mediated migration and invasion of U87MG cells. This is the first report of the effects of PDIA6 on migration and invasion in glioblastoma.


Asunto(s)
Proteína ADAM17/metabolismo , Movimiento Celular/fisiología , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Invasividad Neoplásica/patología , Proteína Disulfuro Isomerasas/metabolismo , Proteína ADAM17/genética , Línea Celular Tumoral , Glioblastoma/patología , Humanos , Proteína Disulfuro Isomerasas/genética , ARN Interferente Pequeño
15.
Anticancer Res ; 26(1A): 249-58, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16475705

RESUMEN

BACKGROUND: Despite the well-documented importance of Nm23 in the control of metastasis, there is currently a paucity of data regarding the role of this gene family in the control of glioma invasion. MATERIALS AND METHODS: Nm23-H1 expression in gliomas was assessed via immunohistochemistry, Western blot, RT-PCR and Northern blot analyses. The migration and invasion ability were also investigated in primary glioma culture cells, human glioma cell lines and nm23-H1 transfectant, using an in vitro brain slice invasion model and a simple scratch technique. RESULTS: Although no significant correlations were detected between nm23-H1 expression and pathological grade, the endogenous nm23-H1 expression in gliomas was found to be inversely correlated with their migratory abilities. Additionally, the nm23-H1 transfectant resulted in a reduction of approximately 45% of the migratory ability and suppressed the invasiveness of the parental cell line. CONCLUSION: Our overall findings suggest that nm23-H1 may play an important role in the suppression of glioma invasion and migration.


Asunto(s)
Astrocitoma/genética , Astrocitoma/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Movimiento Celular/genética , Nucleósido-Difosfato Quinasa/genética , Astrocitoma/metabolismo , Neoplasias Encefálicas/metabolismo , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Expresión Génica , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/aislamiento & purificación , Humanos , Nucleósido Difosfato Quinasas NM23 , Invasividad Neoplásica , Nucleósido-Difosfato Quinasa/biosíntesis , Transfección
16.
J Pharm Sci ; 95(11): 2348-60, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16886178

RESUMEN

The goal of this study is to develop novel types of polyion complex micelles for the drug delivery to brain tumor. Methoxy poly(ethylene glycol) (mPEG)-grafted chitosan (CP) was synthesized in order to make polymeric micelles encapsulating all-trans retinoic acid (ATRA) based on polyion complex formation. Polyion complex micelles were found to have spherical shapes with sizes of about 50 approximately 200 nm. The loading efficiency of micelle was higher than 80% (w/w) for all formulations. 1H nuclear magnetic resonance (NMR) spectra confirmed the formation of polymeric micelles. The CP graft copolymer and ATRA have distinguishing peaks in their 1H NMR spectra. The specific peaks of ATRA disappeared in D2O or DMSO while it appeared at mixtures of D2O/DMSO, indicating that ATRA and chitosan formed ion complex inner-core. In the cell cytotoxicity study using U87MG cells in vitro, polyion complex micelles showed similar cytotoxicity to that of free ATRA. A migration test was performed to investigate the inhibition of tumor cell invasion in vitro. The results suggested that the polyion complex micelles was more effective at inhibiting tumor cell migration than free ATRA.


Asunto(s)
Antineoplásicos/química , Quitosano/química , Polietilenglicoles/química , Tretinoina/química , Adsorción , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Secuencia de Carbohidratos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dimetilsulfóxido , Portadores de Fármacos , Composición de Medicamentos , Sistemas de Liberación de Medicamentos , Excipientes , Glioma/tratamiento farmacológico , Humanos , Espectroscopía de Resonancia Magnética , Micelas , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Sistema Mononuclear Fagocítico , Nanopartículas , Invasividad Neoplásica , Tamaño de la Partícula , Tretinoina/administración & dosificación , Tretinoina/farmacología
17.
J Korean Neurosurg Soc ; 59(2): 106-16, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26962415

RESUMEN

OBJECTIVE: Protein disulfide isomerase (PDI) acts as a chaperone on the cell surface, and it has been reported that PDI is associated with the tumor cell migration and invasion. The aims of this study are to investigate the anti-migration effect of bacitracin, which is an inhibitor of PDI, and the associated factor in this process. METHODS: U87-MG glioma cells were treated with bacitracin in 1.25, 2.5, 3.75, and 5.0 mM concentrations. Western blot with caspase-3 was applied to evaluate the cytotoxicity of bacitracin. Adhesion, morphology, migration assays, and organotypic brain-slice culture were performed to evaluate the effect of bacitracin to the tumor cell. Western blot, PCR, and gelatin zymography were performed to investigate the associated factors. Thirty glioma tissues were collected following immunohistochemistry and Western blot. RESULTS: Bacitracin showed a cytotoxicity in 3rd (p<0.05) and 4th (p<0.001) days, in 5.0 Mm concentration. The cell adhesion significantly decreased and the cells became a round shape after treated with bacitracin. The migration ability, the expression of phosphorylated focal adhesion kinase (p-FAK) and matrix metalloproteinase-2 (MMP-2) decreased in a bacitracin dose- and time-dependent manner. The U87-MG cells exhibited low-invasiveness in the 2.5 mM, compared with the untreated in organotypic brain-slice culture. PDI was expressed in the tumor margin, and significantly increased with histological glioma grades (p<0.001). CONCLUSION: Bacitracin, as a functional inhibitor of PDI, decreased the phosphorylated FAK and the secreted MMP-2, which are the downstream of integrin and play a major role in cell migration and invasion, might become one of the feasible therapeutic strategies for glioblastoma.

18.
Oncol Rep ; 36(6): 3397-3404, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27779695

RESUMEN

Stereotactic radiosurgery has been recognized as an effective treatment approach for metastatic brain tumors. By increasing the sensitivity of the tumor to radiation and decreasing the marginal dose, it is possible to improve therapeutic efficacy and decrease side-effects. In radiation-induced cells, c-Jun N-terminal kinase (JNK) signaling mediates the phosphorylation of H2AX, which indicates DNA damage sensitivity and modulates the effect of radiation. Lewis lung cancer (LLC) and breast cancer (4T1) cells were irradiated with a Gamma Knife in cell culture tubes. To evaluate the relationship between radiosensitivity and JNK activity, clonogenic assay was performed. DNA damage response was estimated by γH2AX focus formation assay and apoptosis­related protein levels were assessed by western blotting. The mice were subcutaneously inoculated with LLC cells, and irradiated concomitantly with JNK inhibitor treatment. The effect of the JNK inhibitor was investigated by tumor volumetry and immunohistochemistry. γH2AX expression, which mediates repair of radiation­induced DNA damage, was reduced in the cancer cell group pretreated with the JNK inhibitor. This finding shows that JNK inhibition may increase the radiosensitivity in radiated lung and breast cancer cells. For the in vivo study, irradiated tumor growth was significantly delayed in the JNK inhibitor-treated mouse group. Blockade of JNK signaling decreased γH2AX expression and increased apoptosis in the radiation-induced cancer cells. JNK inhibitor may be useful for enhancing the radiosensitivity of lung and breast cancer cells and improving the treatment efficacy of radiosurgical approaches for metastatic brain tumors.


Asunto(s)
Antracenos/farmacología , Antineoplásicos/farmacología , Carcinoma Pulmonar de Lewis/enzimología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Animales , Apoptosis , Carcinoma Pulmonar de Lewis/terapia , Línea Celular Tumoral , Quimioradioterapia , Relación Dosis-Respuesta en la Radiación , Femenino , Histonas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Fosforilación , Procesamiento Proteico-Postraduccional , Tolerancia a Radiación , Transducción de Señal
19.
J Korean Neurosurg Soc ; 59(6): 551-558, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27847566

RESUMEN

Malignant glioma cells invading surrounding normal brain are inoperable and resistant to radio- and chemotherapy, and eventually lead to tumor regrowth. Identification of genes related to motility is important for understanding the molecular biological behavior of invasive gliomas. According to our previous studies, Metallothionein 1E (MT1E) was identified to enhance migration of human malignant glioma cells. The purpose of this study was to confirm that MT1E could modulate glioma invasion in vivo. Firstly we established 2 cell lines; MTS23, overexpressed by MT1E complementary DNA construct and pV12 as control. The expression of matrix metalloproteinases (MMP)-2, -9 and a disintegrin and metalloproteinase 17 were increased in MTS23 compared with pV12. Furthermore it was confirmed that MT1E could modulate MMPs secretion and translocation of NFkB p50 and B-cell lymphoma-3 through small interfering ribonucleic acid knocked U87MG cells. Then MTS23 and pV12 were injected into intracranial region of 5 week old male nude mouse. After 4 weeks, for brain tissues of these two groups, histological analysis, and immunohistochemical stain of MMP-2, 9 and Nestin were performed. As results, the group injected with MTS23 showed irregular margin and tumor cells infiltrating the surrounding normal brain, while that of pV12 (control) had round and clear margin. And regrowth of tumor cells in MTS23 group was observed in another site apart from tumor cell inoculation. MT1E could enhance tumor proliferation and invasion of malignant glioma through regulation of activation and expression of MMPs.

20.
Oncol Rep ; 35(6): 3395-402, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27109183

RESUMEN

Nogo or reticulon-4 (RTN4), also known as neurite outgrowth inhibitor, is a member of the reticulon family of genes. Nogo-A, one of the three isoforms, is enriched in the central nervous system (CNS). The extracellular domain of Nogo-A, Nogo-66, has neurite growth inhibitory activity that is specific for neurons and is mediated by the Nogo receptor. However, most of its functions are not known yet. We investigated whether Nogo-A modulates the migration and invasion of a glioblastoma cell line, as well as the factors that have an effect on Nogo-A. The expression of Nogo-A was evaluated using western blotting and immunohistochemistry in human brain tumor specimens. U87MG cells were transfected with a sense-Nogo-A cDNA construct (U87-Nogo-A cells expressing Nogo-A) and an empty vector (U87MG-E cells not expressing Nogo-A). The migration and invasion abilities of these cells were investigated using simple scratch and Matrigel invasion assays. Morphologic and cytoskeletal changes were documented by confocal microscopy. The proliferation rate was estimated using doubling time assay. The effects of Nogo-A on Rho activity and phosphorylated cofilin were determined by a Rho activity assay and western blotting. Among primary brain tumors, Nogo-A expression was found in a higher percentage of oligodendrogliomas (90.0%) compared with the percentage in the glioblastomas (68.4%). In addition, the percentage in mixed gliomas was 42.9%, while it was not expressed in pituitary adenomas or schwannomas. The migration and invasion abilities of the U87-Nogo-A cells were decreased compared with the control. In the U87-Nogo-A cell line, Rho activity and phosphorylated cofilin expression were also decreased and morphology became more flat in comparison with the U87MG-E cell line. Nogo-A may inhibit the migration and invasion of human malignant glioma cells via the downregulation of RhoA-cofilin signaling.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Proteínas Nogo/fisiología , Factores Despolimerizantes de la Actina/metabolismo , Actinas/análisis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Invasividad Neoplásica , Proteínas Nogo/análisis , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA