Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
PLoS One ; 18(3): e0278793, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36893156

RESUMO

Chronic exposure to the Cyanobacteria biotoxin Beta-methylamino-L-alanine (BMAA) has been associated with development of a sporadic form of ALS called Amyotrophic Lateral Sclerosis/Parkinsonism-Dementia Complex (ALS/PDC), as observed within certain Indigenous populations of Guam and Japan. Studies in primate models and cell culture have supported the association of BMAA with ALS/PDC, yet the pathological mechanisms at play remain incompletely characterized, effectively stalling the development of rationally-designed therapeutics or application of preventative measures for this disease. In this study we demonstrate for the first time that sub-excitotoxic doses of BMAA modulate the canonical Wnt signaling pathway to drive cellular defects in human neuroblastoma cells, suggesting a potential mechanism by which BMAA may promote neurological disease. Further, we demonstrate here that the effects of BMAA can be reversed in cell culture by use of pharmacological modulators of the Wnt pathway, revealing the potential value of targeting this pathway therapeutically. Interestingly, our results suggest the existence of a distinct Wnt-independent mechanism activated by BMAA in glioblastoma cells, highlighting the likelihood that neurological disease may result from the cumulative effects of distinct cell-type specific mechanisms of BMAA toxicity.


Assuntos
Diamino Aminoácidos , Esclerose Lateral Amiotrófica , Glioblastoma , Neuroblastoma , Transtornos Parkinsonianos , Animais , Humanos , Glioblastoma/induzido quimicamente , Esclerose Lateral Amiotrófica/patologia , Toxinas de Cianobactérias , Diamino Aminoácidos/toxicidade , Diamino Aminoácidos/metabolismo , Neurotoxinas/toxicidade
2.
Toxicol Appl Pharmacol ; 459: 116356, 2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36563751

RESUMO

Although there is an association between heavy metals and glioma, the molecular mechanisms involved in glioma development remain unclear. Therefore, this study aimed to assess the molecular mechanisms implicated in glioma development induced by heavy metals and their mixtures using various methodologies and databases (CTD, Google Scholar, PubMed, ScienceDirect, SpringerLink, miRNAsong, GeneMANIA, Metascape, MIENTURNET, UALCAN). I found that heavy metals, particularly arsenic, mercury, lead, and cadmium, as well as their mixtures, have substantial influences on the etiology of gliomas. "glioblastoma signaling pathways," "integrated cancer pathway," "central carbon metabolism in cancer," "microRNAs in cancer," "p53 signaling pathway," "chemical carcinogenesis-DNA adducts," "glioma," "TP53 network," and "MAPK signaling pathway" were the predominant molecular pathways implicated in the glioma development induced by the studied heavy metals and their mixtures. Five genes (SOD1, CAT, GSTP1, PTGS2, TNF), two miRNAs (hsa-miR-26b-5p and hsa-miR-143-3p), and transcription factors (DR1 and HNF4) were identified as key components related to combined heavy metal and glioma development. Physical interactions were found to be the most common among the heavy metals and their mixtures studied (ranging from 45.2% to 77.6%). The expression level of SOD1 was significantly lower in glioblastoma multiforma samples compared to normal samples, whereas GSTP1 and TP53 expression levels were significantly higher. Brain lower and grade glioma patients who had higher levels of TP53, hsa-miR-25, hsa-miR-34, hsa-miR-222, and hsa-miR-143 had a reduced likelihood of survival. Our findings suggest that further priority should be given to investigating the impact of specific heavy metals or their mixtures on these molecular processes.


Assuntos
Glioblastoma , Glioma , Metais Pesados , MicroRNAs , Humanos , Glioblastoma/induzido quimicamente , Glioblastoma/genética , Glioblastoma/metabolismo , Prognóstico , Superóxido Dismutase-1 , MicroRNAs/genética , MicroRNAs/metabolismo , Glioma/genética , Metais Pesados/toxicidade , Biomarcadores
3.
Neuro Oncol ; 24(12): 2172-2179, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-35551405

RESUMO

BACKGROUND: Glioblastoma (GBM) is associated with a high incidence of venous thromboembolism (VTE), but there are little data to guide anticoagulation in patients with GBM, in whom the risks of VTE must be balanced against the risk of intracranial hemorrhage (ICH). METHODS: We performed a single-institution retrospective cohort study of patients with GBM diagnosed with VTE from 2014 to 2021 who were treated with low molecular weight heparin (LMWH) or a direct oral anticoagulant (DOAC). The incidence of ICH was compared between the LMWH and DOAC groups. The primary outcome was clinically relevant ICH within the first 30 days of anticoagulation, defined as any ICH that was fatal, symptomatic, required surgical intervention, and/or led to cessation of anticoagulation. Secondary outcomes included clinically relevant ICH within 6 months, fatal ICH within 30 days and 6 months, and any bleeding within 30 days and 6 months. RESULTS: One hundred twenty-one patients were identified in the cohort for 30-day outcome analyses (DOAC, n = 33; LMWH, n = 88). For 6-month outcome analyses, the cohort included only patients who were maintained on their initial anticoagulant (DOAC, n = 32; LMWH, n = 75). The incidence of clinically relevant ICH at 30 days was 0% in the DOAC group and 9% in the LMWH group (P = .11). The cumulative incidence of clinically relevant ICH at 6 months was 0% in the DOAC group and 24% in the LMWH group (P = .001), with 4 fatal ICHs in the LMWH group. CONCLUSIONS: DOACs are associated with a lower incidence of clinically relevant ICH in patients with GBM-associated VTE compared to LMWH.


Assuntos
Glioblastoma , Tromboembolia Venosa , Humanos , Heparina de Baixo Peso Molecular/efeitos adversos , Tromboembolia Venosa/tratamento farmacológico , Tromboembolia Venosa/epidemiologia , Tromboembolia Venosa/etiologia , Glioblastoma/complicações , Glioblastoma/tratamento farmacológico , Glioblastoma/induzido quimicamente , Estudos Retrospectivos , Anticoagulantes/efeitos adversos , Hemorragias Intracranianas/induzido quimicamente , Hemorragias Intracranianas/epidemiologia , Hemorragias Intracranianas/complicações , Estudos de Coortes
4.
Neurotoxicology ; 87: 219-230, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34687775

RESUMO

Galaxolide and tonalide are well-known polycyclic musks whose intensive use without limitations in numerous cleaning, hygiene, and personal care products has resulted in widespread direct human exposure via absorption, inhalation, and oral ingestion. Latest data shows that long-term, low-dose exposure to toxic chemicals can induce unpredictable harmful effects in a variety of living systems, however, interactions between synthetic musks and brain tumours remain largely unexplored. Glioblastoma (GB) accounts for nearly half of all tumours of the central nervous system and is characterized by very poor prognosis. The aims of this study were (1) to investigate the potential effect of long-term (20-generation) single and combined application of galaxolide and tonalide at sub-lethal doses (5-2.5 u M) on the angiogenesis, invasion, and migration of human U87 cells or tumour spheroids, and (2) to explore the underlying molecular mechanisms. Random amplified polymorphic DNA assays revealed significant DNA damage and increased total mutation load in galaxolide- and/or tonalide-treated U87 cells. In those same groups, we also detected remarkable tumour spheroid invasion and up-regulation of both HIF1-α/VEGF/MMP9 and IL6/JAK2/STAT3 signals, known to have important roles in hypoxia-related angiogenesis and/or proliferation. Prolonged musk treatment further altered angio-miRNA expression in a manner consistent with poor prognosis in GB. We also detected significant over-expression of the genes Slug, Snail, ZEB1, and Vimentin, which are biomarkers of epithelial to mesenchymal transition. In addition, matrigel, transwell, and wound healing assays clearly showed that long-term sub-lethal exposure to galaxolide and/or tonalide induced invasion and migration proposing a high metastatic potential. Our results suggest that assessing expression of HIF-1a, VEGF, STAT3, and the miR-17-92 cluster in biopsy samples of GB patients who have a history of possible long-term exposure to galaxolide or tonalide could be beneficial for deciding a therapy regime. Additionally, we recommend that extensively-used hygiene and cleaning materials be selected from synthetic musk-free products, especially when used in palliative care processes for GB patients.


Assuntos
Benzopiranos/toxicidade , Carcinógenos/toxicidade , Glioblastoma/induzido quimicamente , Tetra-Hidronaftalenos/toxicidade , Benzopiranos/administração & dosagem , Carcinógenos/administração & dosagem , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Glioblastoma/patologia , Humanos , Esferoides Celulares/efeitos dos fármacos , Tetra-Hidronaftalenos/administração & dosagem
5.
Chem Pharm Bull (Tokyo) ; 69(9): 832-839, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34470947

RESUMO

Thiamine (vitamin B1), which is synthesized only in bacteria, fungi and plants and which humans should take with diet, participates in basic biochemical and physiological processes in a versatile way and its deficiency is associated with neurological problems accompanied by cognitive dysfunctions. The rat glioblastoma (C6) model was used, which was exposed to a limited environment and toxicity with glutamate. The cells were stressed by exposure to glutamate in the presence and absence of thiamine. The difference in cell proliferation was evaluated in the XTT assay. Oxidative stress (OS) markers malondialdehyde (MDA), superoxide dismutase (SOD), and catalase (CAT) levels, as well as endoplasmic reticulum (ER) stress markers 78-kDa glucose-regulated protein (GRP78), activating transcription factor-4 (ATF-4), and C/EBP homologous protein (CHOP) levels, were measured with commercial kits. Apoptosis determined by flow cytometry was confirmed by 4',6-diamidino-2-phenylindole (DAPI) staining. At all concentrations, thiamine protects the cells and increased the viability against glutamate-induced toxicity. Thiamine also significantly decreased the levels of MDA, while increasing SOD and CAT levels. Moreover, thiamine reduced ER stress proteins' levels. Moreover, it lessened the apoptotic cell amount and enhanced the live-cell percentage in the flow cytometry and DAPI staining. As a result, thiamine may be beneficial nutritional support for individuals with a predisposition to neurodegenerative disorders due to its protective effect on glutamate cytotoxicity in glioblastoma cells by suppressing OS and ER stress.


Assuntos
Glioblastoma/tratamento farmacológico , Substâncias Protetoras/farmacologia , Tiamina/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Glioblastoma/induzido quimicamente , Glioblastoma/patologia , Ácido Glutâmico , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/química , Ratos , Tiamina/química , Células Tumorais Cultivadas
6.
Neurotoxicol Teratol ; 86: 106998, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34048896

RESUMO

Exposure to N-nitroso compounds (NOCs) during pregnancy has been associated with an increase in brain tumors in the progeny. This study investigated the brain tumorigenic effect of N-ethyl N-nitrosourea (ENU) after differential exposure of rats during pregnancy. Sprague Dawley rats were exposed to a single dose of ENU (80 mg/kg) in three different circumstances: 1) at first, second or third week of gestation; 2) at the 15th embryonic day (E15) in consecutive litters and 3) at E15 in three successive generations. Location and characterization of the offspring's brain tumors were performed by magnetic resonance imaging and histopathological studies. Finally, tumor incidence and latency and the animals' survival were recorded. ENU-exposure in the last two weeks of pregnancy induced intracranial tumors in over 70% of the offspring rats, these being mainly gliomas with some peripheral nerve sheath tumors (PNSTs). Tumors appeared in young adults; glioma-like small multifocal neoplasias converged on large glioblastomas in senescence and PNSTs in the sheath of the trigeminal nerve, extending to cover the brain convexity. ENU-exposure at E15 in subsequent pregnancies lead to an increase in glioma and PNST incidence. However, consecutive generational ENU-exposure (E15) decreased the animals' survival due to an early onset of both types of tumors. Moreover, PNST presented an inheritable component because progeny, which were not themselves exposed to ENU but whose progenitors were, developed PNSTs. Our results suggest that repeated exposure to ENU later in pregnancy and in successive generations favours the development of intracranial gliomas and PNSTs in the offspring.


Assuntos
Alquilantes/toxicidade , Neoplasias Encefálicas/induzido quimicamente , Etilnitrosoureia/toxicidade , Glioma/induzido quimicamente , Neoplasias de Bainha Neural/induzido quimicamente , Envelhecimento , Animais , Feminino , Idade Gestacional , Glioblastoma/induzido quimicamente , Glioblastoma/patologia , Glioma/diagnóstico por imagem , Glioma/patologia , Estimativa de Kaplan-Meier , Imageamento por Ressonância Magnética , Neoplasias de Bainha Neural/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Ratos Sprague-Dawley , Análise de Sobrevida
7.
Neuro Oncol ; 22(8): 1150-1161, 2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32296841

RESUMO

BACKGROUND: Imagining ways to prevent or treat glioblastoma (GBM) has been hindered by a lack of understanding of its pathogenesis. Although overexpression of platelet derived growth factor with two A-chains (PDGF-AA) may be an early event, critical details of the core biology of GBM are lacking. For example, existing PDGF-driven models replicate its microscopic appearance, but not its genomic architecture. Here we report a model that overcomes this barrier to authenticity. METHODS: Using a method developed to establish neural stem cell cultures, we investigated the effects of PDGF-AA on subventricular zone (SVZ) cells, one of the putative cells of origin of GBM. We microdissected SVZ tissue from p53-null and wild-type adult mice, cultured cells in media supplemented with PDGF-AA, and assessed cell viability, proliferation, genome stability, and tumorigenicity. RESULTS: Counterintuitive to its canonical role as a growth factor, we observed abrupt and massive cell death in PDGF-AA: wild-type cells did not survive, whereas a small fraction of null cells evaded apoptosis. Surviving null cells displayed attenuated proliferation accompanied by whole chromosome gains and losses. After approximately 100 days in PDGF-AA, cells suddenly proliferated rapidly, acquired growth factor independence, and became tumorigenic in immune-competent mice. Transformed cells had an oligodendrocyte precursor-like lineage marker profile, were resistant to platelet derived growth factor receptor alpha inhibition, and harbored highly abnormal karyotypes similar to human GBM. CONCLUSION: This model associates genome instability in neural progenitor cells with chronic exposure to PDGF-AA and is the first to approximate the genomic landscape of human GBM and the first in which the earliest phases of the disease can be studied directly.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Células-Tronco Neurais , Fator de Crescimento Derivado de Plaquetas , Proteína Supressora de Tumor p53 , Animais , Neoplasias Encefálicas/induzido quimicamente , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Células Cultivadas , Glioblastoma/induzido quimicamente , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Ventrículos Laterais/efeitos dos fármacos , Ventrículos Laterais/metabolismo , Ventrículos Laterais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/patologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/metabolismo
9.
World Neurosurg ; 107: 1047.e9-1047.e15, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28804045

RESUMO

BACKGROUND: Glioblastoma (GBM) is a lesion radiologically characterized by magnetic resonance imaging findings, such as ring enhancement with extensive perifocal edema and a butterfly appearance extending into the bilateral lobes. However, these characteristic findings could be changed by antiangiogenic therapy, with decreased contrast enhancement and improved perifocal edema. Herein, we report a case of GBM that arose during treatment with a tyrosine kinase inhibitor for another cancer. CASE DESCRIPTION: A 57-year-old man presented with seizures. Until the seizure onset, he had been treated with the multireceptor tyrosine kinase inhibitor lenvatinib for 4 years for thyroid cancer and its metastasis to the thoracic vertebral body. Magnetic resonance imaging revealed a slightly high intensity lesion in the left frontal base area on T2-weighted or fluid-attenuated inversion recovery images, and the lesion showed only faint enhancement on T1-weighted images after gadolinium administration. Total resection was performed and the histopathologic diagnosis was GBM. However, grade IV histology was observed in only a limited area, and most of the specimen showed lower grade histology with moderate vascularization that lacked microvascular proliferation. CONCLUSIONS: Lenvatinib, which is antiangiogenic, might have affected the radiologic characteristics, as well as the pathology of the tumor. Brain tumors arising during treatment with receptor tyrosine kinases for other cancers could show atypical imaging findings.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Glioblastoma/diagnóstico por imagem , Imageamento por Ressonância Magnética/tendências , Compostos de Fenilureia/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Quinolinas/uso terapêutico , Neoplasias da Glândula Tireoide/tratamento farmacológico , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/induzido quimicamente , Glioblastoma/induzido quimicamente , Humanos , Masculino , Pessoa de Meia-Idade , Compostos de Fenilureia/efeitos adversos , Inibidores de Proteínas Quinases/efeitos adversos , Quinolinas/efeitos adversos , Neoplasias da Glândula Tireoide/diagnóstico por imagem , Resultado do Tratamento
10.
J Clin Neurosci ; 30: 166-168, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26970935

RESUMO

Glioblastoma is an uncommon and aggressive primary brain tumor with incidence of 3 per 100,000 annually. We report a 50-year-old woman diagnosed with glioblastoma within threeyears of induction of fingolimod therapy for relapsing-remitting multiple sclerosis. Fingolimod, an immunomodulating agent used in the treatment of relapsing-remitting multiple sclerosis, has also been suggested to impart a cardioprotective role in heart failure and arrhythmia via activation of P21-activated kinase-1 (Pak1). In the brain, Pak1 activation has been shown to correlate with decreased survival time amongst patients with glioblastoma. A molecular mechanism underlying a link between fingolimod use and glioblastoma development may involve activation of Pak1. To our knowledge, this is the first report of a potential association between fingolimod use and glioblastoma development.


Assuntos
Neoplasias Encefálicas/induzido quimicamente , Neoplasias Encefálicas/diagnóstico por imagem , Cloridrato de Fingolimode/efeitos adversos , Glioblastoma/induzido quimicamente , Glioblastoma/diagnóstico por imagem , Esclerose Múltipla Recidivante-Remitente/diagnóstico por imagem , Feminino , Cloridrato de Fingolimode/uso terapêutico , Humanos , Imunossupressores/efeitos adversos , Imunossupressores/uso terapêutico , Pessoa de Meia-Idade , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico
11.
Pharmacotherapy ; 36(4): 449-54, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26931498

RESUMO

PURPOSE: To quantify the risk of glioblastoma (GBM) and its most aggressive form, glioblastoma multiforme (GBM-M), in patients treated with tumor necrosis factor (TNF) inhibitors. METHODS: Data from the U.S. Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) and the World Health Organization (WHO) Collaborating Centre for International Drug Monitoring were used to perform a disproportionality analysis. We computed reporting odds ratios (RORs) and corresponding 95% confidence intervals for the association between use of TNF inhibitors (infliximab, adalimumab, etanercept, certolizumab, and golimumab) and GBM or GBM-M compared to all other drugs with adverse events reported in the databases. A harmful signal was deemed for a lower limit of the 95% confidence interval above 1. RESULTS: We identified 81 cases of GBM or GBM-M with adalimumab in the U.S. FDA FAERS and 49 cases in the WHO drug monitoring database. For infliximab, 40 and 32 cases were identified in the FAERS and WHO databases, respectfully. Infliximab had the highest association with GBM (WHO: ROR = 7.41 (5.19-10.57), FAERS: ROR = 2.80 [1.89-4.15]). Adalimumab was also highly associated with GBM (WHO: ROR = 3.54 [2.58-4.89], FAERS: ROR = 1.99 [1.41-2.80]). CONCLUSION: Several TNF inhibitors appear to be more strongly associated with GBM compared to other drugs in both the FAERS and WHO databases. Large epidemiologic studies are needed to confirm these findings. Although these results do not demonstrate a cause-and-effect relationship, they warrant further investigation by well-designed epidemiologic studies.


Assuntos
Adalimumab/efeitos adversos , Antirreumáticos/efeitos adversos , Neoplasias Encefálicas/induzido quimicamente , Glioblastoma/induzido quimicamente , Saúde Global , Infliximab/efeitos adversos , Inibidores do Fator de Necrose Tumoral , Sistemas de Notificação de Reações Adversas a Medicamentos , Neoplasias Encefálicas/epidemiologia , Glioblastoma/epidemiologia , Humanos , Risco , Fatores de Necrose Tumoral/metabolismo , Estados Unidos/epidemiologia , United States Food and Drug Administration , Organização Mundial da Saúde
12.
Biomed Res Int ; 2013: 390714, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24307995

RESUMO

Radiosurgery for glioblastoma is limited to the development of resistance, allowing tumor cells to survive and initiate tumor recurrence. Based on our previous work that coadministration of tissue factor and lipopolysaccharide following radiosurgery selectively induced thrombosis in cerebral arteriovenous malformations, achieving thrombosis of 69% of the capillaries and 39% of medium sized vessels, we hypothesized that a rapid and selective shutdown of the capillaries in glioblastoma vasculature would decrease the delivery of oxygen and nutrients, reducing tumor growth, preventing intracranial hypertension, and improving life expectancy. Glioblastoma was formed by implantation of GL261 cells into C57Bl/6 mouse brain. Mice were intravenously injected tissue factor, lipopolysaccharide, a combination of both, or placebo 24 hours after radiosurgery. Control mice received both agents after sham irradiation. Coadministration of tissue factor and lipopolysaccharide led to the formation of thrombi in up to 87 ± 8% of the capillaries and 46 ± 4% of medium sized vessels within glioblastoma. The survival rate of mice in this group was 80% versus no survivor in placebo controls 30 days after irradiation. Animal body weight increased with time in this group (r = 0.88, P = 0.0001). Thus, radiosurgery enhanced treatment with tissue factor, and lipopolysaccharide selectively induces thrombosis in glioblastoma vasculature, improving life expectancy.


Assuntos
Glioblastoma/cirurgia , Neoplasias Experimentais/cirurgia , Radiocirurgia , Trombose/cirurgia , Animais , Terapia Combinada , Modelos Animais de Doenças , Glioblastoma/induzido quimicamente , Glioblastoma/complicações , Glioblastoma/patologia , Humanos , Lipopolissacarídeos/toxicidade , Camundongos , Recidiva Local de Neoplasia , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/patologia , Tromboplastina/administração & dosagem , Trombose/induzido quimicamente , Trombose/complicações , Trombose/patologia
13.
Neurotoxicology ; 35: 1-14, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23261424

RESUMO

Glioblastoma multiforme (GBM) is the most common and aggressive type of human brain tumor. Although considerable efforts to delineate the underlying pathophysiological pathways have been made during the last decades, only very limited progress on treatment have been achieved because molecular pathways that drive the aggressive nature of GBM are largely unknown. Recent studies have emphasized the importance of environmental factors and the role of gene-environment interactions (GEI) in the development of GBM. Factors such as small sample sizes and study costs have limited the conduct of GEI studies in brain tumors however. Additionally, advances in high-throughput microarrays have produced a wealth of information concerning molecular biology of glioma. In particular, microarrays have been used to obtain genetic and epigenetic changes between normal non-tumor tissue and glioma tissue. Due to the relative rarity of gliomas, microarray data for these tumors is often the product of small studies, and thus pooling this data becomes desirable. To address the challenge of small sample sizes and GEI study difficulties, we introduce a comprehensive bioinformatics method using genetic variations (copy number variations and small-scale variations) and environmental data integration that links with glioblastoma (GEG) to identify: (1) genes that interact with chemicals and have genetic variants linked to the development of GBM, (2) important pathways that may be influenced by environmental exposures (or endogenous chemicals), and (3) genes with variants in GBM that have been understudied in relation to GBM development. The first step in our GEG method identified genes responsive to environmental exposures using the Environmental Genome Project, Comparative Toxicology, and Seattle SNPs databases. These environmentally responsive genes were then compared to a curated list of genes containing copy number variation and/or mutations in GBM. This comparison produced a list of genes responsive to the environment and important to GBM that was then further analyzed using gene networking tools such as RSpider, Cytoscape, and DAVID. Using this GEG bioinformatics method we were able to identify 173 genes with the potential to be involved in GEI that may be important to the development of GBM. Sixty five of these environmentally responsive genes have not been reported as important to GBM development, despite several of them having substantial potential for response to chemicals and subsequent disease related actions. The main biological functions of these 173 genes include signaling by nerve growth factor, DNA repair, integrin cell surface interactions, biological oxidations, apoptosis, synaptic transmission, cell cycle checkpoints, and arachidonic acid metabolism. Importantly, some of these functions have been implicated in the development of several cancers, including glioma. In summary, our GEG bioinformatics approach revealed potential gene-environment interactions, and generated new data for hypothesis generation, in GBM.


Assuntos
Neoplasias Encefálicas/induzido quimicamente , Neoplasias Encefálicas/genética , Carcinógenos/toxicidade , Biologia Computacional , Interação Gene-Ambiente , Glioblastoma/induzido quimicamente , Glioblastoma/genética , Algoritmos , Neoplasias Encefálicas/patologia , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Variações do Número de Cópias de DNA , Mineração de Dados , Bases de Dados Genéticas , Exposição Ambiental , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Predisposição Genética para Doença , Glioblastoma/patologia , Humanos , Mutação , Fenótipo , Polimorfismo de Nucleotídeo Único , Medição de Risco , Fatores de Risco
14.
J Toxicol Environ Health A ; 75(8-10): 544-50, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22686314

RESUMO

1,3-Propane sultone (1,3-PS) is a directly alkylating, genotoxic and carcinogenic substance. In rats, 1,3-PS induces local and systemic tumors at multiple sites, including the mammary gland, intestine, hematopoietic system, kidneys, and the central nervous system (CNS), especially in the form of gliomas. In one particular company, 1,3-PS had been manufactured in limited amounts in the 1950s and 1960s, and for a few purposes until the 1970s. The number of individuals having been in contact with the compound occupationally comprised 55 in total. Data were obtained from this group with an open question of legal compensation regarding an occupational disease. Particular emphasis was placed on malignancies occurring among the occupationally exposed persons. As cerebral gliomas are a major type of tumor induced by 1,3-PS experimentally, the occurrence of two glioblastomas among the previously exposed persons was significant. Three intestinal malignancies were recorded within the cases observed. It is also noteworthy that there was one case of a duodenal carcinoma, which is normally a rare human malignancy. Two hematopoietic/lymphatic malignancies were observed, and there was one case of a renal cell carcinoma. The types of malignancies within a group of only 55 exposed persons are surprisingly consistent with the results from rodent studies. Data clearly indicate that 1,3-PS is carcinogenic in humans. Evidence indicates that 12 cases with various neoplasms were legally compensated within the period of 1985-2010 as an occupational disease, based on the "opening clause" of § 9 (2) SGB VII of legislation in Germany.


Assuntos
Carcinógenos/toxicidade , Tiofenos/efeitos adversos , Tiofenos/toxicidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Encefálicas/induzido quimicamente , Neoplasias Encefálicas/epidemiologia , Indústria Química , Estudos de Coortes , Alemanha/epidemiologia , Glioblastoma/induzido quimicamente , Glioblastoma/epidemiologia , Humanos , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/epidemiologia , Masculino , Pessoa de Meia-Idade , Neoplasias/induzido quimicamente , Neoplasias/epidemiologia , Doenças Profissionais/induzido quimicamente , Doenças Profissionais/epidemiologia , Exposição Ocupacional/efeitos adversos , Exposição Ocupacional/estatística & dados numéricos
15.
J Clin Neurosci ; 19(1): 75-80, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22169598

RESUMO

The aim of this study was to determine if there was an association between expression of toll-like receptor 9 (TLR9) in glioblastoma tissue and patient outcome in glioblastoma multiforme. Further, we characterized the direct in vitro effects of the TLR9 agonist, CpG oligodeoxynucleotide (ODN), commonly used as a vaccine adjuvant in cancer immunotherapy, on glioma cells. TLR9 expression was assessed using immunohistochemical techniques, and enzyme-linked immunosorbent assays were used to investigate the expression of other proteins in glioma cells relevant to immunotherapy. There was a highly significant difference in both progression-free survival and overall survival between TLR9+ and TLR9- patients, with poorer outcome in TLR9+ patients. In in vitro glioma cells, there was a positive correlation between the protein levels of TLR9 and both matrix metalloproteinase (MMP)-2 and MMP-9 (p<0.05), but no relationship between TLR9 levels and levels of interleukin-6, transforming growth factor-ß2 or signal transducer and activator of transcription (STAT)-3 (p>0.05). Our data indicate that expression of TLR9 correlates with shorter progression-free survival and overall survival in patients with glioblastoma multiforme. Our findings also indicate that caution is warranted when directly injecting the TLR9 agonist CpG ODN into glioma tissues as part of glioma immunotherapy. Because the CpG ODN is a TLR9 agonist, we recommend caution when using CpG ODN in immunotherapy.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Glioblastoma/metabolismo , Glioblastoma/mortalidade , Receptor Toll-Like 9/biossíntese , Adjuvantes Imunológicos/efeitos adversos , Adulto , Idoso , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/induzido quimicamente , Vacinas Anticâncer/efeitos adversos , Linhagem Celular Tumoral , Intervalo Livre de Doença , Feminino , Glioblastoma/induzido quimicamente , Humanos , Masculino , Pessoa de Meia-Idade , Oligodesoxirribonucleotídeos/efeitos adversos , Receptor Toll-Like 9/genética
16.
Cancer Res ; 71(11): 3963-71, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21464045

RESUMO

The contribution of microenvironment to tumor growth has important implications for optimizing chemotherapeutic response and understanding the biology of recurrent tumors. In this study, we tested the effects of locally administered topotecan on a rat model of glioblastoma that is induced by intracerebral injection of PDGF (platelet-derived growth factor)-IRES (internal ribosome entry site)-GFP (green fluorescent protein)-expressing retrovirus, treated the tumors by convection-enhanced delivery (CED) of topotecan (136 µmol/L) for 1, 4, or 7 days, and then characterized the effects on both the retrovirus-transformed tumor cells (GFP(+) cells) as well as the uninfected glial progenitor cells (GFP(-) cells) that are recruited to the tumor. Topotecan treatment reduced GFP(+) cells about 10-fold and recruited progenitors by about 80-fold while providing a significant survival advantage that improved with greater treatment duration. Regions of glial progenitor ablation occurred corresponding to the anatomic distribution of topotecan as predicted by MRI of a surrogate tracer. Histopathologic changes in recurrent tumors point to a decrease in recruitment, most likely due to the chemotherapeutic ablation of the recruitable progenitor pool.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Topotecan/administração & dosagem , Animais , Neoplasias Encefálicas/induzido quimicamente , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Glioblastoma/induzido quimicamente , Glioblastoma/metabolismo , Glioblastoma/patologia , Imuno-Histoquímica , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neuroglia/metabolismo , Neuroglia/patologia , Fator de Crescimento Derivado de Plaquetas/administração & dosagem , Ratos , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Eur J Med Chem ; 46(5): 1827-34, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21388717

RESUMO

Glycoglycerolipid analogues, derived from 2-O-ß-D-galactosylglycerol, have been synthesized on the base of the structure of natural glycoglycerolipids showing anti-tumor and anti-inflammatory efficacy. These compounds have been previously demonstrated to inhibit phorbol 12-myristate-13-acetate (PMA) induced tumor promotion in mouse skin, but their mechanism of action has never been elucidated. In this work, we studied the effects of glycoglycerolipid analogues on PKC activation induced by PMA and its downstream target molecules, in human fibroblasts. Our results proved that: a) the tested compounds were able to block PKC translocation to the plasma membrane, promoted by PMA, in a dose-dependent manner (IC50: 0.48 µM for the most active compound 2); b) the efficacy of these compounds was strongly connected to their acyl chain linked to galactose; in particular, the addition of hexanoyl and branched chains enhanced PKC inhibition, the presence of a cyclohexane ring and an excessive length of the acyl chain, or its lack, exerted a negative effect; c) the inhibition of PKC translocation blocked enzyme activation and downstream signaling pathways, MAPK and FAK, involved in proliferation and adhesion/migration control. In addition, the branched glycoglycerolipid (compound 2) was able to inhibit PKC translocation and activation in naturally highly PKC activating glioblastoma cells, U87MG. As consequence, U87MG cell proliferation and, especially, migration potential resulted to be markedly reduced (-30% and -84%, respectively). Thus, these results reveal the role of a PKC-dependent mechanism in glycoglycerolipid analogues mediated protective effects and highlight their possible employment in the field of prevention/treatment of cancer.


Assuntos
Antineoplásicos/farmacologia , Membrana Celular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Glicolipídeos/farmacologia , Proteína Quinase C/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos/síntese química , Antineoplásicos/química , Adesão Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Fibroblastos/metabolismo , Glioblastoma/induzido quimicamente , Glioblastoma/metabolismo , Glicolipídeos/síntese química , Glicolipídeos/química , Humanos , Estrutura Molecular , Proteína Quinase C/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/antagonistas & inibidores
18.
J Neurooncol ; 102(3): 485-90, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20680396

RESUMO

The purpose of this case series is to report the unexpected occurrence of four cases of late radiation-induced neurotoxicity with bevacizumab use following radiotherapy to the CNS. We retrospectively reviewed the case records of four patients, three with glioblastoma and one with bone metastases secondary to metastatic breast cancer, who were treated with radiotherapy and developed late radiation-induced neurotoxicity following bevacizumab use. Three cases of optic neuropathy in glioblastoma patients and a single case of Brown-Séquard syndrome in the thoracic spine of a patient with metastatic breast cancer are reported. We hypothesize that bevacizumab use following radiotherapy to the CNS may inhibit vascular endothelial growth factor-dependent repair of normal neural tissue, and thus may increase the risk of late radiation neurotoxicity. Phase III data on the safety and efficacy of bevacizumab use with radiation in the setting of glioblastoma is awaited.


Assuntos
Inibidores da Angiogênese/efeitos adversos , Anticorpos Monoclonais/efeitos adversos , Neoplasias Ósseas/induzido quimicamente , Glioblastoma/induzido quimicamente , Adulto , Idoso , Anticorpos Monoclonais Humanizados , Bevacizumab , Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Feminino , Humanos , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
19.
Horm Metab Res ; 42(11): 781-6, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20665426

RESUMO

Glioblastoma multiforme is the most common and most aggressive type of high grade tumor with a poor prognosis upon discovery. Based on earlier promising results earned with AN-162, a doxorubicin molecule linked to somatostatin (SST) analogue RC-160, it was our aim to determine the effect of AN-162 on DBTRG-05 glioblastoma cell line, and to test its efficacy in experimental brain tumors. We detected the expression of mRNA for somatostatin receptor (SSTR) subtypes 2 and 3 in DBTRG-05 cells with RT-PCR. Using ligand competition assay, specific high affinity receptors for somatostatin were found. The MTT assay showed that both AN-162 and doxorubicin (DOX) significantly inhibited cell proliferation and that there was no significant difference between the effects in vitro. Nude mice were xenografted with DBTRG-05 glioblastoma tumors. AN-162 showed a significant inhibition of tumor growth compared with the control group and the groups treated with equimolar doses of doxorubicin, somatostatin analogue RC-160, or the unconjugated mixture of doxorubicin plus RC-160. The tumor doubling time in the group of animals treated with AN-162 was extended and was significantly different from doubling times in the control group and in the other treatment groups. Our study clearly demonstrates a potent inhibitory effect of AN-162 in experimental glioblastoma, thus suggesting the possibility of its utilization in patients suffering from malignant brain cancer.


Assuntos
2-Hidroxifenetilamina/análogos & derivados , Compostos de Anilina/farmacologia , Glioblastoma/patologia , Somatostatina/análogos & derivados , 2-Hidroxifenetilamina/farmacologia , 2-Hidroxifenetilamina/uso terapêutico , Compostos de Anilina/uso terapêutico , Animais , Bioensaio , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Doxorrubicina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/induzido quimicamente , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
PLoS One ; 4(12): e8536, 2009 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-20046875

RESUMO

Extensive angiogenesis, formation of new capillaries from pre-existing blood vessels, is an important feature of malignant glioma. Several antiangiogenic drugs targeting vascular endothelial growth factor (VEGF) or its receptors are currently in clinical trials as therapy for high-grade glioma and bevacizumab was recently approved by the FDA for treatment of recurrent glioblastoma. However, the modest efficacy of these drugs and emerging problems with anti-VEGF treatment resistance welcome the development of alternative antiangiogenic therapies. One potential candidate is histidine-rich glycoprotein (HRG), a plasma protein with antiangiogenic properties that can inhibit endothelial cell adhesion and migration. We have used the RCAS/TV-A mouse model for gliomas to investigate the effect of HRG on brain tumor development. Tumors were induced with platelet-derived growth factor-B (PDGF-B), in the presence or absence of HRG. We found that HRG had little effect on tumor incidence but could significantly inhibit the development of malignant glioma and completely prevent the occurrence of grade IV tumors (glioblastoma).


Assuntos
Glioblastoma/patologia , Glioblastoma/prevenção & controle , Proteínas/metabolismo , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Galinhas , Vetores Genéticos/genética , Glioblastoma/irrigação sanguínea , Glioblastoma/induzido quimicamente , Humanos , Injeções , Camundongos , Neovascularização Patológica/patologia , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Proteínas Proto-Oncogênicas c-sis/farmacologia , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA