Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
IEEE Trans Nanobioscience ; 22(4): 845-854, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37022021

RESUMO

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.


Assuntos
Neoplasias Encefálicas , Carcinoma Pulmonar de Lewis , Nanopartículas , Camundongos , Animais , Carcinoma Pulmonar de Lewis/terapia , Caspase 3/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Apoptose
2.
Artigo em Inglês | MEDLINE | ID: mdl-32419816

RESUMO

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.

3.
Macromol Biosci ; 17(5)2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27906510

RESUMO

Melanin is an effective absorber of light and can extend to near infrared (NIR) regions. In this study, a natural melanin is presented as a photothermal therapeutic agent (PTA) because it provides a good photothermal conversion efficiency, shows biodegradability, and does not induce long-term toxicity during retention in vivo. Poloxamer solution containing melanin (Pol-Mel) does not show any precipitation and shows sol-gel transition at body temperature. After irradiation from 808 nm NIR laser at 1.5 W cm-2 for 3 min, the photothermal conversion efficiency of Pol-Mel is enough to kill cancer cells in vitro and in vivo. The tumor growth of mice bearing CT26 tumors treated with Pol-Mel injection and laser irradiation is suppressed completely without recurrence postirradiation. All these results indicate that Pol-Mel can become an attractive PTA for photothermal cancer therapy.


Assuntos
Hidrogéis/química , Hipertermia Induzida/métodos , Melaninas/uso terapêutico , Neoplasias/terapia , Fototerapia/métodos , Animais , Linhagem Celular Tumoral , Humanos , Raios Infravermelhos , Masculino , Melaninas/administração & dosagem , Melaninas/química , Camundongos , Camundongos Endogâmicos BALB C , Poloxâmero , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Neurosurg ; 126(6): 1829-1838, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27540907

RESUMO

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.


Assuntos
Proteína ADAM17/metabolismo , Movimento Celular/fisiologia , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Invasividade Neoplásica/patologia , Isomerases de Dissulfetos de Proteínas/metabolismo , Proteína ADAM17/genética , Linhagem Celular Tumoral , Glioblastoma/patologia , Humanos , Isomerases de Dissulfetos de Proteínas/genética , RNA Interferente Pequeno
5.
J Korean Neurosurg Soc ; 59(6): 551-558, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27847566

RESUMO

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.

6.
Oncol Rep ; 36(6): 3397-3404, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27779695

RESUMO

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.


Assuntos
Antracenos/farmacologia , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Lewis/enzimologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Animais , Apoptose , Carcinoma Pulmonar de Lewis/terapia , Linhagem Celular Tumoral , Quimiorradioterapia , Relação Dose-Resposta à Radiação , Feminino , Histonas/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Fosforilação , Processamento de Proteína Pós-Traducional , Tolerância a Radiação , Transdução de Sinais
7.
Oncol Rep ; 35(6): 3395-402, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27109183

RESUMO

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.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Proteínas Nogo/fisiologia , Fatores de Despolimerização de Actina/metabolismo , Actinas/análise , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Humanos , Invasividade Neoplásica , Proteínas Nogo/análise , Proteínas rho de Ligação ao GTP/metabolismo
8.
J Korean Neurosurg Soc ; 59(2): 106-16, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26962415

RESUMO

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.

9.
J Nanosci Nanotechnol ; 15(1): 125-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26328315

RESUMO

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.


Assuntos
Dextranos/química , Sistemas de Liberação de Medicamentos , Nanopartículas/química , Transferrina/farmacocinética , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quitosana/química , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacologia , Endocitose , Nanopartículas/toxicidade , Tamanho da Partícula , Ratos , Receptores da Transferrina/metabolismo , Transferrina/química
10.
Radiat Oncol ; 10: 164, 2015 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-26245666

RESUMO

BACKGROUND: Glioblastoma is a highly lethal neoplasm that frequently recurs locally after radiotherapy, and most of these recurrences originate from near the irradiated target field. In the present study, we identified the effects of radiation on glioma invasion and p53, TIMP-2, and MMP-2 expression through in vitro and in vivo experiments. METHODS: The U87MG (wt p53) and U251 (mt p53) human malignant glioma cell lines were prepared, and the U2OS (wt 53) and Saos2 (del p53) osteosarcoma cell lines were used as p53 positive and negative controls. The four cell lines and p53 knock-downed U87MG cells received radiation (2-6 Gy) and were analyzed for expression of p53 and TIMP-2 by Western blot, and MMP-2 activity was detected by zymography. In addition, the effects of irradiation on directional invasion of malignant glioma were evaluated by implanting nude mice with bioluminescent u87-Fluc in vivo followed by MMP-2, p53, and TIMP-2 immunohisto-chemistry and in situ zymography. RESULTS: MMP-2 activity and p53 expression increased in proportional to the radiation dose in cell lines with wt p53, but not in the cell lines with del or mt p53. TIMP-2 expression did not increase in U87MG cells. MMP-2 activity decreased in p53 knock-downed U87MG cells but increased in the control group. Furthermore, radiation enhanced MMP-2 activity and increased tumor margin invasiveness in vivo. Tumor cells invaded by radiation overexpressed MMP-2 and p53 and revealed high gelatinolytic activity compared with those of non-radiated tumor cells. CONCLUSION: Radiation-induced upregulation of p53 modulated MMP-2 activity, and the imbalance between MMP-2 and TIMP-2 may have an important role in glioblastoma invasion by degrading the extracellular matrix. Bioluminescent "U87-Fluc"was useful for observing tumor formation without sacrifice after implanting tumor cells in the mouse brain. These findings suggest that the radiotherapy involved field for malignant glioma needs to be reconsidered, and that future trials should investigate concurrent pharmacologic therapies that inhibit invasion associated with radiotherapy.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Metaloproteinase 2 da Matriz/metabolismo , Transdução de Sinais/efeitos da radiação , Proteína Supressora de Tumor p53/metabolismo , Animais , Western Blotting , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Glioma/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica/patologia , RNA Interferente Pequeno , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Childs Nerv Syst ; 31(11): 2063-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26293677

RESUMO

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.


Assuntos
Envelhecimento , Astrocitoma/diagnóstico , Astrocitoma/terapia , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/terapia , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Gerenciamento Clínico , Progressão da Doença , Feminino , Humanos , Lactente , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Adulto Jovem
12.
Oncotarget ; 6(5): 3240-53, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25605251

RESUMO

KITENIN (KAI1 COOH-terminal interacting tetraspanin) promotes tumor invasion and metastasis in various cancers. This study assessed the association between KITENIN expression and advanced glioma grade in patients. In vitro assays revealed that KITENIN knockdown inhibited the invasion and migration of glioma cells, whereas KITENIN overexpression promoted their invasion and migration. In orthotopic mouse tumor models, mice transplanted with KITENIN-transfected glioma cells had significantly shorter survival than mice transplanted with mock-transfected cells. Patients with low KITENIN expression showed a significantly longer progression-free survival than patients with high KITENIN expression. KITENIN induced the expression of the epithelial-mesenchymal transition (EMT) markers (N-cadherin, ZEB1, ZEB2, SNAIL and SLUG) as well as the glioma stemness markers (CD133, ALDH1 and EPH-B1). Taken together, these findings showed that high levels of KITENIN increased glioma invasiveness and progression, associated with the up-regulation of EMT and stemness markers.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/metabolismo , Proteínas de Transporte/metabolismo , Movimento Celular , Transição Epitelial-Mesenquimal , Glioma/metabolismo , Proteínas de Membrana/metabolismo , Células-Tronco Neoplásicas/metabolismo , Animais , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Feminino , Glioma/genética , Glioma/mortalidade , Glioma/patologia , Glioma/cirurgia , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica , Células-Tronco Neoplásicas/patologia , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Transfecção , Regulação para Cima
13.
Acta Neurochir (Wien) ; 156(12): 2263-73, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25238986

RESUMO

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.


Assuntos
Biomarcadores Tumorais/metabolismo , Dura-Máter/metabolismo , Neoplasias Meníngeas/diagnóstico , Meningioma/diagnóstico , Adulto , Idoso , Estudos de Casos e Controles , Dura-Máter/patologia , Feminino , Humanos , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Masculino , Neoplasias Meníngeas/patologia , Neoplasias Meníngeas/cirurgia , Meningioma/patologia , Meningioma/cirurgia , Pessoa de Meia-Idade , Mucina-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Vimentina/metabolismo
14.
Brain Tumor Res Treat ; 2(1): 22-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24926468

RESUMO

BACKGROUND: To investigate the molecular basis for invasion of malignant gliomas, proteomic analysis approach was carried out using two human glioma cell lines, U87MG and U343MG-A that demonstrate different motility and invasiveness in in vitro experiments. METHODS: High-resolution two-dimensional gel electrophoresis and matrix-assisted laser-desorption/ionization time-of-flight mass spectrometry analysis were performed. RESULTS: Nine distinct protein spots that were recognized with significant alteration between the two cell lines. Five of these protein spots were up-regulated in U87MG and four were up-regulated in U343MG-A. CONCLUSION: Among these proteins, cathepsin D was shown to be one of the important proteins which are related with glioma invasion. However, further studies are necessary to reveal the exact role and mechanism of cathepsin D in glioma invasion.

15.
Int J Nanomedicine ; 8: 4351-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24231999

RESUMO

BACKGROUND: In this study, 293T cells were genetically engineered to secrete tissue inhibitor of metalloproteinase-2 (TIMP2) and encapsulated into alginate microcapsules to continuously release TIMP2 protein. METHODS: The anti-invasive potential of the microcapsules was studied in vitro using brain tumor cells. The TIMP2 gene was transfected to 293T cells, and genetically engineered 293TIMP2 cells were encapsulated into alginate microcapsules. Release of TIMP2 protein was detected with Western blot analysis and the anti-invasive potential against U87MG cells was tested using gelatin zymography and a Matrigel assay. RESULTS: Cell viability within the alginate microcapsules was maintained at a cell density of 5 × 10(6). Because polycationic polymers are helpful for maintaining the mechanical strength of microcapsules with good cell viability, the alginate microcapsules were reinforced with chitosan (0.1% w/v). Expression of TIMP2 protein in cell lysates and secretion of TIMP2 into the conditioned medium was confirmed by Western blot analysis. Alginate microcapsules encapsulating 293TIMP2 cells released TIMP2 protein into the medium efficiently, where the TIMP2 protein participated in degradation of the matrix metalloproteinase-2 enzyme and inhibited invasion of U87MG cells. CONCLUSION: Alginate microcapsules encapsulating 293TIMP2 cells are promising candidates for anti-invasive treatment of glioma.


Assuntos
Alginatos/química , Cápsulas/química , Engenharia Genética/métodos , Inibidor Tecidual de Metaloproteinase-2/genética , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Cápsulas/metabolismo , Engenharia Celular , Ácido Glucurônico/química , Células HEK293 , Ácidos Hexurônicos/química , Humanos , Tamanho da Partícula , Transfecção
16.
Neurol Res ; 34(9): 893-900, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22925547

RESUMO

PURPOSE: To assess the influence of 14-3-3-beta in modulating the migration and invasion of human glioma cells. METHODS: To profile the genes associated with malignant glioma cell motility, differential display-polymerase chain reaction was performed and the findings were validated by Northern blotting in the U343MG-A, U87MG, and U87MG-10' human glioma cell lines. Antisense 14-3-3-beta cDNA plasmid was transfected into U87MG ('U87-YA-3'). To follow motility changes after transfection, simple scratch test and matrigel assay were performed. Morphological and cytoskeletal changes were documented by light and confocal microscopy. In addition, doubling times of the transfectant and endogenous 14-3-3-beta levels were determined in various glioma cell lines with different motilities. RESULTS: 14-3-3-beta was highly expressed in U87MG cells. U87-YA-3 cells became small and flat, and actin was depolarized. Furthermore, U87-YA-3 cell motility was inhibited markedly versus parental U87MG cells. The doubling times of transfected and parent cells were 32 and 37 hours, respectively. Endogenous 14-3-3-beta expression in the human glioma cell lines was proportional to their migratory and invasive abilities. CONCLUSION: 14-3-3-beta modulates the migration and invasion in U87MG cells, which may be useful in developing therapeutic approaches for the treatment of glioma.


Assuntos
Proteínas 14-3-3/genética , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteínas 14-3-3/metabolismo , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Glioma/patologia , Humanos , Invasividade Neoplásica/genética , Reprodutibilidade dos Testes , Fatores de Tempo , Transfecção
17.
Int J Oncol ; 41(4): 1305-13, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22843066

RESUMO

Metallothionein 1E (MT1E) has been found to be highly expressed in motile cell lines. We investigated whether MT1E actually modulates the migration and invasion of human glioma cell lines and the types of factors that have an effect on MT1E. RNA differential display was performed using Genefishing™ technology in the human glioma cell lines U343MG-A, U87MG and U87MG-10'; the results were validated by RT-PCR and northern blot analysis, in order to detect possible genetic changes as the determining factors for migration ability in malignant glioma. MT1E was identified in U87MG, a highly motile cell line. The migration and invasion abilities of human glioma cell lines, and MT1E transfectants were investigated using simple scratch testing and Matrigel invasion assays. Morphological and cytoskeletal (actin, vimentin) changes were documented by light and confocal microscopy. The expression of MT1E in four glioma cell lines was assessed by RT-PCR and western blotting. In addition, the effects of MT1E on the activity of the NF-κB p50/p65 transcription factor, MMP-2 and -9 were examined by western blotting and zymography. The endogenous MT1E expression in the human glioma cell lines was statistically correlated with their migratory abilities and invasion. The U87-MT-AS cells became more round and had decreased stress fibers, compared with the U87MG cells. Endogenous MT1E expression in the four human glioma cell lines was directly correlated with migration. Two antisense MT1E-transfected cell lines showed decreased NF-κB p50 translocation into the nucleus, which led to decreased activity of MMP-9 in conditioned media. It may be postulated that MT1E can enhance the migration and invasion of human glioma cells by inducing MMP-9 inactivation via the upregulation of NF-κB p50.


Assuntos
Neoplasias Encefálicas/genética , Movimento Celular/genética , Glioma/genética , Metalotioneína/genética , Invasividade Neoplásica/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Metalotioneína/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Ativação Transcricional , Transfecção
18.
Acta Neurochir (Wien) ; 154(4): 611-20; discussion 620, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22327326

RESUMO

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.


Assuntos
Hiperostose/enzimologia , Metaloproteinases da Matriz/fisiologia , Neoplasias Meníngeas/enzimologia , Meningioma/enzimologia , Crânio/enzimologia , Biomarcadores Tumorais/fisiologia , Feminino , Humanos , Hiperostose/patologia , Hiperostose/fisiopatologia , Masculino , Metaloproteinase 13 da Matriz/fisiologia , Metaloproteinase 14 da Matriz/fisiologia , Metaloproteinase 2 da Matriz/fisiologia , Metaloproteinase 9 da Matriz/fisiologia , Neoplasias Meníngeas/patologia , Neoplasias Meníngeas/fisiopatologia , Meningioma/patologia , Meningioma/fisiopatologia , Invasividade Neoplásica/patologia , Invasividade Neoplásica/fisiopatologia , Osteólise/enzimologia , Osteólise/patologia , Osteólise/fisiopatologia , Crânio/patologia , Crânio/fisiopatologia
19.
J Korean Neurosurg Soc ; 50(3): 157-65, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22102942

RESUMO

OBJECTIVE: We have introduced a method of characterization of invading glioma cells by using molecular analysis of marginal invading tumor cells and molecular profiles of glioma tumor margin. METHODS: Each of tumor core and marginal tissues was obtained in 22 glioma patients. Tumor core cells and marginal cells from each glial tumor were collected by laser capture microdissection or intraoperative microdissection under the operating microscope. Expression of MMP-2, MMP-9, CD44 and RHAMM mRNA by invading glioma cells compared with tumor core was confirmed by realtime-PCR of twenty-four glioma specimens. Clinical data also were reviewed for invasion and recurrence pattern of the gliomas radiologically and invasive rim pattern microscopically. RESULTS: Overall results of the molecular analysis showed that relative overexpression of MMP-2, MMP-9 and RHAMM were noted at the invasive edge of human glioma specimens comparing to the tumor core but CD44 was highly expressed in the tumor core comparing to the margin. High marginal expression of MMP-2 and MMP-9 were noted in poorly ill-defined margin on the pathological finding. High marginal expression of CD44 and MMP-2 were demonstrated in the midline cross group on the radiological review, and that of RHAMM and MMP-2 were showed in the aggressive recurrence group. High expression of MMP-2 seems to be involved in the various invasion-related phenomenons. CONCLUSION: Up-regulation of MMP-2, MMP-9, CD44 and RHAMM was noted in invasive edge of gliomas according to the various clinical situations.

20.
Int J Pharm ; 387(1-2): 209-14, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19945519

RESUMO

Although matrix metalloproteinases (MMPs) play a crucial role in the invasion and growth of malignant gliomas, their increased activity in tumor environment can be used as a specific target for chemotherapy. We investigated whether polymer-drug conjugates formed via MMP-cleavable peptide linkages could provide MMP-responsive tumor targeting and cytotoxicity for malignant glioma cells. One end of an MMP-cleavable peptide was attached to the end of methoxy polyethylene glycol (MPEG) while the other end was attached to adriamycin (ADR). The release of drugs in the presence of conditioned media of U87MG cells was investigated. The cytotoxicities of the MMP-cleavable MPEG-peptide-ADR (PPA) conjugates and non-cleavable MPEG-ADR (PA) conjugates were investigated using U87MG cells. The (1)H nuclear magnetic resonance (NMR) spectra confirmed the conjugation of the two ends of the peptide to the ends of MPEG and ADR, respectively. Gelatin zymography showed that MMP-2 was strongly expressed in the media of U87MG cells. The PA conjugate did not release ADR either in the phosphate buffered saline (PBS) or conditioned media of U87MG cells. The PPA conjugate released ADR in the presence of the conditioned media of U87MG cells, but not in PBS only. In the cytotoxicity test using U87MG cells, ADR and PPA conjugate showed similar anti-proliferative activities, while the cytotoxicity of PA conjugate was lower than that of ADR. Considering that the cytotoxicity of the PPA conjugate was similar to that of ADR, MMP-cleavable polymer-drug conjugates can be used as targeting carriers for the purpose of inhibiting the proliferation of malignant glioma cells.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Glioma/tratamento farmacológico , Metaloproteinases da Matriz/metabolismo , Antibióticos Antineoplásicos/química , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Doxorrubicina/química , Doxorrubicina/farmacologia , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Glioma/patologia , Humanos , Espectroscopia de Ressonância Magnética , Polietilenoglicóis/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA