Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.048
Filtrar
2.
Adv Sci (Weinh) ; 9(21): e2201436, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35619544

RESUMO

The perivascular niche (PVN) is a glioblastoma tumor microenvironment (TME) that serves as a safe haven for glioma stem cells (GSCs), and acts as a reservoir that inevitably leads to tumor recurrence. Understanding cellular interactions in the PVN that drive GSC treatment resistance and stemness is crucial to develop lasting therapies for glioblastoma. The limitations of in vivo models and in vitro assays have led to critical knowledge gaps regarding the influence of various cell types in the PVN on GSCs behavior. This study developed an organotypic triculture microfluidic model as a means to recapitulate the PVN and study its impact on GSCs. This triculture platform, comprised of endothelial cells (ECs), astrocytes, and GSCs, is used to investigate GSC invasion, proliferation and stemness. Both ECs and astrocytes significantly increased invasiveness of GSCs. This study futher identified 15 ligand-receptor pairs using single-cell RNAseq with putative chemotactic mechanisms of GSCs, where the receptor is up-regulated in GSCs and the diffusible ligand is expressed in either astrocytes or ECs. Notably, the ligand-receptor pair SAA1-FPR1 is demonstrated to be involved in chemotactic invasion of GSCs toward PVN. The novel triculture platform presented herein can be used for therapeutic development and discovery of molecular mechanisms driving GSC biology.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioma/irrigação sanguínea , Glioma/metabolismo , Glioma/patologia , Humanos , Ligantes , Microfluídica , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Microambiente Tumoral
3.
Sci Rep ; 12(1): 2121, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35136119

RESUMO

The aim of the study was to evaluate the role of pseudocontinuous arterial spin labeling perfusion (pCASL-perfusion) in preoperative assessment of cerebral glioma grades. The study group consisted of 253 patients, aged 7-78 years with supratentorial gliomas (65 low-grade gliomas (LGG), 188 high-grade gliomas (HGG)). We used 3D pCASL-perfusion for each patient in order to calculate the tumor blood flow (TBF). We obtained maximal tumor blood flow (maxTBF) in small regions of interest (30 ± 10 mm2) and then normalized absolute maximum tumor blood flow (nTBF) to that of the contralateral normal-appearing white matter of the centrum semiovale. MaxTBF and nTBF values significantly differed between HGG and LGG groups (p < 0.001), as well as between patient groups separated by the grades (grade II vs. grade III) (p < 0.001). Moreover, we performed ROC-analysis which demonstrated high sensitivity and specificity in differentiating between HGG and LGG. We found significant differences for maxTBF and nTBF between grade III and IV gliomas, however, ROC-analysis showed low sensitivity and specificity. We did not observe a significant difference in TBF for astrocytomas and oligodendrogliomas. Our study demonstrates that 3D pCASL-perfusion as an effective diagnostic tool for preoperative differentiation of glioma grades.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Glioma/diagnóstico por imagem , Angiografia por Ressonância Magnética/estatística & dados numéricos , Imagem de Perfusão/estatística & dados numéricos , Adolescente , Adulto , Idoso , Neoplasias Encefálicas/irrigação sanguínea , Criança , Feminino , Glioma/irrigação sanguínea , Humanos , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Estudos Retrospectivos , Adulto Jovem
4.
Angiogenesis ; 25(3): 355-371, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35112158

RESUMO

Glioblastoma stem cells (GSCs) reside close to blood vessels (BVs) but vascular cues contributing to GSC stemness and the nature of GSC-BVs cross talk are not fully understood. Here, we dissected vascular cues influencing GSC gene expression and function to perfusion-based vascular cues, as well as to those requiring direct GSC-endothelial cell (EC) contacts. In light of our previous finding that perivascular tumor cells are metabolically different from tumor cells residing further downstream, cancer cells residing within a narrow, < 60 µm wide perivascular niche were isolated and confirmed to possess a superior tumor-initiation potential compared with those residing further downstream. To circumvent reliance on marker expression, perivascular GSCs were isolated from the respective locales based on their relative state of quiescence. Combined use of these procedures uncovered a large number of previously unrecognized differentially expressed GSC genes. We show that the unique metabolic milieu of the perivascular niche dominated by the highly restricted zone of mTOR activity is conducive for acquisition of GSC properties, primarily in the regulation of genes implicated in cell cycle control. A complementary role of vascular cues including those requiring direct glioma/EC contacts was revealed using glioma/EC co-cultures. Outstanding in the group of glioma cells impacted by nearby ECs were multiple genes responsible for maintaining GSCs in an undifferentiated state, a large fraction of which also relied on Notch-mediated signaling. Glioma-EC communication was found to be bidirectional, evidenced by extensive Notch-mediated EC reprogramming by contacting tumor cells, primarily metabolic EC reprogramming.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Sinais (Psicologia) , Glioblastoma/patologia , Glioma/irrigação sanguínea , Glioma/genética , Humanos , Células-Tronco Neoplásicas/patologia
5.
Sci Rep ; 12(1): 652, 2022 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-35027580

RESUMO

Glutamate carboxypeptidase II (GCP), also known as prostate specific membrane antigen (PSMA) has been found to be expressed in glioma vasculature in in-vitro studies. GCP expression can be traced with the use of [68Ga]Ga-PSMA-11 PET/CT used routinely for prostate cancer imaging. The aim of this paper was to analyze GCP expression in the recurrent glial tumors in vivo. 34 patients (pts.) aged 44.5 ± 10.3 years with suspicion of recurrence of histologically confirmed glioma grade III (6 pts.) and grade IV (28 pts.) were included in the study. All patients underwent contrast-enhanced MR and [68Ga]Ga-PSMA-11 PET/CT. No radiopharmaceutical-related adverse events were noted. PET/CT was positive in all the areas suspected for recurrence at MR in all the patients. The recurrence was confirmed by histopathological examinations or follow-up imaging in all cases. The images showed a very low background activity of the normal brain. Median maximal standard uptake value (SUVmax) of the tumors was 6.5 (range 0.9-15.6) and mean standard uptake value (SUVmean) was 3.5 (range 0.9-7.5). Target-to-background (TBR) ratios varied between 15 and 1400 with a median of 152. Target-to-liver background ratios (TLR) ranged from 0.2 to 2.6, the median TLR was 1.3. No significant difference of the measured parameters was found between the subgroups according to the glioma grade. High GCP expression in the recurrent glioma was demonstrated in-vivo with the use of [68Ga]Ga-PSMA-11 PET/CT. As the treatment options in recurrent glioma are limited, this observation may open new therapeutic perspectives with the use of radiolabeled agents targeting the GCP.


Assuntos
Expressão Gênica , Glioma/diagnóstico por imagem , Glioma/metabolismo , Glutamato Carboxipeptidase II/genética , Glutamato Carboxipeptidase II/metabolismo , Recidiva Local de Neoplasia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Adulto , Feminino , Glioma/irrigação sanguínea , Glioma/patologia , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade
6.
MAGMA ; 35(1): 17-27, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34910266

RESUMO

OBJECTIVE: To evaluate the feasibility of intravoxel incoherent motion (IVIM) in assessing blood-brain barrier (BBB) integrity and microvasculature in tumoral tissue of glioma patients. METHODS: Images from 8 high-grade and 4 low-grade glioma patients were acquired on a 3 T MRI scanner. Acquisition protocol included pre- and post-contrast T1- and T2-weighted imaging, FLAIR, dynamic susceptibility contrast (DSC), and susceptibility-weighted imaging (SWI). In addition, IVIM was acquired with 15 b-values and fitted under the non-negative least square (NNLS) model to output the diffusion (D) and pseudo-diffusion (D*) coefficients, perfusion fraction (f), and f times D* (fD*) maps. RESULTS: IVIM perfusion-related maps were sensitive to (1) blood flow and perfusion alterations within the microvasculature of brain tumors, in agreement with intra-tumoral susceptibility signal (ITSS); (2) enhancing areas of BBB breakdown in agreement with DSC maps as well as areas of BBB abnormality that was not detected on DSC maps; (3) enhancing perfusion changes within edemas; (4) detecting early foci of increased perfusion within low-grade gliomas. CONCLUSION: The results suggest IVIM may be a promising approach to delineate tumor extension and progression in size, and to predict histological grade, which are clinically relevant information that characterize tumors and guide therapeutic decisions in patients with glioma.


Assuntos
Barreira Hematoencefálica , Glioma , Microvasos , Barreira Hematoencefálica/diagnóstico por imagem , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Imagem de Difusão por Ressonância Magnética/métodos , Estudos de Viabilidade , Glioma/irrigação sanguínea , Glioma/diagnóstico por imagem , Glioma/patologia , Glioma/fisiopatologia , Humanos , Microcirculação , Microvasos/diagnóstico por imagem , Microvasos/patologia , Movimento (Física)
7.
Cells ; 10(12)2021 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-34944101

RESUMO

Chondroitin sulfate (CS) is a major component of the extracellular matrix found to be abnormally accumulated in several types of cancer tissues. Previous studies have indicated that CS synthases and modification enzymes are frequently elevated in human gliomas and are associated with poor prognosis. However, the underlying mechanisms of CS in cancer progression and approaches for interrupting its functions in cancer cells remain largely unexplored. Here, we have found that CS was significantly enriched surrounding the vasculature in a subset of glioma tissues, which was akin to the perivascular niche for cancer-initiating cells. Silencing or overexpression of the major CS synthase, chondroitin sulfate synthase 1 (CHSY1), significantly regulated the glioma cell invasive phenotypes and modulated integrin expression. Furthermore, we identified CD44 as a crucial chondroitin sulfate proteoglycan (CSPG) that was modified by CHSY1 on glioma cells, and the suppression of CS formation on CD44 by silencing the CHSY1-inhibited interaction between CD44 and integrin ß1 on the adhesion complex. Moreover, we tested the CS-specific binding peptide, resulting in the suppression of glioma cell mobility in a fashion similar to that observed upon the silencing of CHSY1. In addition, the peptide demonstrated significant affinity to CD44, promoted CD44 degradation, and suppressed integrin ß1 expression in glioma cells. Overall, this study proposes a potential regulatory loop between CS, CD44, and integrin ß1 in glioma cells, and highlights the importance of CS in CD44 stability. Furthermore, the targeting of CS by specific binding peptides has potential as a novel therapeutic strategy for glioma.


Assuntos
Sulfatos de Condroitina/metabolismo , Glioma/metabolismo , Glioma/patologia , Receptores de Hialuronatos/metabolismo , Integrina beta1/metabolismo , Animais , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Glioma/irrigação sanguínea , Glioma/genética , Glucuronosiltransferase/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Enzimas Multifuncionais/metabolismo , N-Acetilgalactosaminiltransferases/metabolismo , Gradação de Tumores , Invasividade Neoplásica , Peptídeos/metabolismo , Fenótipo , Proteólise
8.
Cell Rep ; 36(5): 109480, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34348160

RESUMO

Recent multi-omics studies show different immune tumor microenvironment (TME) compositions in glioblastoma (GBM). However, temporal comprehensive knowledge of the TME from initiation of the disease remains sparse. We use Cre recombinase (Cre)-inducible lentiviral murine GBM models to compare the cellular evolution of the immune TME in tumors initiated from different oncogenic drivers. We show that neutrophils infiltrate early during tumor progression primarily in the mesenchymal GBM model. Depleting neutrophils in vivo at the onset of disease accelerates tumor growth and reduces the median overall survival time of mice. We show that, as a tumor progresses, bone marrow-derived neutrophils are skewed toward a phenotype associated with pro-tumorigenic processes. Our findings suggest that GBM can remotely regulate systemic myeloid differentiation in the bone marrow to generate neutrophils pre-committed to a tumor-supportive phenotype. This work reveals plasticity in the systemic immune host microenvironment, suggesting an additional point of intervention in GBM treatment.


Assuntos
Medula Óssea/patologia , Neoplasias Encefálicas/patologia , Carcinogênese/patologia , Glioma/patologia , Neutrófilos/patologia , Microambiente Tumoral , Animais , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Modelos Animais de Doenças , Progressão da Doença , Feminino , Glioma/irrigação sanguínea , Glioma/tratamento farmacológico , Humanos , Terapia de Imunossupressão , Integrases/metabolismo , Camundongos Endogâmicos C57BL , Mutação/genética , Estadiamento de Neoplasias , Neovascularização Patológica/patologia , Análise de Sobrevida
9.
Int J Mol Sci ; 22(11)2021 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-34200145

RESUMO

Angiogenesis, a complex, multistep process of forming new blood vessels, plays crucial role in normal development, embryogenesis, and wound healing. Malignant tumors characterized by increased proliferation also require new vasculature to provide an adequate supply of oxygen and nutrients for developing tumor. Gliomas are among the most frequent primary tumors of the central nervous system (CNS), characterized by increased new vessel formation. The processes of neoangiogenesis, necessary for glioma development, are mediated by numerous growth factors, cytokines, chemokines and other proteins. In contrast to other solid tumors, some biological conditions, such as the blood-brain barrier and the unique interplay between immune microenvironment and tumor, represent significant challenges in glioma therapy. Therefore, the objective of the study was to present the role of various proangiogenic factors in glioma angiogenesis as well as the differences between normal and tumoral angiogenesis. Another goal was to present novel therapeutic options in oncology approaches. We performed a thorough search via the PubMed database. In this paper we describe various proangiogenic factors in glioma vasculature development. The presented paper also reviews various antiangiogenic factors necessary in maintaining equilibrium between pro- and antiangiogenic processes. Furthermore, we present some novel possibilities of antiangiogenic therapy in this type of tumors.


Assuntos
Indutores da Angiogênese/farmacologia , Inibidores da Angiogênese/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Animais , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Glioma/irrigação sanguínea , Glioma/patologia , Humanos , Neovascularização Patológica/patologia
10.
Cell Death Dis ; 12(6): 615, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34131109

RESUMO

Glioma is one of the most lethal cancers with highly vascularized networks and growing evidences have identified glioma stem cells (GSCs) to account for excessive angiogenesis in glioma. Aberrant expression of paired-related homeobox1 (Prrx1) has been functionally associated with cancer stem cells including GSCs. In this study, Prrx1 was found to be markedly upregulated in glioma specimens and elevated Prrx1 expression was inversely correlated with prognosis of glioma patients. Prrx1 potentiated stemness acquisition in non-stem tumor cells (NSTCs) and stemness maintenance in GSCs, accompanied with increased expression of stemness markers such as SOX2. Prrx1 also promoted glioma angiogenesis by upregulating proangiogenic factors such as VEGF. Consistently, silencing Prrx1 markedly inhibited glioma proliferation, stemness, and angiogenesis in vivo. Using a combination of subcellular proteomics and in vitro analyses, we revealed that Prrx1 directly bound to the promoter regions of TGF-ß1 gene, upregulated TGF-ß1 expression, and ultimately activated the TGF-ß/smad pathway. Silencing TGF-ß1 mitigated the malignant behaviors induced by Prrx1. Activation of this pathway cooperates with Prrx1 to upregulate the expression of stemness-related genes and proangiogenic factors. In summary, our findings revealed that Prrx1/TGF-ß/smad signal axis exerted a critical role in glioma stemness and angiogeneis. Disrupting the function of this signal axis might represent a new therapeutic strategy in glioma patients.


Assuntos
Neoplasias Encefálicas , Glioma , Proteínas de Homeodomínio/fisiologia , Células-Tronco Neoplásicas/fisiologia , Neovascularização Patológica/genética , Animais , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Células Cultivadas , Embrião de Galinha , Regulação Neoplásica da Expressão Gênica , Glioma/irrigação sanguínea , Glioma/genética , Glioma/patologia , Células HEK293 , Proteínas de Homeodomínio/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Neovascularização Patológica/metabolismo , Transdução de Sinais/genética , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima/genética
11.
Br J Radiol ; 94(1125): 20201450, 2021 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-34106749

RESUMO

OBJECTIVE: Blood flow is the rate of blood movement and relevant to numerous processes, though understudied in gliomas. The aim of this review was to pool blood flow metrics obtained from MRI modalities in adult supratentorial gliomas. METHODS: MEDLINE, EMBASE and the Cochrane database were queried 01/01/2000-31/12/2019. Studies measuring blood flow in adult Grade II-IV supratentorial gliomas using dynamic susceptibility contrast (DSC) MRI, dynamic contrast enhanced MRI (DCE-MRI) or arterial spin labelling (ASL) were included. Absolute and relative cerebral blood flow (CBF), peritumoral blood flow and tumoral blood flow (TBF) were reported. RESULTS: 34 studies were included with 1415 patients and 1460 scans. The mean age was 52.4 ± 7.3 years. Most patients had glioblastoma (n = 880, 64.6%). The most common imaging modality was ASL (n = 765, 52.4%) followed by DSC (n = 538, 36.8%). Most studies were performed pre-operatively (n = 1268, 86.8%). With increasing glioma grade (II vs IV), TBF increased (70.8 vs 145.5 ml/100 g/min, p < 0.001) and CBF decreased (85.3 vs 49.6 ml/100 g/min, p < 0.001). In Grade IV gliomas, following treatment, CBF increased in ipsilateral (24.9 ± 1.2 vs 26.1 ± 0.0 ml/100 g/min, p < 0.001) and contralateral white matter (25.6 ± 0.2 vs 26.0± 0.0 ml/100 g/min, p < 0.001). CONCLUSION: Our findings demonstrate that increased mass effect from high-grade gliomas impairs blood flow within the surrounding brain that can improve with surgery. ADVANCES IN KNOWLEDGE: This systematic review demonstrates how mass effect from brain tumours impairs blood flow in the surrounding brain parenchyma that can improve with treatment.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/diagnóstico por imagem , Circulação Cerebrovascular , Glioma/irrigação sanguínea , Glioma/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Adulto , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Glioma/patologia , Humanos , Gradação de Tumores
12.
NMR Biomed ; 34(7): e4516, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33817893

RESUMO

The effect of a human vascular endothelial growth factor antibody on the vasculature of human tumor grown in rat brain was studied. Using dynamic contrast-enhanced magnetic resonance imaging, the effects of intravenous bevacizumab (Avastin; 10 mg/kg) were examined before and at postadministration times of 1, 2, 4, 8, 12 and 24 h (N = 26; 4-5 per time point) in a rat model of orthotopic, U251 glioblastoma (GBM). The commonly estimated vascular parameters for an MR contrast agent were: (i) plasma distribution volume (vp ), (ii) forward volumetric transfer constant (Ktrans ) and (iii) reverse transfer constant (kep ). In addition, extracellular distribution volume (VD ) was estimated in the tumor (VD-tumor ), tumor edge (VD-edge ) and the mostly normal tumor periphery (VD-peri ), along with tumor blood flow (TBF), peri-tumoral hydraulic conductivity (K) and interstitial flow (Flux) and tumor interstitial fluid pressure (TIFP). Studied as % changes from baseline, the 2-h post-treatment time point began showing significant decreases in vp , VD-tumor, VD-edge and VD-peri , as well as K, with these changes persisting at 4 and 8 h in vp , K, VD-tumor, -edge and -peri (t-tests; p < 0.05-0.01). Decreases in Ktrans were observed at the 2- and 4-h time points (p < 0.05), while interstitial volume fraction (ve ; = Ktrans /kep ) showed a significant decrease only at the 2-h time point (p < 0.05). Sustained decreases in Flux were observed from 2 to 24 h (p < 0.01) while TBF and TIFP showed delayed responses, increases in the former at 12 and 24 h and a decrease in the latter only at 12 h. These imaging biomarkers of tumor vascular kinetics describe the short-term temporal changes in physical spaces and fluid flows in a model of GBM after Avastin administration.


Assuntos
Bevacizumab/uso terapêutico , Glioma/irrigação sanguínea , Glioma/tratamento farmacológico , Animais , Bevacizumab/farmacologia , Linhagem Celular Tumoral , Feminino , Glioma/diagnóstico por imagem , Humanos , Cinética , Imageamento por Ressonância Magnética , Modelos Biológicos , Ratos , Distribuição Tecidual
13.
Br J Radiol ; 94(1121): 20201321, 2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-33876653

RESUMO

OBJECTIVE: This meta-analysis was carried out for assessing the accuracy of intravoxel incoherent motion (IVIM) parameters true diffusion coefficient (D), pseudo-diffusion coefficient (D*), and perfusion fraction (f) in differentiating low-grade gliomas (LGGs) from high-grade gliomas (HGGs). METHODS: Literatures concerning IVIM in the grading of brain gliomas published prior to October 20, 2020, searched in the Embase, PubMed, and Cochrane library. Use the quality assessment of diagnostic accuracy studies 2 (QUADAS 2) to evaluate the quality of studies. We estimated the pooled sensitivity, specificity, and the area under the summary ROC (SROC) curve to identification the accuracy of IVIM parameters D, D*, and f evaluation in grading gliomas. RESULTS: Totally, 6 articles including 252 brain gliomas conform to the inclusion criteria. The pooled sensitivity of parameters D, D*, and f derived from IVIM were 0.85 (95%Cl, 0.76-0.91), 0.78 (95%Cl, 0.71-0.85), and 0.89 (95%Cl, 0.76-0.96), respectively. The pooled specificity were 0.78 (95%Cl, 0.60-0.90), 0.68 (95%Cl, 0.56-0.79), and 0.88 (95%Cl, 0.76-0.94), respectively. Meanwhile, the AUC of SROC curve were 0.89 (95%Cl, 0.86-0.92) , 0.81 (95%Cl, 0.77-0.84), and 0.94 (95%Cl, 0.92-0.96), respectively. CONCLUSION: This meta-analysis suggested that IVIM parameters D, D*, and f have moderate or high diagnosis value accuracy in differentiating HGGs from LGGs, and the parameter f has greater sensitivity and specificity. Standardized methodology is warranted to guide the use of this method for clinical decision-making. However, more clinical studies are needed to prove our view. ADVANCES IN KNOWLEDGE: IVIM parameter f showed greater sensitivity and specificity, as well as excellent performance than parameter D* and D.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Glioma/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Microcirculação , Área Sob a Curva , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Intervalos de Confiança , Imagem de Difusão por Ressonância Magnética , Glioma/irrigação sanguínea , Glioma/patologia , Humanos , Gradação de Tumores , Razão de Chances , Viés de Publicação , Sensibilidade e Especificidade
14.
Cancer Res ; 81(8): 2142-2156, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33593822

RESUMO

The extraordinary plasticity of glioma cells allows them to contribute to different cellular compartments in tumor vessels, reinforcing the vascular architecture. It was recently revealed that targeting glioma-derived pericytes, which represent a big percentage of the mural cell population in aggressive tumors, increases the permeability of the vessels and improves the efficiency of chemotherapy. However, the molecular determinants of this transdifferentiation process have not been elucidated. Here we show that mutations in EGFR stimulate the capacity of glioma cells to function as pericytes in a BMX- (bone marrow and X-linked) and SOX9-dependent manner. Subsequent activation of platelet-derived growth factor receptor beta in the vessel walls of EGFR-mutant gliomas stabilized the vasculature and facilitated the recruitment of immune cells. These changes in the tumor microenvironment conferred a growth advantage to the tumors but also rendered them sensitive to pericyte-targeting molecules such as ibrutinib or sunitinib. In the absence of EGFR mutations, high-grade gliomas were enriched in blood vessels, but showed a highly disrupted blood-brain barrier due to the decreased BMX/SOX9 activation and pericyte coverage, which led to poor oxygenation, necrosis, and hypoxia. Overall, these findings identify EGFR mutations as key regulators of the glioma-to-pericyte transdifferentiation, highlighting the intricate relationship between the tumor cells and their vascular and immune milieu. Our results lay the foundations for a vascular-dependent stratification of gliomas and suggest different therapeutic vulnerabilities determined by the genetic status of EGFR. SIGNIFICANCE: This study identifies the EGFR-related mechanisms that govern the capacity of glioma cells to transdifferentiate into pericytes, regulating the vascular and immune phenotypes of the tumors. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2142/F1.large.jpg.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Transdiferenciação Celular , Microambiente Celular , Glioma/irrigação sanguínea , Mutação , Pericitos/fisiologia , Adenina/análogos & derivados , Adenina/farmacologia , Inibidores da Angiogênese/farmacologia , Animais , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Barreira Hematoencefálica/metabolismo , Medula Óssea , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Cromossomos Humanos X , Receptores ErbB/genética , Glioma/imunologia , Glioma/patologia , Humanos , Imunidade Celular , Isocitrato Desidrogenase/genética , Camundongos , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Piperidinas/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fatores de Transcrição SOX9 , Sunitinibe/farmacologia , Hipóxia Tumoral , Microambiente Tumoral
15.
Aging (Albany NY) ; 13(4): 5055-5068, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33535172

RESUMO

Vasculogenic mimicry (VM), the formation of an alternative microvascular circulation independent of VEGF-driven angiogenesis, is reluctant to anti-angiogenesis therapy for glioma patients. However, treatments targeting VM are lacking due to the poor understanding of the molecular mechanism involved in VM formation. By analysing the TCGA database, microRNA-29a-3p (miR-29a-3p) was found to be highly expressed in normal brain tissue compared with glioma. An in vitro study revealed an inhibitory role for miR-29a-3p in glioma cell migration and VM formation, and further study confirmed that ROBO1 is a direct target of miR-29a-3p. Based on this, we engineered human mesenchymal stem cells (MSCs) to produce miR-29a-3p-overexpressing exosomes. Treatment with these exosomes attenuated migration and VM formation in glioma cells. Moreover, the anti-glioma role of miR-29a-3p and miR-29a-3p-overexpressing exosomes were confirmed in vivo. Overall, the present study demonstrates that MSCs can be used to produce miR-29a-3p-overexpressing exosomes, which have great potential for anti-VM therapy and may act as supplements to anti-angiogenetic therapy in the clinic.


Assuntos
Exossomos/metabolismo , Glioma/genética , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , Neovascularização Patológica/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Técnicas de Silenciamento de Genes , Glioma/irrigação sanguínea , Glioma/metabolismo , Humanos , Camundongos , Camundongos Nus , MicroRNAs/metabolismo , Transplante de Neoplasias , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/genética
16.
Methods ; 185: 94-104, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-31981608

RESUMO

This paper develops a three-dimensional in silico hybrid model of cancer, which describes the multi-variate phenotypic behaviour of tumour and host cells. The model encompasses the role of cell migration and adhesion, the influence of the extracellular matrix, the effects of oxygen and nutrient availability, and the signalling triggered by chemical cues and growth factors. The proposed in silico hybrid modelling framework combines successfully the advantages of continuum-based and discrete methods, namely the finite element and agent-based method respectively. The framework is thus used to realistically model cancer mechano-biology in a multiscale fashion while maintaining the resolution power of each method in a computationally cost-effective manner. The model is tailored to simulate glioma progression, and is subsequently used to interrogate the balance between the host cells and small sized gliomas, while the go-or-grow phenotype characteristic in glioblastomas is also investigated. Also, cell-cell and cell-matrix interactions are examined with respect to their effect in (macroscopic) tumour growth, brain tissue perfusion and tumour necrosis. Finally, we use the in silico framework to assess differences between low-grade and high-grade glioma growth, demonstrating significant differences in the distribution of cancer as well as host cells, in accordance with reported experimental findings.


Assuntos
Simulação por Computador , Glioma/patologia , Modelos Biológicos , Neovascularização Patológica , Progressão da Doença , Glioma/irrigação sanguínea , Humanos , Necrose , Invasividade Neoplásica
17.
Cell Prolif ; 54(2): e12929, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33300633

RESUMO

Glioma is the most commonly observed primary intracranial tumour and is associated with massive angiogenesis. Glioma neovascularization provides nutrients for the growth and metabolism of tumour tissues, promotes tumour cell division and proliferation, and provides conditions ideal for the infiltration and migration of tumour cells to distant places. Growing evidence suggests that there is a correlation between the activation of nuclear factor (NF)-κB and the angiogenesis of glioma. In this review article, we highlighted the functions of NF-κB in the angiogenesis of glioma, showing that NF-κB activation plays a pivotal role in the growth and progression of glioma angiogenesis and is a rational therapeutic target for antiangiogenic strategies aimed at glioma.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , NF-kappa B/metabolismo , Antineoplásicos Imunológicos/uso terapêutico , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Caspases/metabolismo , Citocinas/metabolismo , Glioma/irrigação sanguínea , Glioma/metabolismo , Glioma/terapia , Humanos , MicroRNAs/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , Neovascularização Patológica , Estresse Oxidativo
18.
J Biomed Mater Res A ; 109(6): 915-925, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32779363

RESUMO

Glioblastoma is the most frequently diagnosed primary malignant brain tumor with unfavourable prognosis and high mortality. One of its key features is the extensive abnormal vascular network. Up to now, the mechanism of angiogenesis and the origin of tumor vascularization remain controversial. It is essential to establish an ideal preclinical tumor model to elucidate the mechanism of tumor vascularization, and the role of tumor cells in this process. In this study, both U118 cell and GSC23 cell exhibited good printability and cell proliferation. Compared with 3D-U118, 3D-GSC23 had a greater ability to form cell spheroids, to secrete vascular endothelial growth factor (VEGFA), and to form tubule-like structures in vitro. More importantly, 3D-glioma stem cells (GSC)23 cells had a greater power to transdifferentiate into functional endothelial cells, and blood vessels composed of tumor cells with an abnormal endothelial phenotype was observed in vivo. In summary, 3D bioprinted hydrogel scaffold provided a suitable tumor microenvironment (TME) for glioma cells and GSCs. This bioprinted model supported a novel TME for the research of glioma cells, especially GSCs in glioma vascularization and therapeutic targeting of tumor angiogenesis.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Glioma/irrigação sanguínea , Neovascularização Patológica/patologia , Impressão Tridimensional , Microambiente Tumoral , Diferenciação Celular , Linhagem Celular Tumoral , Células Endoteliais , Humanos , Hidrogéis , Microtúbulos/química , Modelos Anatômicos , Fluxo Sanguíneo Regional , Tecidos Suporte , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Aging (Albany NY) ; 12(24): 24995-25004, 2020 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-33229627

RESUMO

BACKGROUND: Malignant glioma is the most common form of primary malignant brain cancer. Heterogeneity is the hallmark of glioma. DAZ-interacting zinc finger 3 (DZIP3), acts as an RNA-binding RING-type ubiquitin ligase; however, its function in glioma is yet unclear. RESULTS: The DZIP3 expression was related to the World Health Organization (WHO) grade and isocitrate dehydrogenase 1(IDH1) status, as well as the clinical outcome. Malignant cases exhibit lower DZIP3 expression. DZIP3 was an independent predictive factor of good prognosis in all grade and lower grade gliomas (p < 0.0001). Gene enrichment analysis and immunohistochemistry indicated that DZIP3 affected the biological behavior of glioma through the angiogenesis pathway. Moreover, based on DZIP3 expression, IDH1 wild-type lower-grade gliomas could be divided into two groups with different survival time. CONCLUSION: In conclusion, the loss of DZIP3 may be involved in the mechanism of angiogenesis in the invasive biological process of glioma. These findings laid an understanding of DZIP3-specific clinical features in glioma. METHODS: A total of 325 glioma patients from the Chinese Glioma Genome Atlas (CGGA) RNA-seq cohort comprised the training cohort, while 265 patients from the GSE 16011 array cohort formed the validation cohort. The mRNA expression of DZIP3 and clinical characteristics was assessed. DZIP3 protein expression and microvessel density (MVD) were evaluated by immunohistochemistry (IHC).


Assuntos
Neoplasias Encefálicas/genética , Glioma/genética , Isocitrato Desidrogenase/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Ubiquitina-Proteína Ligases/genética , Astrocitoma/irrigação sanguínea , Astrocitoma/genética , Astrocitoma/patologia , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Glioblastoma/irrigação sanguínea , Glioblastoma/genética , Glioblastoma/patologia , Glioma/irrigação sanguínea , Glioma/patologia , Humanos , Imuno-Histoquímica , Gradação de Tumores , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Oligodendroglioma/irrigação sanguínea , Oligodendroglioma/genética , Oligodendroglioma/patologia , Prognóstico , RNA-Seq , Taxa de Sobrevida
20.
Sci Rep ; 10(1): 17324, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-33057180

RESUMO

Many neurological diseases present with substantial genetic and phenotypic heterogeneity, making assessment of these diseases challenging. This has led to ineffective treatments, significant morbidity, and high mortality rates for patients with neurological diseases, including brain cancers and neurodegenerative disorders. Improved understanding of this heterogeneity is necessary if more effective treatments are to be developed. We describe a new method to measure phenotypic heterogeneity across the whole rodent brain at multiple spatial scales. The method involves co-registration and localized comparison of in vivo radiologic images (e.g. MRI, PET) with ex vivo optical reporter images (e.g. labeled cells, molecular targets, microvasculature) of optically cleared tissue slices. Ex vivo fluorescent images of optically cleared pathology slices are acquired with a preclinical in vivo optical imaging system across the entire rodent brain in under five minutes, making this methodology practical and feasible for most preclinical imaging labs. The methodology is applied in various examples demonstrating how it might be used to cross-validate and compare in vivo radiologic imaging with ex vivo optical imaging techniques for assessing hypoxia, microvasculature, and tumor growth.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Glioma/diagnóstico por imagem , Gliossarcoma/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Imagem Multimodal/métodos , Neuroimagem/métodos , Imagem Óptica/métodos , Tomografia por Emissão de Pósitrons/métodos , Animais , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/química , Hipóxia Celular , Linhagem Celular Tumoral , Corantes Fluorescentes/análise , Genes Reporter , Glioma/irrigação sanguínea , Glioma/química , Gliossarcoma/irrigação sanguínea , Gliossarcoma/química , Processamento de Imagem Assistida por Computador , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Camundongos , Camundongos Nus , Microtomia , Microvasos/diagnóstico por imagem , Fenótipo , Ratos , Ratos Endogâmicos F344 , Ratos Wistar , Carga Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...