Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 900
Filtrar
1.
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
2.
Nature ; 573(7775): 526-531, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31534217

RESUMO

Metastasis-the disseminated growth of tumours in distant organs-underlies cancer mortality. Breast-to-brain metastasis (B2BM) is a common and disruptive form of cancer and is prevalent in the aggressive basal-like subtype, but is also found at varying frequencies in all cancer subtypes. Previous studies revealed parameters of breast cancer metastasis to the brain, but its preference for this site remains an enigma. Here we show that B2BM cells co-opt a neuronal signalling pathway that was recently implicated in invasive tumour growth, involving activation by glutamate ligands of N-methyl-D-aspartate receptors (NMDARs), which is key in model systems for metastatic colonization of the brain and is associated with poor prognosis. Whereas NMDAR activation is autocrine in some primary tumour types, human and mouse B2BM cells express receptors but secrete insufficient glutamate to induce signalling, which is instead achieved by the formation of pseudo-tripartite synapses between cancer cells and glutamatergic neurons, presenting a rationale for brain metastasis.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Neoplasias Encefálicas/secundário , Receptores de N-Metil-D-Aspartato/fisiologia , Transdução de Sinais/fisiologia , Sinapses/fisiologia , Animais , Neoplasias Encefálicas/ultraestrutura , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Metástase Neoplásica , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/ultraestrutura , Transmissão Sináptica
3.
Ultrastruct Pathol ; 48(5): 323-337, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-38916264

RESUMO

Glioblastoma tumors are the most aggressive primary brain tumors that develop resistance to temozolomide (TMZ). Eribulin (ERB) exhibits a unique mechanism of action by inhibiting microtubule dynamics during the G2/M cell cycle phase. We utilized the T98G human glioma cell line to investigate the effects of ERB and TMZ, both individually and in combination. The experimental groups were established as follows: control, E5 (5 nM ERB), T0.75 (0.75 mM TMZ), T1 (1.0 mM TMZ), and combination groups (E5+T0.75 and E5+T1). All groups showed a significant decrease in cell proliferation. Apoptotic markers revealed a time-dependent increase in annexin-V expression, across all treatment groups at the 48-hour time point. Caspase-3, exhibited an increase in the combination treatment groups at the 48-hour mark. Transmission electron microscopy (TEM) revealed normal ultrastructural features in the glioma cells of the control group. However, treatments induced ultrastructural changes within the spheroid glioblastoma model, particularly in the combination groups. These changes included a dose-dependent increase in autophagic vacuoles and apoptotic morphology of the cells. In conclusion, the similarity in the mechanism of action between ERB and TMZ suggests the potential for synergistic effects when combined. Our results highlight that this combination induced severe damage and autophagy in glioma spheroids after 48 hours.


Assuntos
Apoptose , Neoplasias Encefálicas , Proliferação de Células , Furanos , Glioblastoma , Cetonas , Temozolomida , Humanos , Temozolomida/farmacologia , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/ultraestrutura , Furanos/farmacologia , Cetonas/farmacologia , Linhagem Celular Tumoral , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/ultraestrutura , Proliferação de Células/efeitos dos fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Microscopia Eletrônica de Transmissão , Autofagia/efeitos dos fármacos , Policetídeos de Poliéter
4.
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
5.
Int J Mol Sci ; 22(4)2021 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-33669859

RESUMO

Titanium dioxide and zinc oxide are two of the most widely used nanomaterials. We assessed the effects of noncytotoxic doses of both nanomaterials on T98G human glioblastoma cells by omic approaches. Surprisingly, no effects on the transcriptome of T98G cells was detected after exposure to 5 µg/mL of zinc oxide nanoparticles during 72 h. Conversely, the transcriptome of the cells exposed to 20 µg/mL of titanium dioxide nanoparticles during 72 h revealed alterations in lots of biological processes and molecular pathways. Alterations to the transcriptome suggests that exposure to titanium dioxide nanoparticles might, potentially, compromise the integrity of the blood brain barrier integrity and cause neuroinflammation. The latter issue was further confirmed phenotypically with a proteomic analysis and by recording the release of interleukin 8. Titanium dioxide also caused autophagy, which was demonstrated through the increase in the expression of the autophagy-related 3 and microtubule associated protein 1 light chain 3 alpha genes. The proteomic analysis revealed that titanium dioxide nanoparticles might have anticancerigen properties by downregulating genes involved in the detoxication of anthracyclines. A risk assessment resulting from titanium dioxide exposure, focusing on the central nervous system as a potential target of toxicity, is necessary.


Assuntos
Neoplasias Encefálicas/genética , Glioblastoma/genética , Nanopartículas/toxicidade , Titânio/toxicidade , Transcriptoma/genética , Óxido de Zinco/toxicidade , Autofagia/efeitos dos fármacos , Autofagia/genética , Neoplasias Encefálicas/ultraestrutura , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Ontologia Genética , Glioblastoma/ultraestrutura , Humanos , Nanopartículas/química , Nanopartículas/ultraestrutura , Tamanho da Partícula , Proteômica , Transcriptoma/efeitos dos fármacos , Água/química
6.
J Cell Mol Med ; 24(6): 3724-3738, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32065471

RESUMO

In solid tumours, elevated interstitial fluid pressure (osmotic and hydrostatic pressure) is a barrier to drug delivery and correlates with poor prognosis. Glioblastoma (GBM) further experience compressive force when growing within a space limited by the skull. Caveolae are proposed to play mechanosensing roles, and caveola-forming proteins are overexpressed in GBM. We asked whether caveolae mediate the GBM response to osmotic pressure. We evaluated in vitro the influence of spontaneous or experimental down-regulation of caveola-forming proteins (caveolin-1, CAVIN1) on the proteolytic profile and invasiveness of GBM cells in response to osmotic pressure. In response to osmotic pressure, GBM cell lines expressing caveola-forming proteins up-regulated plasminogen activator (uPA) and/or matrix metalloproteinases (MMPs), some EMT markers and increased their in vitro invasion potential. Down-regulation of caveola-forming proteins impaired this response and prevented hyperosmolarity-induced mRNA expression of the water channel aquaporin 1. CRISPR ablation of caveola-forming proteins further lowered expression of matrix proteases and EMT markers in response to hydrostatic pressure, as a model of mechanical force. GBM respond to pressure by increasing matrix-degrading enzyme production, mesenchymal phenotype and invasion. Caveola-forming proteins mediate, at least in part, the pro-invasive response of GBM to pressure. This may represent a novel target in GBM treatment.


Assuntos
Neoplasias Encefálicas/metabolismo , Cavéolas/metabolismo , Caveolina 1/metabolismo , Glioblastoma/metabolismo , Pressão Hidrostática , Osmose , Aquaporina 1/genética , Aquaporina 1/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/ultraestrutura , Cavéolas/ultraestrutura , Linhagem Celular Tumoral , Matriz Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Glioblastoma/ultraestrutura , Humanos , Invasividade Neoplásica
7.
J Cell Mol Med ; 24(5): 2847-2856, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31778016

RESUMO

Proteolipid protein 2 (PLP2) is an integral ion channel membrane protein of the endoplasmic reticulum. The protein has been shown to be highly expressed in many cancer types, but its importance in glioma progression is poorly understood. Using publicly available datasets (Rembrandt, TCGA and CGGA), we found that the expression of PLP2 was significantly higher in high-grade gliomas than in low-grade gliomas. We confirmed these results at the protein level through IHC staining of high-grade (n = 56) and low-grade glioma biopsies (n = 16). Kaplan-Meier analysis demonstrated that increased PLP2 expression was associated with poorer patient survival. In functional experiments, siRNA and shRNA PLP2 knockdown induced ER stress and increased apoptosis and autophagy in U87 and U251 glioma cell lines. Inhibition of autophagy with chloroquine augmented apoptotic cell death in U87- and U251-siPLP2 cells. Finally, intracranial xenografts derived from U87- and U251-shPLP2 cells revealed that loss of PLP2 reduced glioma growth in vivo. Our results therefore indicate that increased PLP2 expression promotes GBM growth and that PLP2 represents a potential future therapeutic target.


Assuntos
Apoptose/genética , Autofagia/genética , Neoplasias Encefálicas/genética , Estresse do Retículo Endoplasmático/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/patologia , Proteínas com Domínio MARVEL/genética , Proteolipídeos/genética , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/ultraestrutura , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação para Baixo/genética , Técnicas de Silenciamento de Genes , Glioblastoma/ultraestrutura , Humanos , Proteínas com Domínio MARVEL/metabolismo , Masculino , Camundongos , Prognóstico , Proteolipídeos/metabolismo , Fator de Transcrição CHOP/metabolismo
8.
Cell Commun Signal ; 18(1): 21, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-32033611

RESUMO

BACKGROUND: Glioblastoma multiforme is an aggressive primary brain tumor that is characterized by local invasive growth and resistance to therapy. The role of the microenvironment in glioblastoma invasiveness remains unclear. While carcinomas release CD147, a protein that signals for increased matrix metalloproteinase (MMP) release by fibroblasts, glioblastoma does not have a significant fibroblast component. We hypothesized that astrocytes release MMPs in response to CD147 contained in glioblastoma-derived extracellular vesicles (EVs) and that ionizing radiation, part of the standard treatment for glioblastoma, enhances this release. METHODS: Astrocytes were incubated with EVs released by irradiated or non-irradiated human glioblastoma cells wild-type, knockdown, or knockout for CD147. Levels of CD147 in glioblastoma EVs and MMPs secreted by astrocytes were quantified. Levels of proteins in the mitogen activated protein kinase (MAPK) pathway, which can be regulated by CD147, were measured in astrocytes incubated with EVs from glioblastoma cells wild-type or knockdown for CD147. Immunofluorescence was performed on the glioblastoma cells to identify changes in CD147 localization in response to irradiation, and to confirm uptake of the EVs by astrocytes. RESULTS: Immunoblotting and mass spectrometry analyses showed that CD147 levels in EVs were transiently increased when the EVs were from glioblastoma cells that were irradiated with γ rays. Specifically, the highly-glycosylated 45 kDa form of CD147 was preferentially present in the EVs relative to the cells themselves. Immunofluorescence demonstrated that astrocytes incorporate glioblastoma EVs and subsequently increase their secretion of active MMP9. The increase was greater if the EVs were from irradiated glioblastoma cells. Testing MAPK pathway activation, which also regulates MMP expression, showed that JNK signaling, but not ERK1/2 or p38, was increased in astrocytes incubated with EVs from irradiated compared to non-irradiated glioblastoma cells. Knockout of CD147 in glioblastoma cells blocked the increased JNK signaling and the rise in secreted active MMP9 levels. CONCLUSIONS: The results support a tumor microenvironment-mediated role of CD147 in glioblastoma invasiveness, and reveal a prominent role for ionizing radiation in enhancing the effect. They provide an improved understanding of glioblastoma intercellular signaling in the context of radiotherapy, and identify pathways that can be targeted to reduce tumor invasiveness. Video abstract.


Assuntos
Astrócitos/metabolismo , Basigina/metabolismo , Neoplasias Encefálicas/metabolismo , Vesículas Extracelulares/metabolismo , Glioblastoma/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Radiação Ionizante , Astrócitos/patologia , Astrócitos/ultraestrutura , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/ultraestrutura , Linhagem Celular Tumoral , Vesículas Extracelulares/efeitos da radiação , Vesículas Extracelulares/ultraestrutura , Glioblastoma/patologia , Glioblastoma/ultraestrutura , Humanos , Invasividade Neoplásica , Proteômica , Transdução de Sinais , Regulação para Cima
9.
Neurosurg Rev ; 43(3): 903-910, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30788677

RESUMO

Telomerase activity and (human) Telomerase Reverse Transcriptase (hTERT) expression are considered hallmarks in oncogenesis of neoplasms and are upregulated by alterations of the hTERT promoter. In meningiomas, numerous studies investigated hTERT expression, telomerase activity, promoter mutations, and methylations. Moreover, reports about hTERT-targeted chemotherapy in meningiomas have recently been published. We provide a systematic review of the literature about the role of hTERT in meningiomas. TERT expression and telomerase activity is found in benign and high-grade meningiomas and increase with WHO grade. Remarkably, rates of TERT expression/telomerase activity usually exceed mutation frequency and both telomerase activity and TERT expression have also been found in hTERT promoter wildtype meningiomas, indicating further mechanisms of TERT upregulation. Although hTERT promoter methylation has been reported in the vast majority of meningiomas, correlation with TERT expression remains controversial. Rates of promoter mutations, and methylation were shown to increase with rising WHO grade. Moreover, promoter methylation and mutations strongly correlate with prognosis. Although mutations predicted malignant progression, de novo mutations in high-grade recurrences of former benign lesions were also observed. Retroviral transduction of the TERT gene enabled immortalization in several grade I-III meningioma cell lines. In vitro analyses revealed significant effects on viability in hTERT-mutated meningioma cells after targeted treatment. Alternative mechanisms of telomere lengthening are usually absent in meningiomas. TERT and hTERT promoter alterations play a major role during oncogenesis of meningiomas with implications for prognosis and potentially treatment.


Assuntos
Neoplasias Encefálicas/genética , Meningioma/genética , Telomerase/genética , Telomerase/metabolismo , Homeostase do Telômero/genética , Telômero/genética , Neoplasias Encefálicas/ultraestrutura , Humanos , Meningioma/ultraestrutura , Telomerase/biossíntese , Telômero/ultraestrutura
10.
Ultrastruct Pathol ; 44(2): 227-236, 2020 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-32148147

RESUMO

The phenomenon of unstable expression of gap junction's proteins connexins remains a "visiting card" of astrocytic tumors with various degrees of malignancy. At the same time, it stays unclear what is detected by the positive expression of connexins in astrocytic tumors: gap junctions, hemi-channels, or connexin proteins in cytosol. In the present work, for the first time, we demonstrate an ultrastructural evidence of gap junctions in pleomorphic xanthoastrocytoma, a rare primary brain tumor, the intercellular characteristics of which are poorly studied and remain very discursive and controversial. The primary tumor mass was resected during craniotomy from a 57-old patient diagnosed with pleomorphic xanthoastrocytoma Grade II based on the histopathological analysis. The immunohistochemical study was conducted with primary antibodies: Neurofilament, Myelin basic protein, Glial fibrillary acidic protein, and Synaptophysin. For electron microscopic examination fragments of tumor tissue were fixed in a glutaraldehyde, postfixed in a 1% OsO4, dehydrated and embedded into resin. After the detailed clinical, histological, and immunohistochemical study we revealed some ultrastructural characteristics of the tumor, as well as the first evidence of direct intercellular connection between the tumor cells via gap junctions. Regularly arranged gap junctions connected the somas of xanthastrocytes with dark cytoplasm containing lipid drops. Besides the localization between the cell bodies, from one to several gap junctions were found between the branches of xanthoastrocytoma in tumor intercellular space in close proximity to tumor cell. Our results may indicate gap junctions as a possible structure for intercellular communication between pleomorphic xanthoastrocytoma cells.


Assuntos
Astrocitoma/ultraestrutura , Neoplasias Encefálicas/ultraestrutura , Junções Comunicantes/ultraestrutura , Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Humanos , Masculino , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade
11.
J Cell Mol Med ; 23(11): 7859-7872, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31532058

RESUMO

Glioblastoma (GBM) is the most frequent and inevitably lethal primary brain cancer in adults. It is recognized that the overexpression of the endosomal Na+ /H+ exchanger NHE9 is a potent driver of GBM progression. Patients with NHE9 overexpression have a threefold lower median survival relative to GBM patients with normal NHE9 expression, using available treatment options. New treatment strategies tailored for this GBM subset are much needed. According to the prevailing model, NHE9 overexpression leads to an increase in plasma membrane density of epidermal growth factor receptors (EGFRs) which consequently enhances GBM cell proliferation and migration. However, this increase is not specific to EGFRs. In fact, the hallmark of NHE9 overexpression is a pan-specific increase in plasma membrane receptors. Paradoxically, we report that this gain of function in NHE9 can be exploited to effectively target GBM cells for destruction. When exposed to gold nanoparticles, NHE9 overexpressing GBM cells accumulated drastically high amounts of gold via receptor-mediated endocytosis, relative to control. Irradiation of these cells with near-infrared light led to apoptotic tumour cell death. A major limitation for delivering therapeutics to GBM cells is the blood-brain barrier (BBB). Here, we demonstrate that macrophages loaded with gold nanoparticles can cross the BBB, deliver the gold nanoparticles and effect the demise of GBM cells. In combination with receptor tyrosine kinase inhibition, we show this approach holds great promise for a new GBM-targeted therapy.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Mutação com Ganho de Função/genética , Glioblastoma/tratamento farmacológico , Terapia de Alvo Molecular , Trocadores de Sódio-Hidrogênio/genética , Animais , Apoptose , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/ultraestrutura , Linhagem Celular Tumoral , Clatrina/metabolismo , Endocitose , Endossomos/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Glioblastoma/patologia , Glioblastoma/ultraestrutura , Ouro , Humanos , Concentração de Íons de Hidrogênio , Hipertermia Induzida , Macrófagos/metabolismo , Nanopartículas Metálicas/ultraestrutura , Camundongos , Fototerapia , Células RAW 264.7 , Trocadores de Sódio-Hidrogênio/metabolismo
12.
Nanotechnology ; 30(27): 275101, 2019 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-30856613

RESUMO

Despite decades of efforts, non-invasive sensitive detection of small malignant brain tumors still remains challenging. Here we report a dual-modality 124I-labeled gold nanostar (124I-GNS) probe for sensitive brain tumor imaging with positron emission tomography (PET) and subcellular tracking with two-photon photoluminescence (TPL) and electron microscopy (EM). Experiment results showed that the developed nanoprobe has potential to reach sub-millimeter intracranial brain tumor detection using PET scan, which is superior to any currently available non-invasive imaging modality. Microscopic examination using TPL and EM further confirmed that GNS nanoparticles permeated the brain tumor leaky vasculature and accumulated inside brain tumor cells following systemic administration. Selective brain tumor targeting by enhanced permeability and retention effect and ultrasensitive imaging render 124I-GNS nanoprobe promise for future brain tumor-related preclinical and translational applications.


Assuntos
Neoplasias Encefálicas/diagnóstico , Diagnóstico por Imagem , Nanopartículas Metálicas/química , Animais , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/ultraestrutura , Fluordesoxiglucose F18/química , Ouro/química , Ouro/toxicidade , Células HEK293 , Humanos , Radioisótopos do Iodo/química , Nanopartículas Metálicas/toxicidade , Nanopartículas Metálicas/ultraestrutura , Camundongos , Imagem Óptica , Especificidade de Órgãos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada
13.
Neuropathology ; 39(2): 120-126, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30588667

RESUMO

A rare case of extraventricular neurocytoma (EVN) arising from the VIIIth cranial nerve in a 34-year-old woman is reported. The patient had a 20-year history of hearing loss and facial palsy. Computed tomography showed a 3-cm enhancing lesion in the left cerebellopontine angle (CPA). At operation, the tumor was seen to originate from the cochlear and vestibular nerves. The tumor was subtotally resected. Histologically, the tumor consisted of uniform cells with oval to round nuclei and scant cytoplasm. Immunohistochemically, the tumor cells were positive for synaptophysin, but negative for glial fibrillary acid protein and S-100 protein. The Ki-67 labeling index was 0%. Twelve years after the operation, magnetic resonance imaging (MRI) showed tumor recurrence at the left CPA. The tumor was subtotally resected, and radiation therapy was given. Histologically, the tumor consisted of round cells with mild atypia and one mitosis/20 high-power fields (HPF). Immunohistochemically, tumor cells showed the same findings as the first operation sample, except for the Ki-67 labeling index (3%). Twelve years after the second operation, MRI showed a second tumor recurrence at the left CPA and surroundings of the brain stem. The tumor was subtotally resected. Histologically, the tumor consisted of anaplastic short spindle cells and five mitoses/10 HPF. The immunohistochemical findings were almost the same as the earlier operation samples. However, the Ki-67 labeling index was 20%. In addition, tumor cells from the third specimen were more strongly and more diffusely positive for GAB1 (growth factor receptor-bound protein 2-associated binding protein 1) compared to those of the earlier specimens. Electron microscopy showed the presence of numerous cell processes with a dense core and clear vesicles and microtubules. GAB1 immunostaining also indicated that malignant progression might be associated with the sonic hedgehog signaling pathways. To the best of our knowledge, this is the first report of an EVN arising from the VIIIth cranial nerve with malignant progression.


Assuntos
Neoplasias Encefálicas/patologia , Neoplasias dos Nervos Cranianos/patologia , Progressão da Doença , Neurocitoma/patologia , Nervo Vestibulococlear/patologia , Adulto , Neoplasias Encefálicas/ultraestrutura , Neoplasias dos Nervos Cranianos/ultraestrutura , Feminino , Humanos , Neurocitoma/ultraestrutura , Nervo Vestibulococlear/ultraestrutura
14.
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
15.
Small ; 14(23): e1800740, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29726109

RESUMO

Difficulty in visualizing glioma margins intraoperatively remains a major issue in the achievement of gross total tumor resection and, thus, better clinical outcome of glioblastoma (GBM) patients. Here, the potential of a new combined optical + optoacoustic imaging method for intraoperative brain tumor delineation is investigated. A strategy using a newly developed gold nanostar synthesis method, Raman reporter chemistry, and silication method to produce dual-modality contrast agents for combined surface-enhanced resonance Raman scattering (SERRS) and multispectral optoacoustic tomography (MSOT) imaging is devised. Following intravenous injection of the SERRS-MSOT-nanostars in brain tumor bearing mice, sequential MSOT imaging is performed in vivo and followed by Raman imaging. MSOT is able to accurately depict GBMs three-dimensionally with high specificity. The MSOT signal is found to correlate well with the SERRS images. Because SERRS enables uniquely sensitive high-resolution surface detection, it could represent an ideal complementary imaging modality to MSOT, which enables real-time, deep tissue imaging in 3D. This dual-modality SERRS-MSOT-nanostar contrast agent reported here is shown to enable high precision depiction of the extent of infiltrating GBMs by Raman- and MSOT imaging in a clinically relevant murine GBM model and could pave new ways for improved image-guided resection of brain tumors.


Assuntos
Neoplasias Encefálicas/diagnóstico , Nanopartículas/química , Técnicas Fotoacústicas/métodos , Análise Espectral Raman/métodos , Tomografia/métodos , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/ultraestrutura , Glioblastoma/diagnóstico , Glioblastoma/patologia , Glioblastoma/ultraestrutura , Humanos , Camundongos
16.
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
17.
Mol Cell ; 39(4): 560-9, 2010 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-20797628

RESUMO

Structural studies have provided detailed insights into different functional states of the ribosome and its interaction with factors involved in nascent peptide folding, processing, and targeting. However, how the translational machinery is organized spatially in native cellular environments is not yet well understood. Here we have mapped individual ribosomes in electron tomograms of intact human cells by template matching and determined the average structure of the ribosome in situ. Characteristic features of active ribosomes in the cellular environment were assigned to the tRNA channel, elongation factors, and additional densities near the peptide tunnel. Importantly, the relative spatial configuration of neighboring ribosomes in the cell is clearly nonrandom. The preferred configurations are specific for active polysomes and were largely abrogated in puromycin-treated control cells. The distinct neighbor orientations found in situ resemble configurations of bacterial polysomes in vitro, indicating a conserved supramolecular organization with implications for nascent polypeptide folding.


Assuntos
Neoplasias Encefálicas/ultraestrutura , Glioblastoma/ultraestrutura , Polirribossomos/ultraestrutura , Sítios de Ligação , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Tomografia com Microscopia Eletrônica , Glioblastoma/metabolismo , Humanos , Imageamento Tridimensional , Modelos Moleculares , Polirribossomos/efeitos dos fármacos , Polirribossomos/metabolismo , Biossíntese de Proteínas , Conformação Proteica , Inibidores da Síntese de Proteínas/farmacologia , Puromicina/farmacologia , Relação Estrutura-Atividade
18.
Neuropathology ; 37(2): 116-121, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27645472

RESUMO

We describe a unique case of pleomorphic xanthoastrocytoma (PXA) in a 19-year-old male presenting with the chief complaint of seizures. On radiology, the tumor was located in the temporal lobe. It was cortically based and solid cystic in nature. Light microscopy showed pleomorphic large polygonal cells with inclusions, nuclear clustering, lipidization, and foamy cytoplasm intermingled with spindle cells arranged in sweeping pattern and focally containing cytoplasmic brownish black pigment. The pigment stained black with Fontana-Masson stain and bleached with potassium permanganate. Gomori silver stain showed reticulin fibers surrounding individual tumor cells as well as groups of cells. On immunohistochemistry, tumor cells were positive for GFAP, S-100 and focally for synaptophysin and CD34 but negative for HMB-45. CD34 revealed a specific membranous pattern around individual cells as well as groups of cells along the fibers replicating a reticulin pattern. The ultrastructural examination showed supporting melanosomes, thus confirming the melanin pigment. Sequencing for BRAF V600E showed a heterozygous mutation. To our knowledge only five cases of PXA with melanin pigment have been reported and none of which described BRAF V600E mutation analysis. This case provides further insight into the origin and pathogenesis of pigmented astrocytic tumor, additionally highlighting the characteristic CD34 staining pattern.


Assuntos
Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Neuroglia/patologia , Neurônios/patologia , Adulto , Astrocitoma/genética , Astrocitoma/ultraestrutura , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/ultraestrutura , Humanos , Masculino , Neuroglia/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas B-raf/genética , Adulto Jovem
19.
Neuropathology ; 36(5): 490-495, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27121356

RESUMO

Ependymomas are gliomas that recapitulate the ependymal cells microscopically and ultrastructurally. They commonly occur along the ventricular surfaces and central canal of the brain and spinal cord. Intracranial extra-axial ependymoma (IEAE) is a rare entity and is commonly misdiagnosed clinically and radiologically as a meningioma. The histogenesis of such IEAEs is obscure. A novel recurrent oncogenic fusion involving the C11orf95 and RELA genes was recently described in supratentorial ependymomas. A 9-year-old girl presented with a dural based parafalcine mass that, in addition to exhibiting classical immunohistochemical features of an ependymoma, also demonstrated C11orf95-RELA fusion, characteristic of supratentorial ependymomas. We suggest that IEAEs share their histogenesis with their intra-axial counterparts, arising either from dural extension of subcortical, subependymal rests or directly from ectopic dural rests.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Ependimoma/genética , Ependimoma/patologia , Proteínas/genética , Fator de Transcrição RelA/genética , Encéfalo/ultraestrutura , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/ultraestrutura , Criança , Ependimoma/diagnóstico por imagem , Feminino , Fusão Gênica , Humanos
20.
Bioconjug Chem ; 26(4): 660-8, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25775241

RESUMO

A current limitation in molecular imaging is that it often requires genetic manipulation of cancer cells for noninvasive imaging. Other methods to detect tumor cells in vivo using exogenously delivered and functionally active reporters, such as ß-gal, are required. We report the development of a platform system for linking ß-gal to any number of different ligands or antibodies for in vivo targeting to tissue or cells, without the requirement for genetic engineering of the target cells prior to imaging. Our studies demonstrate significant uptake in vitro and in vivo of an EGFR-targeted ß-gal complex. We were then able to image orthotopic brain tumor accumulation and localization of the targeted enzyme when a fluorophore was added to the complex, as well as validate the internalization of the intravenously administered ß-gal reporter complex ex vivo. After fluorescence imaging localized the ß-gal complexes to the brain tumor, we topically applied a bioluminescent ß-gal substrate to serial sections of the brain to evaluate the delivery and integrity of the enzyme. Finally, robust bioluminescence of the EGFR-targeted ß-gal complex was captured within the tumor during noninvasive in vivo imaging.


Assuntos
Neoplasias Encefálicas/ultraestrutura , Receptores ErbB/genética , Imagem Molecular/métodos , Proteínas de Neoplasias/genética , Imagem Óptica/métodos , beta-Galactosidase/genética , Animais , Biotina/química , Biotina/metabolismo , Biotinilação , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Fator de Crescimento Epidérmico/química , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Feminino , Expressão Gênica , Genes Reporter , Histidina/química , Histidina/genética , Histidina/metabolismo , Humanos , Camundongos , Camundongos Nus , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Oligopeptídeos/química , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Técnicas Estereotáxicas , Estreptavidina/química , Estreptavidina/metabolismo , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA