Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38639614

RESUMO

Objective: Glioblastoma is the most common and aggressive type of the central nervous system cancers. Although radiotherapy and chemotherapy are used in the treatment of glioblastoma, survival rates remain unsatisfactory. This study aimed to explore differentially expressed genes (DEGs) based on the survival prognosis of patients with glioblastoma and to establish a model for classifying patients into different risk groups for overall survival. Methods: DEGs from 160 tumor samples from patients with glioblastoma and 5 nontumor samples from other patients in The Cancer Genome Atlas database were identified. Functional enrichment analysis and a protein-protein interaction network were used to analyze the DEGs. The prognostic DEGs were identified by univariate Cox regression analysis. We split patient data from The Cancer Genome Atlas database into a high-risk group and a low-risk group as the training data set. Least absolute shrinkage and selection operator and multiple Cox regression were used to construct a prognostic risk model, which was validated in a test data set from The Cancer Genome Atlas database and was analyzed using external data sets from the Chinese Glioma Genome Atlas database and the GSE74187 and GSE83300 data sets. Furthermore, we constructed and validated a nomogram to predict survival of patients with glioblastoma. Results: A total of 3572 prognostic DEGs were identified. Functional analysis indicated that these DEGs were mainly involved in the cell cycle and focal adhesion. Least absolute shrinkage and selection operator regression identified 3 prognostic DEGs (EFEMP2, PTPRN, and POM121L9P), and we constructed a prognostic risk model. The receiver operating characteristic curve analysis showed that the areas under the curve were 0.83 for the training data set and 0.756 for the test data set. The predictive performance of the prognostic risk model was validated in the 3 external data sets. The nomogram showed that the prognostic risk model was reliable and that the accuracy of predicting survival in each patient was high. Conclusion: The prognostic risk model can effectively classify patients with glioblastoma into high-risk and low-risk groups in terms of overall survival rate, which may help select high-risk patients with glioblastoma for more intensive treatment.

2.
BMC Surg ; 24(1): 4, 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-38166900

RESUMO

BACKGROUND: Corpus callosum glioblastoma (ccGBM) is a specific type of GBM and has worse outcomes than other non-ccGBMs. We sought to identify whether en-bloc resection of ccGBMs based on T2-FLAIR imaging contributes to clinical outcomes and can achieve a satisfactory balance between maximal resection and preservation of neurological function. METHODS: A total of 106 adult ccGBM patients (including astrocytoma, WHO grade 4, IDH mutation, and glioblastoma) were obtained from the Department of Neurosurgery in Nanfang Hospital between January 2008 and December 2018. The clinical data, including gender, age, symptoms, location of tumor, involvement of eloquent areas, extent of resection (EOR), pre- and postoperative Karnofsky Performance Status (KPS) scales, and National Institute of Health stroke scale (NIHSS) scores were collected. Propensity score matching (PSM) analysis was applied to control the confounders for analyzing the relationship between the en-bloc technique and EOR, and the change in the postoperative KPS scales and NIHSS scores. RESULTS: Applying the en-bloc technique did not negatively affect the postoperative KPS scales compared to no-en-bloc resection (P = 0.851 for PSM analysis) but had a positive effect on preserving or improving the postoperative NIHSS scores (P = 0.004 for PSM analysis). A positive correlation between EOR and the en-bloc technique was identified (r = 0.483, P < 0.001; r = 0.720, P < 0.001 for PSM analysis), indicating that applying the en-bloc technique could contribute to enlarged maximal resection. Further survival analysis confirmed that applying the en-bloc technique and achieving supramaximal resection could significantly prolong OS and PFS, and multivariate analysis suggested that tumor location, pathology, EOR and the en-bloc technique could be regarded as independent prognostic indicators for OS in patients with ccGBMs, and pathology, EOR and the en-bloc technique were independently correlated with patient's PFS. Interestingly, the en-bloc technique also provided a marked reduction in the risk of tumor recurrence compared with the no-en-bloc technique in tumors undergoing TR, indicating that the essential role of the en-bloc technique in ccGBM surgery (HR: 0.712; 95% CI: 0.535-0.947; P = 0.02). CONCLUSIONS: The en-bloc technique could contribute to achieving an enlarged maximal resection and could significantly prolong overall survival and progression-free survival in patients with ccGBMs.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Adulto , Humanos , Glioblastoma/cirurgia , Estudos Retrospectivos , Resultado do Tratamento , Corpo Caloso/cirurgia , Corpo Caloso/patologia , Neoplasias Encefálicas/cirurgia , Recidiva Local de Neoplasia/cirurgia , Prognóstico , Procedimentos Neurocirúrgicos/métodos
3.
J Neuroinflammation ; 20(1): 80, 2023 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-36944954

RESUMO

BACKGROUND: The unique intracranial tumor microenvironment (TME) contributes to the immunotherapy failure for glioblastoma (GBM), thus new functional protein targets are urgently needed. Alternative splicing is a widespread regulatory mechanism by which individual gene can express variant proteins with distinct functions. Moreover, proteins located in the cell plasma membrane facilitate targeted therapies. This study sought to obtain functional membrane protein isoforms from GBM TME. METHODS: With combined single-cell RNA-seq and bulk RNA-seq analyses, novel candidate membrane proteins generated by prognostic splicing events were screened within GBM TME. The short isoform of MS4A7 (MS4A7-s) was selected for evaluation by RT-PCR and western blotting in clinical specimens. Its clinical relevance was evaluated in a GBM patient cohort. The function of MS4A7-s was identified by in vitro and in vivo experiments. MS4A7-s overexpression introduced transcriptome changes were analyzed to explore the potential molecular mechanism. RESULTS: The main expression product, isoform MS4A7-s, generated by exon skipping, is an M2-specific plasma membrane protein playing a pro-oncogenic role in GBM TME. Higher expression of MS4A7-s correlates with poor prognosis in a GBM cohort. In vitro cell co-culture experiments, intracranial co-injection tumorigenesis assay, and RNA-seq suggest MS4A7-s promotes activation of glioma-associated macrophages' (GAMs) PI3K/AKT/GSK3ß pathway, leading to M2 polarization, and drives malignant progression of GBM. CONCLUSIONS: MS4A7-s, a novel splicing isoform of MS4A7 located on the surface of GAMs in GBM TME, is a predictor of patient outcome, which contributes to M2 polarization and the malignant phenotype of GBM. Targeting MS4A7-s may constitute a promising treatment for GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Proteínas de Membrana , Humanos , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Macrófagos/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Isoformas de Proteínas/genética , Fatores de Transcrição/metabolismo , Microambiente Tumoral
4.
J Neurooncol ; 164(3): 557-568, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37783878

RESUMO

PURPOSE: Resection beyond the contrast-enhanced zone contributed to reduce tumor burden and prolong survival in glioblastomas. The optimal extent of resection (EOR) and how to achieve it are worthy of continuous investigation for obtaining a satisfactory balance between maximal resection and the preservation of neurological function. METHODS: A total of 340 adult supratentorial lobar glioblastomas (included astrocytoma, WHO 4, IDH mutation and glioblastoma) were retrospectively evaluated. The clinical data, EOR, technique of resection, postoperative complications, overall survival (OS) and progression-free survival (PFS) were assessed by univariate, multivariate and propensity score matched analysis. Histological staining was performed to comprehend the effect of the membranous structures and the cell distribution in tumoral and peritumoral regions. RESULTS: Supramaximal resection (SMR) was confirmed as resection with 100% EORCE and > 50% EORnCE in glioblastomas by Cox proportional hazards model. Histological results showed SMR reduced the cell density of surgical edge compared to total resection. En-bloc technique based on membranous structures, which had blocking effect on tumoral invasion, contributed to achieve SMR. Moreover, applying en-bloc technique and achieving SMR did not additionally deteriorate neurological function and had similarly effects on the improvement of neurological function. Multivariate analysis confirmed that IDH1 status, technique of resection and EOR were independently correlated with PFS, and > 64 years old, IDH1 status, technique of resection, EOR and preoperative NIHSS were independently correlated with OS. CONCLUSIONS: Applying en-bloc technique and achieving SMR, which could reduce tumor burden and did not increase additional complications, both had remarkedly positive effects on clinical outcomes in patients with primary supratentorial lobar glioblastomas.


Assuntos
Astrocitoma , Neoplasias Encefálicas , Glioblastoma , Neoplasias Supratentoriais , Adulto , Humanos , Pessoa de Meia-Idade , Glioblastoma/patologia , Estudos Retrospectivos , Carga Tumoral , Neoplasias Supratentoriais/genética , Astrocitoma/cirurgia , Procedimentos Neurocirúrgicos/métodos , Neoplasias Encefálicas/cirurgia , Neoplasias Encefálicas/patologia , Resultado do Tratamento
5.
J Phys Chem A ; 127(45): 9473-9482, 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37824456

RESUMO

The detailed mechanism for NHC-Cu(I)-catalyzed intermolecular nucleophilic substitution of the C-H bonds at aniline (2-methyl-N-methoxyaniline) was studied via DFT methods to reveal the essence of the selectivity. Calculations revealed that the meta C-H functionalization proceeds via two nucleophilic attacks on the aromatic ring rather than a one-step meta C-H substitution to give the experimentally observed major product. The reaction is initiated by activation of the substrate via oxidative addition with an NHC-Cu(I) catalyst, through which an umpolung occurs at the ring. From the activated intermediate, methoxyl group transfer to benzyl forms a resting state, while a nucleophile can attack the ortho position of benzyl to form a more stable intermediate. The nucleophile group can then transfer to the meta position by a 1,2-Wagner-Meerwein rearrangement to form the final product through a proton shuttle. In contrast, other transfer processes affording ortho- or para-substituted products encounter higher activation barriers. This work investigates the relationship of product selectivity with the umpolung of the aromatic ring, as well as the priority of a nucleophilic attack at the ortho position of the aromatic, 1,2-Wagner-Meerwein rearrangement from the ortho-substituted intermediate, and proton shuttle from the meta-substituted intermediate.

6.
Angew Chem Int Ed Engl ; 62(19): e202218577, 2023 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-36716145

RESUMO

Iron-catalyzed asymmetric amination of C(sp3 )-H bonds is appealing for synthetic applications due to the biocompatibility and high earth abundance of iron, but examples of such reactions are sparse. Herein we describe chiral iron complexes of meso- and ß-substituted-porphyrins that can catalyze asymmetric intramolecular C(sp3 )-H amination of aryl and arylsulfonyl azides to afford chiral indolines (29 examples) and benzofused cyclic sulfonamides (17 examples), respectively, with up to 93 % ee (yield: up to 99 %) using 410 nm light under mild conditions. Mechanistic studies, including DFT calculations, for the reactions of arylsulfonyl azides reveal that the Fe(NSO2 Ar) intermediate generated in situ under photochemical conditions reacts with the C(sp3 )-H bond through a stepwise hydrogen atom transfer/radical rebound mechanism, with enantioselectivity arising from cooperative noncovalent interactions between the Fe(NSO2 Ar) unit and the peripheral substituents of the chiral porphyrin scaffold.

7.
J Am Chem Soc ; 144(27): 11975-11979, 2022 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-35763716

RESUMO

Countless inorganic materials are prepared via high temperature solid-state reaction of mixtures of reagents powders. Understanding and controlling the phenomena that limit these solid-state reactions is crucial to designing reactions for new materials synthesis. Here, focusing on topotactic ion-exchange between NaFeO2 and LiBr as a model reaction, we manipulate the mesoscale reaction architecture and transport pathways by changing the packing and interfacial contact between reagent particles. Through analysis of in situ synchrotron X-ray diffraction data, we identify multiple kinetic regimes that reflect transport limitations on different length scales: a fast kinetic regime in the first minutes of the reaction and a slow kinetic regime that follows. The fast kinetic regime dominates the observed reaction progress and depends on the reagent packing; this challenges the view that solid-state reactions are necessarily slow. Using a phase-field model, we simulated the reaction process and showed that particles without direct contact to the other reactant phases experience large reduction in the reaction rate, even when transport hindrance at particle-particle contacts is not considered.

8.
Cancer Cell Int ; 22(1): 151, 2022 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-35410293

RESUMO

BACKGROUND: Compared with the proneural (PN) subtype of glioblastoma (GBM), the mesenchymal (MES) subtype is more invasive and immune evasive and is closely related to poor prognosis. Here, we used transcriptome data and experimental evidence to indicate that CUB domain-containing protein 1 (CDCP1) is a novel regulator that facilitates the transformation of PN-GBM to MES-GBM. METHODS: The mRNA expression data of CDCP1 in glioma were collected from the TCGA, CGGA and GEO databases, and in vitro experiments verified CDCP1 expression in glioma tissue samples. Independent prognostic analysis revealed the correlation of the CDCP1 expression level and patient survival. Bioinformatics analysis and experiments verified the biological function of CDCP1. Multivariate proportional hazards models and a PPI network were used to select key genes. A prognostic risk model for predicting the survival of glioma patients was constructed based on the selected genes. RESULTS: The results showed that the expression of CDCP1 increased with increasing tumor grade and that the overexpression of CDCP1 correlated with a poor prognosis. CDCP1 was highly expressed in MES-GBM but weakly expressed in PN-GBM. The risk model (considering CDCP1 combined with CD44 and ITGAM expression) could represent a tool for predicting survival and prognosis in glioma patients. CONCLUSIONS: Our study indicates that CDCP1 plays an important role in facilitating the transformation of PN-GBM to MES-GBM.

9.
Anticancer Drugs ; 33(1): 100-104, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34232949

RESUMO

Epithelioid glioblastoma (E-GBM) is a recently described variant of glioblastoma (GBM) which is associated with short survival and now added as a provisional entity to WHO 2016 classification of central nervous system tumors. About half of these tumors show the BRAF mutant. Therefore, this is a target of special interest for this group of patients. Meanwhile, unlike conventional glioblastoma, E-GBM lacks specific prognostic markers. We described a case of a long-term surviving 37-years-old men patient diagnosed with a BRAF V600E and TERT mutated E-GBM with wild-type in the isocitrate dehydrogenase gene (IDH wild-type). The tumor displayed atypical exophytic growth, an obvious proliferation of vascular endothelial cells, especially tumor tissue can be seen under subarachnoid space. Notably, tumor tissue was found under subarachnoid space. After postoperative conventional treatment options were exhausted, vemurafenib treatment was initiated. The patient remained clinically stable, and follow-up magnetic resonance images were consistent with stable disease for the following fifteen months up to now. Whole-exome sequencing analysis and RNA-seq results of formalin-fixed and paraffin-embedded tissue revealed nine mutant genes (AHNAK2, BFSP1, BRAF, CNTNAP3, DNHD1, MTOR, NFATC3, NOM1). For E-GBM patients, the use of BRAF inhibitors combined with inhibitors of these seven genes may be a useful remedial treatment option.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Vemurafenib/uso terapêutico , Adulto , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Masculino , Proteínas Proto-Oncogênicas B-raf/genética , Telomerase/genética , Sequenciamento do Exoma
10.
J Neurooncol ; 157(1): 15-26, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35187626

RESUMO

PURPOSE: Glioblastoma multiforme (GBM) is a primary brain tumor with devastating prognosis. Although the O6-methylguanine-DNA methyltransferase (MGMT) leads to inherent temozolomide (TMZ) resistance, approximately half of GBMs were sufficient to confer acquired TMZ resistance, which express low levels of MGMT. The purpose of this study was to investigate the underlying mechanisms of the acquired TMZ resistance in MGMT-deficient GBM. METHODS: The function of Down syndrome critical region protein 3 (DSCR3) on MGMT-deficient GBM was investigated in vitro and in an orthotopic brain tumor model in mice. Purification of plasma membrane proteins by membrane-cytoplasmic separation and subsequent label free-based quantitative proteomics were used to identified potential protein partners for DSCR3. Immunofluorescence was performed to show the reverse transport of solute carrier family 38 member 1 (SLC38A1) mediated by DSCR3. RESULTS: DSCR3 is upregulated in MGMT-deficient GBM cells during TMZ treatment. Both DSCR3 and SLC38A1 were highly expressed in recurrent GBM patients. Silencing DSCR3 or SLC38A1 expression can increase TMZ sensitivity in MGMT-deficient GBM cells. Combination of proteomics and in vitro experiments show that DSCR3 directly binds internalized SLC38A1 to mediate its sorting into recycling pathway, which maintains the abundance on plasma membrane and enhances uptake of glutamine in MGMT-deficient GBM cells. CONCLUSIONS: DSCR3 is a crucial regulator of acquired TMZ resistance in MGMT-deficient GBM. The DSCR3-dependent recycling of SLC38A1 maintains its abundance on plasma membrane, leading to tumor progression and acquired TMZ resistance in MGMT-deficient GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Sistema A de Transporte de Aminoácidos , Animais , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Resistencia a Medicamentos Antineoplásicos , Glioblastoma/patologia , Humanos , Camundongos , Temozolomida/farmacologia , Temozolomida/uso terapêutico
11.
BMC Neurol ; 22(1): 390, 2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36271359

RESUMO

BACKGROUND: The incidence of osteosarcoma as a secondary neoplasm in glioblastoma patient is extremely rare. The genetic characteristic still remains unclear until now. CASE DESCRIPTION: We reported a 47-year-old female patient with multiple intracranial disseminations and infiltrations (splenium of the corpus callosum and lateral ventricular wall) of a rapid progressive glioblastoma underwent occipital craniotomy and total resection of all the enhancing lesions. Whole-exome sequencing and pathological examination revealed glioblastoma, IDH1 wild type, PTEN deficient, TERT mutated, NF1mutated, MGMT unmethylated. After surgery, the patient received combined therapeutic regimen of TTFields (tumor-treating fields) plus pembrolizumab plus temozolomide and TTFields plus everolimus, which displayed significant clinical benefits. During the combined therapeutic course, an extremely rare secondary malignant neoplasm occurred, femur MR and pathological detection of biopsy tissue demonstrated osteosarcoma. The result of whole-exome sequencing revealed 7 germline mutated genes (EPAS1, SETD2, MSH3, BMPR1A, ERCC4, CDH1, AR). Bioinformatic analysis showed the two germline mutations (MSH3 and ERCC4) induced deficiency in the DNA repair machinery, which resulting in the accumulation of mutations and may generate neoantigens contributing to the development of a secondary osteosarcoma in this case. CONCLUSION: Individualized combination therapies based on whole-exome sequencing displayed significant clinical benefits in this case. Germline MSH3 and ERCC4 mutation may induce a secondary osteosarcoma in glioblastoma patients.


Assuntos
Neoplasias Ósseas , Neoplasias Encefálicas , Glioblastoma , Osteossarcoma , Feminino , Humanos , Pessoa de Meia-Idade , Glioblastoma/complicações , Glioblastoma/genética , Glioblastoma/terapia , Temozolomida/uso terapêutico , Sequenciamento do Exoma , Everolimo/uso terapêutico , Osteossarcoma/complicações , Osteossarcoma/genética , Osteossarcoma/tratamento farmacológico , Mutação/genética , Neoplasias Ósseas/complicações , Neoplasias Ósseas/genética , Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética
12.
Environ Res ; 204(Pt A): 112011, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34492276

RESUMO

Human brain has a complex structure and is able to perform powerful functions. Blood-brain barrier blocks the entry of foreign substances and maintains the homeostasis of the brain. However, some exogenous substances are still able to pass through the blood-brain barrier, with distribution patterns yet to be clarified. Perfluoroalkyl and polyfluoroalkyl substances (PFASs), including perfluoroalkyl carboxylic acids (PFCAs), perfluoroalkyl sulfonic acids (PFSAs), a precursor (perfluorooctane sulfonamide that can be degraded to other substances), and emerging PFASs, were analyzed for the first time in living human brain glioma. The target compounds were detected and quantified in 25 out of 26 glioma samples. The concentration range of ∑PFAS was < RL-51 ng g-1 wet weight (applied to all reported concentrations), with a median of 2.9 ng g-1. The most abundant compound was PFCAs (40%), followed by PFSAs (28%), emerging PFASs (22%), and perfluorooctane sulfonamide (10%). Abundant alternatives PFASs, including short-chain PFCAs, short-chain PFSAs, and emerging PFASs (52% of ∑PFAS), were found in the glioma samples, supporting the notion that low molecular weight exogenous compounds have high permeability to cross the blood-brain barrier and accumulate in brain tissue. Gender difference was not significant (p > 0.05) in the concentrations of PFASs in the glioma samples. Concentrations of PFASs increased with increasing age, from 0.61 ng g-1 (0-14 years old) to 1.6 ng g-1 (>48 years old), with no significant linear correlation with age. The present study suggested that glioma is an effective indicator for monitoring exogenous contaminants in brain tissues.


Assuntos
Ácidos Alcanossulfônicos , Fluorocarbonos , Glioma , Poluentes Químicos da Água , Adolescente , Ácidos Alcanossulfônicos/análise , Encéfalo , Criança , Pré-Escolar , Monitoramento Ambiental , Fluorocarbonos/análise , Humanos , Lactente , Recém-Nascido , Pessoa de Meia-Idade , Poluentes Químicos da Água/análise
13.
BMC Neurol ; 20(1): 310, 2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-32819307

RESUMO

BACKGROUND: Age is associated with the prognosis of glioma patients, but there is no uniform standard of age-group classification to evaluate the prognosis of glioma patients. In this study, we aimed to establish an age group classification for risk stratification in glioma patients. METHODS: 1502 patients diagnosed with gliomas at Nanfang Hospital between 2000 and 2018 were enrolled. The WHO grade of glioma was used as a dependent variable to evaluate the effect of age on risk stratification. The evaluation model was established by logistic regression, and the Akaike information criterion (AIC) value of the model was used to determine the optimal cutoff points for age-classification. The differences in gender, WHO grade, pathological subtype, tumor cell differentiation, tumor size, tumor location, and molecular markers between different age groups were analyzed. The molecular markers included GFAP, EMA, MGMT, P53, NeuN, Oligo2, EGFR, VEGF, IDH1, Ki-67, PR, CD3, H3K27M, TS, and 1p/19q status. RESULTS: The proportion of men with glioma was higher than that of women with glioma (58.3% vs 41.7%). Analysis of age showed that appropriate classifications of age group were 0-14 years old (pediatric group), 15-47 years old (youth group), 48-63 years old (middle-aged group), and ≥ 64 years old (elderly group).The proportions of glioblastoma and large tumor size (4-6 cm) increased with age (p = 0.000, p = 0.018, respectively). Analysis of the pathological molecular markers across the four age groups showed that the proportion of patients with larger than 10% area of Ki-67 expression or positive PR expression increased with age (p = 0.000, p = 0.017, respectively). CONCLUSIONS: Appropriate classifications of the age group for risk stratification are 0-14 years old (pediatric group), 15-47 years old (young group), 48-63 years old (middle age group) and ≥ 64 years old (elderly group). This age group classification is effective in evaluating the risk of glioblastoma in glioma patients.


Assuntos
Fatores Etários , Neoplasias Encefálicas/classificação , Glioma/classificação , Medição de Risco , Adolescente , Adulto , Idoso , Neoplasias Encefálicas/patologia , Criança , Pré-Escolar , Feminino , Glioma/patologia , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Prognóstico , Adulto Jovem
14.
Brain ; 142(8): 2352-2366, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31347685

RESUMO

The acquisition of temozolomide resistance is a major clinical challenge for glioblastoma treatment. Chemoresistance in glioblastoma is largely attributed to repair of temozolomide-induced DNA lesions by O6-methylguanine-DNA methyltransferase (MGMT). However, some MGMT-deficient glioblastomas are still resistant to temozolomide, and the underlying molecular mechanisms remain unclear. We found that DYNC2H1 (DHC2) was expressed more in MGMT-deficient recurrent glioblastoma specimens and its expression strongly correlated to poor progression-free survival in MGMT promotor methylated glioblastoma patients. Furthermore, silencing DHC2, both in vitro and in vivo, enhanced temozolomide-induced DNA damage and significantly improved the efficiency of temozolomide treatment in MGMT-deficient glioblastoma. Using a combination of subcellular proteomics and in vitro analyses, we showed that DHC2 was involved in nuclear localization of the DNA repair proteins, namely XPC and CBX5, and knockdown of either XPC or CBX5 resulted in increased temozolomide-induced DNA damage. In summary, we identified the nuclear transportation of DNA repair proteins by DHC2 as a critical regulator of acquired temozolomide resistance in MGMT-deficient glioblastoma. Our study offers novel insights for improving therapeutic management of MGMT-deficient glioblastoma.


Assuntos
Neoplasias Encefálicas/genética , Dineínas do Citoplasma/genética , Reparo do DNA/genética , Resistencia a Medicamentos Antineoplásicos/genética , Glioblastoma/genética , Animais , Antineoplásicos Alquilantes , Neoplasias Encefálicas/metabolismo , Homólogo 5 da Proteína Cromobox , Dineínas do Citoplasma/metabolismo , Metilases de Modificação do DNA/deficiência , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/deficiência , Enzimas Reparadoras do DNA/genética , Glioblastoma/metabolismo , Xenoenxertos , Humanos , Camundongos , Temozolomida , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
15.
Cell Biochem Funct ; 38(2): 185-194, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31833081

RESUMO

Glioblastoma (GBM) is the most malignant and aggressive glioma, which has a very poor prognosis. Temozolomide (TMZ) is still a first-line treatment, but resistance is inevitable even in MGMT-deficient glioblastoma cells. The aims of this study were to comprehend the effect of TMZ on nucleus and the underlying mechanism of acquired TMZ resistance in MGMT-deficient GBM. We show the changes of nuclear proteome in the MGMT-deficient GBM U87 cells treated with TMZ for 1 week. Label-free-based quantitative proteomics were used to investigate nuclear protein abundance change. Subsequently, gene ontology function annotation, KEGG pathway analysis, protein-protein interaction (PPI) network construction analysis of DAPs, and immunofluorescence were applied to validate the quality of proteomics. In total, 457 (455 gene products) significant DAPs were identified, of which 327 were up-regulated and 128 were down-regulated. Bioinformatics analysis uncovered RAD50, MRE11, UBR5, MSH2, MSH6, DDB1, DDB2, RPA1, RBX1, CUL4A, and CUL4B mainly enriched in DNA damage repair related pathway and constituted a protein-protein interaction network. Ribosomal proteins were down-regulated. Cells were in a stress-responsive state, while the entire metabolic level was lowered. SIGNIFICANCE OF THE STUDY: In U87 cell treated with TMZ for 1 week, which resulted in DNA damage, we found various proteins dysregulated in the nucleus. Some proteins related to the DNA damage repair pathway were up-regulated, and there was a strong interaction. We believe this is the potential clues of chemotherapy resistance in tumour cells. These proteins can be used as indicators of tumour resistance screening in the future.


Assuntos
Neoplasias Encefálicas/patologia , Núcleo Celular/efeitos dos fármacos , Dano ao DNA , Glioblastoma/patologia , Glioma/patologia , Temozolomida/farmacologia , Linhagem Celular Tumoral , Núcleo Celular/patologia , Biologia Computacional , Reparo do DNA , Humanos , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteoma , Proteômica/métodos , Espectrometria de Massas por Ionização por Electrospray
16.
Neurodegener Dis ; 20(4): 123-130, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33735873

RESUMO

OBJECTIVES: The present study aimed to study whether combined inflow-based vascular-space-occupancy (iVASO) MR imaging (MRI) and diffusion-weighted imaging (DWI) improve the diagnostic accuracy in the preoperative grading of gliomas. METHODS: Fifty-one patients with histopathologically confirmed diffuse gliomas underwent preoperative structural MRI, iVASO, and DWI. We performed 2 qualitative consensus reviews: (1) structural MR images alone and (2) structural MR images with iVASO and DWI. Relative arteriolar cerebral blood volume (rCBVa) and minimum apparent diffusion coefficient (mADC) were compared between low-grade and high-grade gliomas. Receiver operating characteristic (ROC) curve analysis was performed to compare the tumor grading efficiency of rCBVa, mADC, and the combination of the two parameters. RESULTS: Two observers diagnosed accurate tumor grade in 40 of 51 (78.4%) patients in the first review and in 46 of 51 (90.2%) in the second review. Both rCBVa and mADC showed significant differences between low-grade and high-grade gliomas. ROC analysis gave a threshold value of 1.52 for rCBVa and 0.85 × 10-3 mm2/s for mADC to provide a sensitivity and specificity of 88.0 and 81.2% and 100.0 and 68.7%, respectively. The area under the ROC curve (AUC) was 0.87 and 0.85 for rCBVa and mADC, respectively. The combination of rCBVa and mADC values increased the AUC to 0.92. CONCLUSION: The combined application of iVASO and DWI may improve the diagnostic accuracy of glioma grading.


Assuntos
Neoplasias Encefálicas , Glioma , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/cirurgia , Imagem de Difusão por Ressonância Magnética , Glioma/diagnóstico por imagem , Glioma/cirurgia , Humanos , Interpretação de Imagem Assistida por Computador , Gradação de Tumores , Curva ROC , Sensibilidade e Especificidade
17.
Pediatr Res ; 83(3): 645-654, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29166381

RESUMO

BackgroundWe aimed to develop a risk score to improve the prediction of severe obesity in pediatric craniopharyngiomas (PCs).MethodsOverall, 612 consecutive PCs were prospectively enrolled from six hospitals. Data from 404 participants were analyzed. Participants from three of the six hospitals (n=290) were used to develop a risk score. External validation of the developed risk score was conducted using the participants from the other three hospitals (n=114). Sequential logistic regression was used to develop and validate the risk score. The c statistic and a calibration plot were used to assess the discrimination and calibration of the proposed risk score.ResultsThe overall frequency of severe obesity was 16.1% (65/404). The risk score employed a scale of 0-16 and demonstrated good discriminative power, with an optimism-corrected c statistic of 0.820. Similar results were obtained from external validation, with a c statistic of 0.821. The risk score showed good calibration, with no apparent over- or under-prediction observed in the calibration plots.ConclusionsThis novel risk score is a simple tool that can help clinicians assess the risk of severe obesity in PCs, thereby helping to plan and initiate the most appropriate disease management for these patients in time.


Assuntos
Craniofaringioma/diagnóstico por imagem , Obesidade Mórbida/diagnóstico , Obesidade Infantil/diagnóstico , Neoplasias Hipofisárias/diagnóstico por imagem , Medição de Risco/métodos , Adolescente , Calibragem , Criança , Pré-Escolar , Craniofaringioma/complicações , Feminino , Seguimentos , Humanos , Modelos Logísticos , Masculino , Modelos Estatísticos , Obesidade Mórbida/complicações , Obesidade Infantil/complicações , Neoplasias Hipofisárias/complicações , Curva ROC , Análise de Regressão , Reprodutibilidade dos Testes , Fatores de Risco
18.
J Craniofac Surg ; 27(4): e409-13, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27213742

RESUMO

To analyze the element composition and microstructure of calcification in craniopharyngiomas and to explore the differences among differing degrees of calcification, 50 consecutive patients with craniopharyngioma were selected. X-ray diffraction analysis and energy-dispersive X-ray spectroscopy analysis were performed on the calcified plaques isolated from the tumor specimens. All calcified plaques were constituted of hydroxyapatite crystals and some amorphous materials. The main elements for the analysis were calcium, phosphate, carbon, and oxygen. There were significant differences among groups of differing degrees of calcification in the percentage composition of calcium, phosphorus, and carbon (P < 0.05), in which the element content of calcium and phosphorus had a positive correlation with the extent of calcification (rp = 0.745 and 0.778, respectively, P < 0.01), while the element content of carbon had a negative correlation with the extent of calcification (rp =-0.526, P <0.01). The calcium, phosphorus, and carbon content are different in calcified plaques with different extents of calcification. The element content of calcium, phosphorus, and carbon influences the degree of calcification.


Assuntos
Craniofaringioma/diagnóstico , Hipófise/ultraestrutura , Neoplasias Hipofisárias/diagnóstico , Adolescente , Adulto , Idoso , Calcinose/patologia , Cálcio/análise , Criança , Pré-Escolar , Craniofaringioma/química , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fosfatos/análise , Fósforo/análise , Hipófise/metabolismo , Neoplasias Hipofisárias/química , Espectrometria por Raios X , Adulto Jovem
19.
Mol Biomed ; 5(1): 6, 2024 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-38342791

RESUMO

Cancer is associated with a high degree of heterogeneity, encompassing both inter- and intra-tumor heterogeneity, along with considerable variability in clinical response to common treatments across patients. Conventional models for tumor research, such as in vitro cell cultures and in vivo animal models, demonstrate significant limitations that fall short of satisfying the research requisites. Patient-derived tumor organoids, which recapitulate the structures, specific functions, molecular characteristics, genomics alterations and expression profiles of primary tumors. They have been efficaciously implemented in illness portrayal, mechanism exploration, high-throughput drug screening and assessment, discovery of innovative therapeutic targets and potential compounds, and customized treatment regimen for cancer patients. In contrast to conventional models, tumor organoids offer an intuitive, dependable, and efficient in vitro research model by conserving the phenotypic, genetic diversity, and mutational attributes of the originating tumor. Nevertheless, the organoid technology also confronts the bottlenecks and challenges, such as how to comprehensively reflect intra-tumor heterogeneity, tumor microenvironment, tumor angiogenesis, reduce research costs, and establish standardized construction processes while retaining reliability. This review extensively examines the use of tumor organoid techniques in fundamental research and precision medicine. It emphasizes the importance of patient-derived tumor organoid biobanks for drug development, screening, safety evaluation, and personalized medicine. Additionally, it evaluates the application of organoid technology as an experimental tumor model to better understand the molecular mechanisms of tumor. The intent of this review is to explicate the significance of tumor organoids in cancer research and to present new avenues for the future of tumor research.


Assuntos
Neoplasias , Organoides , Medicina de Precisão , Humanos , Organoides/patologia , Organoides/efeitos dos fármacos , Medicina de Precisão/métodos , Neoplasias/patologia , Neoplasias/genética , Neoplasias/tratamento farmacológico , Animais , Microambiente Tumoral
20.
Environ Int ; 187: 108719, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38718677

RESUMO

Per- and polyfluoroalkyl substances (PFAS) have been shown to penetrate the blood-brain barrier (BBB) and accumulate in human brain. The BBB transmission and accumulation efficiency of PFAS, as well as the potential health risks from human co-exposure to legacy and emerging PFAS due to differences in transport efficiency, need to be further elucidated. In the present pilot study, 23 plasma samples from glioma patients were analyzed for 17 PFAS. The concentrations of PFAS in six paired brain tissue and plasma samples were used to calculate the BBB transmission efficiency of PFAS (RPFAS). This RPFAS analysis was conducted with utmost care and consideration amid the limited availability of valuable paired samples. The results indicated that low molecular weight PFAS, including short-chain and emerging PFAS, may have a greater potential for accumulation in brain tissue than long-chain PFAS. As an alternative to perfluorooctane sulfonic acid (PFOS), 6:2 chlorinated polyfluorinated ether sulfonate (6:2 Cl-PFESA) exhibited brain accumulation potential similar to that of PFOS, suggesting it may not be a suitable substitute concerning health risk in brain. The BBB transmission efficiencies of perfluorooctanoic acid, PFOS, and 6:2 Cl-PFESA showed similar trends with age, which may be an important factor influencing the entry of exogenous compounds into the brain. A favorable link between perfluorooctane sulfonamide (FOSA) and the development and/or progression of glioma may be implicated by a strong positive correlation (r2 = 0.94; p < 0.01) between RFOSA and Ki-67 (a molecular marker of glioma). However, a causal relationship between RFOSA and glioma incidence were not established in the present study. The present pilot study conducted the first examination of BBB transmission efficiency of PFAS from plasma to brain tissue and highlighted the importance of reducing and/or controlling exposure to PFAS.


Assuntos
Barreira Hematoencefálica , Fluorocarbonos , Humanos , Barreira Hematoencefálica/metabolismo , Projetos Piloto , Fluorocarbonos/sangue , Pessoa de Meia-Idade , Feminino , Adulto , Masculino , Glioma , Idoso , Poluentes Ambientais/sangue , Exposição Ambiental , Ácidos Alcanossulfônicos/sangue , Encéfalo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA