Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 458
Filtrar
1.
Sci Rep ; 11(1): 16790, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34408195

RESUMO

With diffuse infiltrative glioma being increasingly recognized as a systemic brain disorder, the macroscopically apparent tumor lesion is suggested to impact on cerebral functional and structural integrity beyond the apparent lesion site. We investigated resting-state functional connectivity (FC) and diffusion-MRI-based structural connectivity (SC) (comprising edge-weight (EW) and fractional anisotropy (FA)) in isodehydrogenase mutated (IDHmut) and wildtype (IDHwt) patients and healthy controls. SC and FC were determined for whole-brain and the Default-Mode Network (DMN), mean intra- and interhemispheric SC and FC were compared across groups, and partial correlations were analyzed intra- and intermodally. With interhemispheric EW being reduced in both patient groups, IDHwt patients showed FA decreases in the ipsi- and contralesional hemisphere, whereas IDHmut patients revealed FA increases in the contralesional hemisphere. Healthy controls showed strong intramodal connectivity, each within the structural and functional connectome. Patients however showed a loss in structural and reductions in functional connectomic coherence, which appeared to be more pronounced in IDHwt glioma patients. Findings suggest a relative dissociation of structural and functional connectomic coherence in glioma patients at the time of diagnosis, with more structural connectomic aberrations being encountered in IDHwt glioma patients. Connectomic profiling may aid in phenotyping and monitoring prognostically differing tumor types.


Assuntos
Encéfalo/diagnóstico por imagem , Conectoma , Glioma/diagnóstico por imagem , Encéfalo/patologia , Encéfalo/ultraestrutura , Imagem de Difusão por Ressonância Magnética , Imagem de Tensor de Difusão , Feminino , Glioma/patologia , Glioma/ultraestrutura , Giro do Cíngulo/diagnóstico por imagem , Giro do Cíngulo/patologia , Giro do Cíngulo/ultraestrutura , Humanos , Masculino , Pessoa de Meia-Idade , Rede Nervosa/diagnóstico por imagem , Rede Nervosa/patologia , Rede Nervosa/ultraestrutura
2.
Cell Mol Neurobiol ; 41(3): 563-587, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32430779

RESUMO

Cisplatin (CDDP) is one of the most effective chemotherapeutic agents, used for the treatment of diverse tumors, including neuroblastoma and glioblastoma. CDDP induces cell death through different apoptotic pathways. Despite its clinical benefits, CDDP causes several side effects and drug resistance.[Pt(O,O'-acac)(γ-acac)(DMS)], namely PtAcacDMS, a new platinum(II) complex containing two acetylacetonate (acac) and a dimethylsulphide (DMS) in the coordination sphere of metal, has been recently synthesized and showed 100 times higher cytotoxicity than CDDP. Additionally, PtAcacDMS was associated to a decreased neurotoxicity in developing rat central nervous system, also displaying great antitumor and antiangiogenic activity both in vivo and in vitro. Thus, based on the knowledge that several chemotherapeutics induce cancer cell death through an aberrant increase in [Ca2+]i, in the present in vitro study we compared CDDP and PtAcacDMS effects on apoptosis and intracellular Ca2+ dynamics in human glioblastoma T98G cells, applying a battery of complementary techniques, i.e., flow cytometry, immunocytochemistry, electron microscopy, Western blotting, qRT-PCR, and epifluorescent Ca2+ imaging. The results confirmed that (i) platinum compounds may induce cell death through an aberrant increase in [Ca2+]i and (ii) PtAcacDMS exerted stronger cytotoxic effect than CDDP, associated to a larger increase in resting [Ca2+]i. These findings corroborate the use of PtAcacDMS as a promising approach to improve Pt-based chemotherapy against gliomas, either by inducing a chemosensitization or reducing chemoresistance in cell lineages resilient to CDDP treatment.


Assuntos
Neoplasias Encefálicas/patologia , Cisplatino/efeitos adversos , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Glioma/patologia , Compostos Organoplatínicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/ultraestrutura , Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/genética , Glioma/ultraestrutura , Homeostase/efeitos dos fármacos , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
3.
Acta Histochem ; 122(7): 151625, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33066845

RESUMO

The aim of this investigation was to determine the effect of doxorubicin on F-actin rearrangement and ß-catenin and cofilin-1 in a rat glioma C6 cell line in combination with changes in their morphology and ultrastructure. The experimental material constituted rat glioma C6 cell line. The cells were incubated with sublethal doses of doxorubicin in the concentration of 50, 100 and 200 nM. The blue trypan dye method was used to determine the number of dead cells. Morphological and ultrastructural changes in the cells were evaluated using light and transmission electron microscope, respectively. In order to determine the rearrangements and level of expression of F-actin, ß-catenin and cofilin-1 they were analyzed using a fluorecence microscope. In turn, cell death and cell cycle were evaluated by Guava 6HT-2 L Cytometer. The performed experiments showed a dose-dependent decrease in the survival of C6 cells after treatment with doxorubicin. The analysis of cell death showed a dose-dependent increase in the population of apoptotic and necrotic cells. These results were confirmed by microscopy observation. The changes in morphology, ultrastructure, and rearrangements of F-actin, ß-catenin and cofilin-1 were also observed. The results obtained in the study showed that sublethal concentrations of doxorubicin influenced the structure of F-actin and other proteins involved in cell-cell interactions. Moreover, mitotic catastrophe may preceding apoptosis, what suggest the cytotoxic effect of low dose of doxorubicin. Furthermore, our results confirmed the multi-dimensional mechanism of DOX action in tumor cells.


Assuntos
Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Glioma/tratamento farmacológico , Actinas/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Glioma/ultraestrutura
4.
Theranostics ; 10(4): 1873-1883, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32042342

RESUMO

Rationale: Glioblastoma is the most frequent, primary brain tumor that is characterized by a highly immunosuppressive tumor microenvironment (TME). The TME plays a key role for tumor biology and the effectiveness of immunotherapies. Composition of the TME correlates with overall survival and governs therapy response. Non invasive assessment of the TME has been notoriously difficult. Methods: We have designed an in vivo imaging approach to non invasively visualize innate immune cell dynamics in the TME in a mouse glioma model by correlated MRI and multiphoton microscopy (MR-MPM) using a bimodal, fluorescently labeled iron oxide nanoparticle (NP). The introduction of Teflon cranial windows instead of conventional Titanium rings dramatically reduced susceptibility artifacts on MRI and allowed longitudinal MR-MPM imaging for innate immune cell tracking in the same animal. Results: We visualized tumor associated macrophage and microglia (TAM) dynamics in the TME and dissect the single steps of NP uptake by blood-born monocytes that give rise to tumor-associated macrophages. Next to peripheral NP-loading, we identified a second route of direct nanoparticle uptake via the disrupted blood-brain barrier to directly label tissue resident TAMs. Conclusion: Our approach allows innate immune cell tracking by MRI and multiphoton microscopy in the same animal to longitudinally investigate innate immune cell dynamics in the TME.


Assuntos
Glioma/diagnóstico por imagem , Imunidade Inata/imunologia , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Microambiente Tumoral/imunologia , Animais , Barreira Hematoencefálica/diagnóstico por imagem , Barreira Hematoencefálica/fisiopatologia , Neoplasias Encefálicas/patologia , Rastreamento de Células/instrumentação , Glioma/patologia , Glioma/ultraestrutura , Imunidade Inata/fisiologia , Imunidade Inata/efeitos da radiação , Imunoterapia/métodos , Macrófagos/imunologia , Nanopartículas Magnéticas de Óxido de Ferro/química , Camundongos , Camundongos Endogâmicos C57BL , Microglia/patologia , Nanopartículas/administração & dosagem , Nanopartículas/química
5.
Nature ; 573(7775): 532-538, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31534219

RESUMO

A network of communicating tumour cells that is connected by tumour microtubes mediates the progression of incurable gliomas. Moreover, neuronal activity can foster malignant behaviour of glioma cells by non-synaptic paracrine and autocrine mechanisms. Here we report a direct communication channel between neurons and glioma cells in different disease models and human tumours: functional bona fide chemical synapses between presynaptic neurons and postsynaptic glioma cells. These neurogliomal synapses show a typical synaptic ultrastructure, are located on tumour microtubes, and produce postsynaptic currents that are mediated by glutamate receptors of the AMPA subtype. Neuronal activity including epileptic conditions generates synchronised calcium transients in tumour-microtube-connected glioma networks. Glioma-cell-specific genetic perturbation of AMPA receptors reduces calcium-related invasiveness of tumour-microtube-positive tumour cells and glioma growth. Invasion and growth are also reduced by anaesthesia and the AMPA receptor antagonist perampanel, respectively. These findings reveal a biologically relevant direct synaptic communication between neurons and glioma cells with potential clinical implications.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Progressão da Doença , Glioma/fisiopatologia , Sinapses/patologia , Animais , Neoplasias Encefálicas/ultraestrutura , Modelos Animais de Doenças , Glioma/ultraestrutura , Humanos , Camundongos , Microscopia Eletrônica de Transmissão , Neurônios/fisiologia , Receptores de AMPA/genética , Receptores de AMPA/metabolismo
6.
Theranostics ; 9(18): 5347-5358, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31410219

RESUMO

Rationale: Glioma is the most common malignant primary brain tumor in the central nervous system (CNS). The lack of reliable noninvasive diagnostic and prognostic methods is one of the main reasons for the high mortality of glioma. Serum has become a useful biomarker for the diagnosis and prognosis prediction of glioma because extracellular vesicles (EVs) carry molecular components from their parental cells. Methods: To detect EVs and perform molecular analysis of serum EVs, we established and optimized a microbead-assisted method based on flow cytometry and estimated the efficacy of EGFR protein expression and NLGN3 and PTTG1 mRNA in serum EVs from glioma patients (n=23) and healthy individuals (n=12). We evaluated the ability of EGFR+ EVs to differentiate high-grade and low-grade glioma patients and checked the correlation between EGFR in EVs and the ki-67 labeling index (LI) in the tumor tissue. Results: We demonstrated that EGFR+ EVs are effective diagnostic and prognostic markers of glioma. The expression of EGFR in serum EVs can accurately differentiate high-grade and low-grade glioma patients, and EGFR in EVs positively correlates with ki-67 LI in the tumor tissue. We also showed the potential of NLGN3 and PTTG1 mRNA in EVs for detecting glioma patients. Conclusions: We demonstrate that the protein expression of EGFR in serum EVs is an effective diagnostic marker of glioma. EGFR in EVs highly correlates with the malignancy of glioma. We also show the potential of NLGN3 and PTTG1 in EVs for detecting glioma. The optimized flow cytometry with the aid of microbead-based EV enrichment show its potential as a noninvasive method for the detection of glioma and will be beneficial to the management of glioma.


Assuntos
Biomarcadores Tumorais/sangue , Neoplasias Encefálicas/sangue , Vesículas Extracelulares/metabolismo , Glioma/sangue , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/ultraestrutura , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Linhagem Celular Tumoral , Receptores ErbB/sangue , Vesículas Extracelulares/ultraestrutura , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/ultraestrutura , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Securina/genética , Securina/metabolismo
7.
Z Naturforsch C J Biosci ; 74(11-12): 295-302, 2019 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-31421049

RESUMO

This research investigated the antiproliferative effects of 1-500 µM fisetin in T98G and BEAS-2B cells by MTT assay. The IC50 of fisetin in T98G cells for 24 and 48 h were 93 and 75 µM, respectively. Apoptotic alterations of fisetin-treated T98G cells were observed by transmission electron microscopy. BEAS-2B was then used in comparison to T98G cells to determine the cytotoxic effects of fisetin. The IC50 of fisetin for 24 and 48 h were recorded as 270 and 90 µM in BEAS-2B cells, respectively. Different concentrations of fisetin were selected to determine the apoptotic and necrotic effects. Consequently, fisetin was determined to have more apoptotic effects in T98G than BEAS-2B cells, dose- and time-dependently. Moreover, fisetin was found to have cytotoxicity at lower doses in T98G cells compared to carmustine, as positive control. CASPASE 3, CASPASE 9, CASPASE 8, and BAX expressions were increased by the selected fisetin doses of 25 and 50 µM, while that of BCL-2 and survivin was reduced in T98G cells. These results will serve as an essential basis of future in vitro and in vivo studies, in the continuous search for alternative treatment agents for gliomas.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Flavonoides/farmacologia , Glioma/patologia , Apoptose/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/ultraestrutura , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Flavonóis , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/genética , Glioma/ultraestrutura , Humanos , Necrose , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
8.
Elife ; 82019 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-31169500

RESUMO

GPIHBP1, a GPI-anchored protein of capillary endothelial cells, binds lipoprotein lipase (LPL) within the subendothelial spaces and shuttles it to the capillary lumen. GPIHBP1-bound LPL is essential for the margination of triglyceride-rich lipoproteins (TRLs) along capillaries, allowing the lipolytic processing of TRLs to proceed. In peripheral tissues, the intravascular processing of TRLs by the GPIHBP1-LPL complex is crucial for the generation of lipid nutrients for adjacent parenchymal cells. GPIHBP1 is absent from the capillaries of the brain, which uses glucose for fuel; however, GPIHBP1 is expressed in the capillaries of mouse and human gliomas. Importantly, the GPIHBP1 in glioma capillaries captures locally produced LPL. We use NanoSIMS imaging to show that TRLs marginate along glioma capillaries and that there is uptake of TRL-derived lipid nutrients by surrounding glioma cells. Thus, GPIHBP1 expression in gliomas facilitates TRL processing and provides a source of lipid nutrients for glioma cells.


Assuntos
Glioma/metabolismo , Lipoproteínas/metabolismo , Receptores de Lipoproteínas/metabolismo , Animais , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Capilares/metabolismo , Isótopos de Carbono/metabolismo , Células Endoteliais/metabolismo , Ácidos Graxos/metabolismo , Glioma/irrigação sanguínea , Glioma/patologia , Glioma/ultraestrutura , Glucose/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Humanos , Lipase Lipoproteica/metabolismo , Camundongos Endogâmicos C57BL , Triglicerídeos/metabolismo
9.
Acta Neuropathol Commun ; 6(1): 79, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30107856

RESUMO

Alpha-synuclein (aSyn) plays a crucial role in Parkinson's disease (PD) and other synucleinopathies, since it misfolds and accumulates in typical proteinaceous inclusions. While the function of aSyn is thought to be related to vesicle binding and trafficking, the precise molecular mechanisms linking aSyn with synucleinopathies are still obscure. aSyn can spread in a prion-like manner between interconnected neurons, contributing to the propagation of the pathology and to the progressive nature of synucleinopathies. Here, we investigated the interaction of aSyn with membranes and trafficking machinery pathways using cellular models of PD that are amenable to detailed molecular analyses. We found that different species of aSyn can enter cells and form high molecular weight species, and that membrane binding properties are important for the internalization of aSyn. Once internalized, aSyn accumulates in intracellular inclusions. Interestingly, we found that internalization is blocked in the presence of dynamin inhibitors (blocked membrane scission), suggesting the involvement of the endocytic pathway in the internalization of aSyn. By screening a pool of small Rab-GTPase proteins (Rabs) which regulate membrane trafficking, we found that internalized aSyn partially colocalized with Rab5A and Rab7. Initially, aSyn accumulated in Rab4A-labelled vesicles and, at later stages, it reached the autophagy-lysosomal pathway (ALP) where it gets degraded. In total, our study emphasizes the importance of membrane binding, not only as part of the normal function but also as an important step in the internalization and subsequent accumulation of aSyn. Importantly, we identified a fundamental role for Rab proteins in the modulation of aSyn processing, clearance and spreading, suggesting that targeting Rab proteins may hold important therapeutic value in PD and other synucleinopathies.


Assuntos
Membrana Celular/metabolismo , Endocitose/fisiologia , Transporte Proteico/fisiologia , alfa-Sinucleína/metabolismo , Biotinilação , Fracionamento Celular , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Dinaminas/farmacologia , Endocitose/efeitos dos fármacos , Glioma/patologia , Glioma/ultraestrutura , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lisossomos/fisiologia , Imagem Molecular , Mutação/genética , Transporte Proteico/efeitos dos fármacos , Transfecção , alfa-Sinucleína/genética , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
10.
Autophagy ; 14(10): 1693-1709, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29938581

RESUMO

In most cases, macroautophagy/autophagy serves to alleviate cellular stress and acts in a pro-survival manner. However, the effects of autophagy are highly contextual, and autophagic cell death (ACD) is emerging as an alternative paradigm of (stress- and drug-induced) cell demise. AT 101 ([-]-gossypol), a natural compound from cotton seeds, induces ACD in glioma cells as confirmed here by CRISPR/Cas9 knockout of ATG5 that partially, but significantly rescued cell survival following AT 101 treatment. Global proteomic analysis of AT 101-treated U87MG and U343 glioma cells revealed a robust decrease in mitochondrial protein clusters, whereas HMOX1 (heme oxygenase 1) was strongly upregulated. AT 101 rapidly triggered mitochondrial membrane depolarization, engulfment of mitochondria within autophagosomes and a significant reduction of mitochondrial mass and proteins that did not depend on the presence of BAX and BAK1. Conversely, AT 101-induced reduction of mitochondrial mass could be reversed by inhibiting autophagy with wortmannin, bafilomycin A1 and chloroquine. Silencing of HMOX1 and the mitophagy receptors BNIP3 (BCL2 interacting protein 3) and BNIP3L (BCL2 interacting protein 3 like) significantly attenuated AT 101-dependent mitophagy and cell death. Collectively, these data suggest that early mitochondrial dysfunction and HMOX1 overactivation synergize to trigger lethal mitophagy, which contributes to the cell killing effects of AT 101 in glioma cells. ABBREVIATIONS: ACD, autophagic cell death; ACN, acetonitrile; AT 101, (-)-gossypol; BAF, bafilomycin A1; BAK1, BCL2-antagonist/killer 1; BAX, BCL2-associated X protein; BH3, BCL2 homology region 3; BNIP3, BCL2 interacting protein 3; BNIP3L, BCL2 interacting protein 3 like; BP, Biological Process; CCCP, carbonyl cyanide m-chlorophenyl hydrazone; CC, Cellular Component; Con, control; CQ, chloroquine; CRISPR, clustered regularly interspaced short palindromic repeats; DMEM, Dulbecco's Modified Eagle Medium; DTT, 1,4-dithiothreitol; EM, electron microscopy; ER, endoplasmatic reticulum; FACS, fluorescence-activated cell sorting; FBS, fetal bovine serum; FCCP, carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone; GO, Gene Ontology; HAcO, acetic acid; HMOX1, heme oxygenase 1; DKO, double knockout; LC-MS/MS, liquid chromatography coupled to tandem mass spectrometry; LPL, lipoprotein lipase, MEFs, mouse embryonic fibroblasts; mPTP, mitochondrial permeability transition pore; MTG, MitoTracker Green FM; mt-mKeima, mito-mKeima; MT-ND1, mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 1; PBS, phosphate-buffered saline; PE, phosphatidylethanolamine; PI, propidium iodide; PRKN, parkin RBR E3 ubiquitin protein ligase; SDS, sodium dodecyl sulfate; SQSTM1/p62, sequestome 1; STS, staurosporine; sgRNA, single guide RNA; SILAC, stable isotope labeling with amino acids in cell culture; TFA, trifluoroacetic acid, TMRM, tetramethylrhodamine methyl ester perchlorate; WM, wortmannin; WT, wild-type.


Assuntos
Glioma/patologia , Gossipol/análogos & derivados , Heme Oxigenase-1/metabolismo , Mitocôndrias/metabolismo , Mitofagia/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Proteína 5 Relacionada à Autofagia/metabolismo , Linhagem Celular Tumoral , Glioma/enzimologia , Glioma/ultraestrutura , Gossipol/farmacologia , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Proteínas de Neoplasias/metabolismo , Proteômica
11.
Biomed Pharmacother ; 102: 699-710, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29604589

RESUMO

Gliomas, the most common primary malignant brain tumor, exhibit high metabolic activity. The targeting of metabolism alterations, particularly in mitochondria, is emerging as an efficient approach for curing cancers. Here, we showed that berberine, a natural compound that is used as an antibacterial agent, could reduce cellular viability and induce oncosis-like death, characterized by cell swelling, cytoplasmic vacuoles and plasma membrane blebbing, in gliomas, and that these effects were correlated with intracellular adenosine triphosphate (ATP) depletion. We also found that berberine induced autophagy as a protective effect and decreased the oxygen consumption rate (OCR), which could inhibit mitochondrial aerobic respiration by repressing phosphorylated extracellular regulated protein kinases (p-ERK1/2). Furthermore, the down-regulation of mitochondrial p-ERK1/2 by berberine inhibited aerobic respiration and led to glycolysis, an inefficient energy production pathway. In addition, berberine reduced tumor growth and inhibited Ki-67 and p-ERK1/2 expression in vivo. The results demonstrate that berberine, which represses aerobic oxidation in mitochondria and decreases their energy production efficiency, decreases metabolic activity by reducing ERK1/2 activity.


Assuntos
Apoptose , Berberina/farmacologia , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/patologia , Glioma/enzimologia , Glioma/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Mitocôndrias/metabolismo , Trifosfato de Adenosina/biossíntese , Aerobiose/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Berberina/química , Neoplasias Encefálicas/ultraestrutura , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Respiração Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Glioma/ultraestrutura , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/ultraestrutura , Ratos Wistar , Vacúolos/efeitos dos fármacos , Vacúolos/metabolismo , Vacúolos/ultraestrutura , Ensaios Antitumorais Modelo de Xenoenxerto
12.
J Neurooncol ; 138(1): 17-27, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29357089

RESUMO

Binding of epsin ubiquitin-interacting motif (UIM) with ubiquitylated VEGFR2 is a critical mechanism for epsin-dependent VEGFR2 endocytosis and physiological angiogenesis. Deletion of epsins in vessel endothelium produces uncontrolled tumor angiogenesis and retards tumor growth in animal models. The aim of this study is to test the therapeutic efficacy and targeting specificity of a chemically-synthesized peptide, UPI, which compete for epsin binding sites in VEGFR2 and potentially inhibits Epsin-VEGFR2 interaction in vivo, in an attempt to reproduce an epsin-deficient phenotype in tumor angiogenesis. Our data show that UPI treatment significantly inhibits and shrinks tumor growth in GL261 glioma tumor model. UPI peptide specifically targets VEGFR2 signaling pathway revealed by genetic and biochemical approaches. Furthermore, we demonstrated that UPI peptide treatment caused serious thrombosis in tumor vessels and damages tumor cells after a long-term UPI peptide administration. Besides, we revealed that UPI peptides were unexpectedly targeted cancer cells and induced apoptosis. We conclude that UPI peptide is a potent inhibitor to glioma tumor growth through specific targeting of VEGFR2 signaling in the tumor vasculature and cancer cells, which may offer a potentially novel treatment for cancer patients who are resistant to current anti-VEGF therapies.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/química , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/ultraestrutura , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/ultraestrutura , Glioma/diagnóstico por imagem , Glioma/genética , Glioma/ultraestrutura , Marcação In Situ das Extremidades Cortadas , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Trombose/tratamento farmacológico , Trombose/etiologia , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
13.
J Cell Physiol ; 233(3): 2434-2443, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28776671

RESUMO

Glioma is the most common type of primary brain tumor and has an undesirable prognosis. Autophagy plays an important role in cancer therapy, but it is effect is still not definite. P53 is an important tumor suppressor gene and protein that is closely to autophagy. Our aim was to study the effect of ß-asarone on inhibiting cell proliferation in human glioma U251 cells and to detect the effect of the inhibition on autophagy through the P53 signal pathway. For cell growth, the cells were divided into four groups: the model, ß-asarone, temozolomide (TMZ), and co-administration groups. For cell autoghapy and the P53 pathway, the cells were divided into six groups: the model, ß-asarone, 3MA, Rapa, Pifithrin-µ, and NSC groups. The counting Kit-8 assay and flow cytometry (FCM) were then used to measure the cell proliferation and cycle. Electron microscopy was used to observe autophagosome formation. Cell immunohistochemistry/-immunofluorescence, FCM and Western blot (WB) were used to examine the expression of Beclin-1 and P53. The levels of P53 and GAPDH mRNA were detected by RT-PCR. Using WB, we determined autophagy-related proteins Beclin-1, LC3-II/I, and P62 and those of the P53 pathway-related proteins P53, Bcl-2, mTOR, P-mTOR, AMPK, P-AMPK, and GAPDH. We got the results that ß-asarone changed the cellular morphology, inhibited cell proliferation, and enhanced the expression of P53, LC3-II/I, Beclin-1, AMPK, and pAMPK while inhibiting the expression of P62, Bcl-2, mTOR, and pmTOR. All the data suggested that ß-asarone could reduce the cell proliferation and promote autophagy possible via the P53 pathway in U251 cells.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Anisóis/farmacologia , Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Proteína Beclina-1/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Glioma/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Derivados de Alilbenzenos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagossomos/ultraestrutura , Proteína Beclina-1/genética , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/ultraestrutura , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Glioma/enzimologia , Glioma/genética , Glioma/ultraestrutura , Humanos , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Temozolomida , Proteína Supressora de Tumor p53/genética
14.
Cell Stem Cell ; 21(5): 591-603.e4, 2017 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-29100012

RESUMO

The blood-tumor barrier (BTB) is a major obstacle for drug delivery to malignant brain tumors such as glioblastoma (GBM). Disrupting the BTB is therefore highly desirable but complicated by the need to maintain the normal blood-brain barrier (BBB). Here we show that targeting glioma stem cell (GSC)-derived pericytes specifically disrupts the BTB and enhances drug effusion into brain tumors. We found that pericyte coverage of tumor vasculature is inversely correlated with GBM patient survival after chemotherapy. Eliminating GSC-derived pericytes in xenograft models disrupted BTB tight junctions and increased vascular permeability. We identified BMX as an essential factor for maintaining GSC-derived pericytes. Inhibiting BMX with ibrutinib selectively targeted neoplastic pericytes and disrupted the BTB, but not the BBB, thereby increasing drug effusion into established tumors and enhancing the chemotherapeutic efficacy of drugs with poor BTB penetration. These findings highlight the clinical potential of targeting neoplastic pericytes to significantly improve treatment of brain tumors.


Assuntos
Barreira Hematoencefálica/patologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Glioma/tratamento farmacológico , Glioma/patologia , Células-Tronco Neoplásicas/patologia , Pericitos/patologia , Adenina/análogos & derivados , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/ultraestrutura , Permeabilidade Capilar/efeitos dos fármacos , Glioma/ultraestrutura , Humanos , Camundongos , Células-Tronco Neoplásicas/metabolismo , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Piperidinas , Prognóstico , Proteínas Tirosina Quinases/metabolismo , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Análise de Sobrevida , Junções Íntimas/metabolismo , Resultado do Tratamento
15.
Cancer Lett ; 410: 12-19, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28939029

RESUMO

Poor tumor perfusion and unfavorable vessel permeability compromise nanomedicine drug delivery to tumors. Captopril dilates blood vessels, reducing blood pressure clinically and bradykinin, as the downstream signaling moiety of captopril, is capable of dilating blood vessels and effectively increasing vessel permeability. The hypothesis behind this study was that captopril can dilate tumor blood vessels, improving tumor perfusion and simultaneously enlarge the endothelial gaps of tumor vessels, therefore enhancing nanomedicine drug delivery for tumor therapy. Using the U87 tumor xenograft with abundant blood vessels as the tumor model, tumor perfusion experiments were carried out using laser Doppler imaging and lectin-labeling experiments. A single treatment of captopril at a dose of 100 mg/kg significantly increased the percentage of functional vessels in tumor tissues and improved tumor blood perfusion. Scanning electron microscopy of tumor vessels also indicated that the endothelial gaps of tumor vessels were enlarged after captopril treatment. Immunofluorescence-staining of tumor slices demonstrated that captopril significantly increased bradykinin expression, possibly explaining tumor perfusion improvements and endothelial gap enlargement. Additionally, imaging in vivo, imaging ex vivo and nanoparticle distribution in tumor slices indicated that after a single treatment with captopril, the accumulation of 115-nm nanoparticles in tumors had increased 2.81-fold with a more homogeneous distribution pattern in comparison to non-captopril treated controls. Finally, pharmacodynamics experiments demonstrated that captopril combined with paclitaxel-loaded nanoparticles resulted in the greatest tumor shrinkage and the most extensive necrosis in tumor tissues among all treatment groups. Taken together, the data from the present study suggest a novel strategy for improving tumor perfusion and enlarging blood vessel permeability simultaneously in order to improve nanomedicine delivery for tumor therapy. As captopril has already been extensively used clinically, such a strategy has great therapeutic potential.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Permeabilidade Capilar/efeitos dos fármacos , Captopril/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Endotélio Vascular/efeitos dos fármacos , Glioma/tratamento farmacológico , Nanomedicina/métodos , Paclitaxel/farmacologia , Vasodilatadores/farmacologia , Animais , Antineoplásicos/administração & dosagem , Bradicinina/metabolismo , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/ultraestrutura , Linhagem Celular Tumoral , Endotélio Vascular/metabolismo , Endotélio Vascular/ultraestrutura , Glioma/irrigação sanguínea , Glioma/metabolismo , Glioma/ultraestrutura , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas , Paclitaxel/administração & dosagem , Fluxo Sanguíneo Regional , Fatores de Tempo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Biomed Pharmacother ; 95: 363-374, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28858735

RESUMO

Glioma is the most common form of malignant brain cancer with high mortality rate in human. Therefore, finding effective therapeutic strategy and revealing the underlying molecular mechanism is necessary. Plant-extracted flavonoid glycosides have been suggested to be bioactive compounds with pleiotropic functions, such as anti-cancer, anti-inflammatory, antioxidant and effects. Our study was attempted to explore the anti-cancer role of linarin (acacetin-7-O-ß-d-rutinoside) in glioma in vitro and in vivo. Nuclear factor kappa-B (NF-κB) activity is a common phenomenon in various cancers, resulting in abnormal cell proliferation, malignant transformation, or resistance to cell death. P53, an essential tumor suppressor, plays an important role in preventing tumor progression. Our data indicated that linarin suppressed glioma cell proliferation and migration by inducing apoptosis, which was through reducing cell cycle-related signals, including Survivin, p-Rb, and Cyclin D1, while promoting p21, Bax, Caspase-3 and poly (ADP-ribose) polymerase (PARP) activation. Also, we found that linarin-reduced cellular proliferation of glioma was dependent on p53 up-regulation and Nuclear factor kappa-B (NF-κB)/p65-down-regulation, thereby inhibiting glioma cell growth. We further conformed the inhibitory effect of linarin in vivo using xenograft tumor model. Linarin significantly triggered apoptosis as well as the tumor growth in animals, accompanied with p53 increase and p65 decrease. Our data illustrated that linarin could be used as a promising candidate against glioma progression.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Glioma/tratamento farmacológico , Glioma/metabolismo , Glicosídeos/uso terapêutico , Fator de Transcrição RelA/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/ultraestrutura , Caspase 3/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Glioma/patologia , Glioma/ultraestrutura , Glicosídeos/farmacologia , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Transdução de Sinais/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco , Regulação para Cima/efeitos dos fármacos
17.
Autophagy ; 13(9): 1528-1542, 2017 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-28812437

RESUMO

Antiangiogenesis with bevacizumab, an antibody against vascular endothelial growth factor (VEGF), has been used for devascularization to limit the growth of malignant glioma. However, the benefits are transient due to elusive mechanisms underlying resistance to the antiangiogenic therapy. Glioma stem cells (GSCs) are capable of forming vasculogenic mimicry (VM), an alternative microvascular circulation independent of VEGF-driven angiogenesis. Herein, we report that the formation of VM was promoted by bevacizumab-induced macroautophagy/autophagy in GSCs, which was associated with tumor resistance to antiangiogenic therapy. We established a 3-dimensional collagen scaffold to examine the formation of VM and autophagy by GSCs, and found that rapamycin increased the number of VM and enhanced KDR/VEGFR-2 phosphorylation. Treatment with chloroquine, or knockdown of the autophagy gene ATG5, inhibited the formation of VM and KDR phosphorylation in GSCs. Notably, neutralization of GSCs-produced VEGF with bevacizumab failed to recapitulate the effect of chloroquine treatment and ATG5 knockdown, suggesting that autophagy-promoted formation of VM was independent of tumor cell-derived VEGF. ROS was elevated when autophagy was induced in GSCs and activated KDR phosphorylation through the phosphoinositide 3-kinase (PI3K)-AKT pathway. A ROS inhibitor, N-acetylcysteine, abolished KDR phosphorylation and the formation of VM by GSCs. By examination of the specimens from 95 patients with glioblastoma, we found that ATG5 and p-KDR expression was strongly associated with the density of VM in tumors and poor clinical outcome. Our results thus demonstrate a crucial role of autophagy in the formation of VM by GSCs, which may serve as a therapeutic target in drug-resistant glioma.


Assuntos
Autofagia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Glioma/metabolismo , Glioma/patologia , Células-Tronco Neoplásicas/patologia , Neovascularização Patológica/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Proteína 5 Relacionada à Autofagia/metabolismo , Bevacizumab/farmacologia , Bevacizumab/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/ultraestrutura , Linhagem Celular Tumoral , Proliferação de Células , Cloroquina/farmacologia , Feminino , Técnicas de Silenciamento de Genes , Glioma/tratamento farmacológico , Glioma/ultraestrutura , Humanos , Camundongos , Camundongos SCID , Modelos Biológicos , Neovascularização Patológica/tratamento farmacológico , Fosforilação/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Sirolimo/farmacologia , Análise de Sobrevida , Alicerces Teciduais/química , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Oncotarget ; 8(28): 45643-45655, 2017 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-28484091

RESUMO

Inhibition of Brd4 by JQ1 treatment showed potential in the treatment of glioma, however, some cases showed low sensitivity of JQ1. In addition, the pre-clinical analysis showed its limitation by demonstrating that transient treatment with JQ1 leads to aggressive tumor development. Thus, an improved understanding of the mechanisms underlying JQ1 is urgently required to design strategies to improve its efficiency, as well as overcome its limitation. HEXIM1 has been confirmed to have an important role in regulating JQ1 sensitivity. In our study, ubenimex, a classical anti-cancer drug showed potential in regulating the JQ1 sensitivity of glioma cells using the WST-1 proliferation assay. Further studies demonstrated that ubenimex inhibited autophagy and downregulated the autophagic degradation of HEXIM1. The role of HEXIM1 in regulating JQ1 sensitivity was verified by the HEXIM1 knockdown. Since ubenimex was verified as an Akt inhibitor, we further studied the role of Akt inhibition in regulating JQ1 sensitivity and migration of glioma cells. Data showed that ubenimex improved the efficiency of JQ1 treatment and suppressed migration both in the in vitro and in vivo xenografts models. The Akt agonist attenuated these effects, pointing to the role of Akt inhibition in JQ1 sensitivity and suppressed migration. Our findings suggest the potential of ubenimex adjuvant treatment to enhance JQ1 efficiency and attenuate parts of its side effect (enhancing tumor aggressive) by regulating the autophagic degradation of HEXIM1 and Akt inhibition.


Assuntos
Autofagia/efeitos dos fármacos , Glioma/metabolismo , Leucina/análogos & derivados , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagossomos/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Técnicas de Silenciamento de Genes , Glioma/tratamento farmacológico , Glioma/patologia , Glioma/ultraestrutura , Humanos , Leucina/farmacologia , Camundongos , Modelos Biológicos , Ensaios Antitumorais Modelo de Xenoenxerto
19.
World Neurosurg ; 103: 484-492, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28427981

RESUMO

OBJECTIVES: Recently, we reported a tendency toward spontaneous hemorrhage in both the preoperative and postoperative periods in patients with intracranial epidermoid cyst (EC). According to our experience, this tendency for spontaneous hemorrhage was partly caused by the pathologic blood vessels adjacent to the EC. This study was designed to testify this hypothesis. METHODS: Twenty-three removable pericystic or intracystic blood vessels from 17 patients with EC were collected during surgery and were then examined by transmission electron microscopy. The microvascular structure in gliomas was chosen as the control. RESULTS: Under electron microscopy, variant pathologic changes of vessels were found in all patients with EC. In the tunicae intima, we found vacuolization, apoptosis, necrosis, and intralumenal protrusion of endothelial cells, as well as swollen basement and highly flexed and discontinued elastic plate. In the tunicae media, vacuolization and swollen mitochondria were found in muscular cells. In the tunicae adventitia, extravascular erythrocytes, edema or apoptosis of pericytes, collagen predominance, and inflammatory cell infiltration and destruction were found. Neuron denature and necrosis were found in the peripheral brain tissue. In the microvascular structure of 5 glioma specimens, we found enlargement and hyperplasia of endothelial cells, swollen basement membrane, swollen pericytes, and astrocytic hyperplasia and neuron denature in adjacent brain tissues. CONCLUSIONS: Our findings provide strong evidence for the hypothesis that intracystic or pericystic vascular degeneration or destruction accounts for the spontaneous hemorrhage tendency before and after surgical resection of ECs.


Assuntos
Vasos Sanguíneos/ultraestrutura , Neoplasias Encefálicas/irrigação sanguínea , Células Endoteliais/ultraestrutura , Cisto Epidérmico/irrigação sanguínea , Glioma/irrigação sanguínea , Miócitos de Músculo Liso/ultraestrutura , Pericitos/ultraestrutura , Adolescente , Adulto , Túnica Adventícia/ultraestrutura , Apoptose , Astrócitos/ultraestrutura , Membrana Basal/ultraestrutura , Encefalopatias/cirurgia , Neoplasias Encefálicas/ultraestrutura , Colágeno/ultraestrutura , Endotélio Vascular/ultraestrutura , Cisto Epidérmico/cirurgia , Feminino , Glioma/ultraestrutura , Humanos , Masculino , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Músculo Liso Vascular/ultraestrutura , Necrose , Procedimentos Neurocirúrgicos , Hemorragia Pós-Operatória , Túnica Íntima/ultraestrutura , Vacúolos/ultraestrutura , Adulto Jovem
20.
Cell Death Dis ; 8(4): e2753, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28425987

RESUMO

Glioblastoma is the most common and aggressive form of intrinsic brain tumor with a very poor prognosis. Thus, novel therapeutic approaches are urgently needed. Tumor-treating fields (TTFields) may represent such a novel treatment option. The aim of this study was to investigate the effects of TTFields on glioma cells, as well as the functional characterization of the underlying mechanisms. Here, we assessed the anti-glioma activity of TTFields in several preclinical models. Applying TTFields resulted in the induction of cell death in a frequency- and intensity-dependent manner in long-term glioma cell lines, as well as glioma-initiating cells. Cell death occurred in the absence of caspase activation, but involved autophagy and necroptosis. Severe alterations in cell cycle progression and aberrant mitotic features, such as poly- and micronucleation, preceded the induction of cell death. Furthermore, exposure to TTFields led to reduced migration and invasion, which are both biological hallmarks of glioma cells. The combination of TTFields with irradiation or the alkylating agent, temozolomide (TMZ), resulted in additive or synergistic effects, and the O6-methyl-guanine DNA methyltransferase status did not influence the efficacy of TTFields. Importantly, TMZ-resistant glioma cells were responsive to TTFields application, highlighting the clinical potential of this therapeutic approach. In summary, our results indicate that TTFields induce autophagy, as well as necroptosis and hamper the migration and invasiveness of glioma cells. These findings may allow for a more detailed clinical evaluation of TTFields beyond the clinical data available so far.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/ultraestrutura , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Metilases de Modificação do DNA/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , Glioma/patologia , Glioma/ultraestrutura , Humanos , Invasividade Neoplásica , Temozolomida , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...