Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Neuropathol Appl Neurobiol ; 45(2): 119-140, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29679380

RESUMEN

AIMS: Resident and peripherally derived glioma associated microglia/macrophages (GAMM) play a key role in driving tumour progression, angiogenesis, invasion and attenuating host immune responses. Differentiating these cells' origins is challenging and current preclinical models such as irradiation-based adoptive transfer, parabiosis and transgenic mice have limitations. We aimed to develop a novel nonmyeloablative transplantation (NMT) mouse model that permits high levels of peripheral chimerism without blood-brain barrier (BBB) damage or brain infiltration prior to tumour implantation. METHODS: NMT dosing was determined in C57BL/6J or Pep3/CD45.1 mice conditioned with concentrations of busulfan ranging from 25 mg/kg to 125 mg/kg. Donor haematopoietic cells labelled with eGFP or CD45.2 were injected via tail vein. Donor chimerism was measured in peripheral blood, bone marrow and spleen using flow cytometry. BBB integrity was assessed with anti-IgG and anti-fibrinogen antibodies. Immunocompetent chimerised animals were orthotopically implanted with murine glioma GL-261 cells. Central and peripheral cell contributions were assessed using immunohistochemistry and flow cytometry. GAMM subpopulation analysis of peripheral cells was performed using Ly6C/MHCII/MerTK/CD64. RESULTS: NMT achieves >80% haematopoietic chimerism by 12 weeks without BBB damage and normal life span. Bone marrow derived cells (BMDC) and peripheral macrophages accounted for approximately 45% of the GAMM population in GL-261 implanted tumours. Existing markers such as CD45 high/low proved inaccurate to determine central and peripheral populations while Ly6C/MHCII/MerTK/CD64 reliably differentiated GAMM subpopulations in chimerised and unchimerised mice. CONCLUSION: NMT is a powerful method for dissecting tumour microglia and macrophage subpopulations and can guide further investigation of BMDC subsets in glioma and neuro-inflammatory diseases.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Macrófagos/patología , Microglía/patología , Animales , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos
2.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 36(3): 515-520, 2019 Jun 25.
Artículo en Zh | MEDLINE | ID: mdl-31232558

RESUMEN

Glioma is one of the most common primary tumors in the human brain with poor prognosis. The local and systemic immunosuppressive environment created by glioma cells enables them to evade immunosurveillance. Myeloid-derived suppressor cells (MDSCs) are a critical component of the immunosuppression system. They are a heterogeneous cell population composed of early myeloid progenitor cells and precursor cells. Although the cells are diverse in phenotypes and functions, they all have strong immunosuppressive functions. MDSCs are extensively infiltrated into tumor tissues and play an important role in the glioma immunosuppressive microenvironment, which also hinders the immunotherapeutic effects of glioma. This article will review the phenotypic characteristics of MDSCs in the glioma microenvironment and their role in the progression of glioma. It is of positive significance to better understand the pathogenesis of glioma and explore effective comprehensive treatments.


Asunto(s)
Glioma/patología , Células Supresoras de Origen Mieloide/citología , Microambiente Tumoral , Humanos , Tolerancia Inmunológica
3.
J Adv Res ; 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39097088

RESUMEN

BACKGROUND: With the gradual understanding of glioma development and the immune microenvironment, many immune cells have been discovered. Despite the growing comprehension of immune cell functions and the clinical application of immunotherapy, the precise roles and characteristics of immune cell subtypes, how glioma induces subtype transformation of immune cells and its impact on glioma progression have yet to be understood. AIM OF THE REVIEW: In this review, we comprehensively center on the four major immune cells within the glioma microenvironment, particularly neutrophils, macrophages, lymphocytes, myeloid-derived suppressor cells (MDSCs), and other significant immune cells. We discuss (1) immune cell subtype markers, (2) glioma-induced immune cell subtype transformation, (3) the mechanisms of each subtype influencing chemotherapy resistance, (4) therapies targeting immune cells, and (5) immune cell-associated single-cell sequencing. Eventually, we identified the characteristics of immune cell subtypes in glioma, comprehensively summarized the exact mechanism of glioma-induced immune cell subtype transformation, and concluded the progress of single-cell sequencing in exploring immune cell subtypes in glioma. KEY SCIENTIFIC CONCEPTS OF REVIEW: In conclusion, we have analyzed the mechanism of chemotherapy resistance detailly, and have discovered prospective immunotherapy targets, excavating the potential of novel immunotherapies approach that synergistically combines radiotherapy, chemotherapy, and surgery, thereby paving the way for improved immunotherapeutic strategies against glioma and enhanced patient outcomes.

4.
Front Immunol ; 14: 1259562, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37781367

RESUMEN

Gliomas account for the majority of brain malignant tumors. As the most malignant subtype of glioma, glioblastoma (GBM) is barely effectively treated by traditional therapies (surgery combined with radiochemotherapy), resulting in poor prognosis. Meanwhile, due to its "cold tumor" phenotype, GBM fails to respond to multiple immunotherapies. As its capacity to prime T cell response, dendritic cells (DCs) are essential to anti-tumor immunity. In recent years, as a therapeutic method, dendritic cell vaccine (DCV) has been immensely developed. However, there have long been obstacles that limit the use of DCV yet to be tackled. As is shown in the following review, the role of DCs in anti-tumor immunity and the inhibitory effects of tumor microenvironment (TME) on DCs are described, the previous clinical trials of DCV in the treatment of GBM are summarized, and the challenges and possible development directions of DCV are analyzed.


Asunto(s)
Neoplasias Encefálicas , Vacunas contra el Cáncer , Glioblastoma , Glioma , Humanos , Células Dendríticas , Vacunas contra el Cáncer/uso terapéutico , Microambiente Tumoral
5.
J Control Release ; 358: 681-705, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37196900

RESUMEN

Gliomas are the most aggressive and lethal tumors of the central nervous system, for which few therapeutic options exist. Surgical resection is the primary treatment for most gliomas; however, tumor recurrence is nearly inevitable. Emerging nanobiotechnology-based strategies have shown great prospects for early glioma diagnosis, physiological barrier traversing, postoperative regrowth suppression, and microenvironment remodeling. Herein, we focus on the postoperative scenario and summarize the key properties of the glioma microenvironment, especially its immune peculiarities. We elucidate the challenges of managing recurrent glioma. We also discuss the potential of nanobiotechnology in addressing the therapeutic challenges of recurrent glioma, including optimizing the design of drug delivery systems, enhancing intracranial accumulation, and restoring the anti-glioma immune response. The development of these technologies offers new opportunities for accelerating the drug development process and treating recurrent glioma.


Asunto(s)
Neoplasias Encefálicas , Glioma , Humanos , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Microambiente Tumoral
6.
Neuro Oncol ; 25(2): 263-276, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-35609569

RESUMEN

BACKGROUND: Dendritic cells (DC), the most potent professional antigen presenting cells capable of effective cross-presentation, have been demonstrated to license T helper cells to induce antitumor immunity in solid tumors. Specific DC subtypes are recruited to the injured brain by microglial chemokines, locally adapting to distinct transcriptional profiles. In isocitrate dehydrogenase (IDH) type 1 mutant gliomas, monocyte-derived macrophages have recently been shown to display an attenuated intratumoral antigen presentation capacity as consequence of the local accumulation of the oncometabolite R-2-hydroxyglutarate. The functionality and the contribution of DC to the IDH-mutant tumor microenvironment (TME) remains unclear. METHODS: Frequencies and intratumoral phenotypes of human DC in IDH-wildtype (IDHwt) and -mutant high-grade gliomas are comparatively assessed by transcriptomic and proteomic profiling. DC functionality is investigated in experimental murine glioblastomas expressing the model antigen ovalbumin. Single-cell sequencing-based pseudotime analyses and spectral flow cytometric analyses are used to profile DC states longitudinally. RESULTS: DC are present in primary and recurrent high-grade gliomas and interact with other immune cell types within the TME. In murine glioblastomas, we find an IDH-status-associated major histocompatibility class I-restricted cross-presentation of tumor antigens by DC specifically in the tumor but not in meninges or secondary lymphoid organs of tumor-bearing animals. In single-cell sequencing-based pseudotime and longitudinal spectral flow cytometric analyses, we demonstrate an IDH-status-dependent differential, exclusively microenvironmental education of DC. CONCLUSIONS: Glioma-associated DCs are relevantly abundant in human IDHwt and mutant tumors. Glioma IDH mutations result in specifically educated, dysfunctional DCs via paracrine reprogramming of infiltrating monocytes, providing the basis for combinatorial immunotherapy concepts against IDH mutant gliomas.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Animales , Ratones , Glioblastoma/patología , Proteómica , Linfocitos T/metabolismo , Glioma/patología , Neoplasias Encefálicas/patología , Células Dendríticas , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Mutación , Microambiente Tumoral
7.
CNS Neurosci Ther ; 28(11): 1748-1766, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35855654

RESUMEN

INTRODUCTION: Pentraxin 3 (PTX3) is an essential regulator of the immune system. However, the immune-modulatory role of PTX3 in the tumor microenvironment of glioma has not been elucidated. METHODS: The RNA seq samples were obtained from The Cancer Genome Atlas (TCGA) and the China Glioma Genome Atlas (CGGA) datasets. The single-cell sequencing data of glioblastoma (GBM) samples were obtained from the Single Cell Portal platform (http://singlecell.broadinstitute.org). Immunohistochemistry was used to assess PTX3 expression, HAVCR2, PD-1, PD-L1, and CD276 in glioma sections from the Xiangya cohort (n = 60). Multiplex immunofluorescence staining of PTX3, CD68, and CD163 was performed in several solid cancer types, including GBM. HMC3 was cocultured with U251 and U87, and transwell assay and flow cytometry assay were performed to explore the migration and polarization activity of HMC3. RESULTS: PTX3 expression is significantly increased in GBM. PTX3 expression predicts worse survival in the Xiangya cohort. PTX3 is closely related to the expression of PD-1, PD-L1, CD276, and HAVCR2 in the tumor microenvironment. Additionally, PTX3 is involved in tumorigenic and immunogenic processes, especially the activity of macrophages based on various signaling pathways in cellular communications and critical transcription factors. Specifically, PTX3 actively mediates macrophages' infiltration, migration, and inflammation-resolving-polarization. PTX3 could also predict immunotherapy response. CONCLUSION: PTX3 is critically involved in macrophage infiltration, migration, and inflammation-resolving-polarization and modulates an immunosuppressive microenvironment.


Asunto(s)
Glioblastoma , Glioma , Antígenos B7/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Proteína C-Reactiva , Glioblastoma/metabolismo , Glioma/genética , Humanos , Inflamación/metabolismo , Macrófagos/patología , Receptor de Muerte Celular Programada 1/metabolismo , Componente Amiloide P Sérico , Factores de Transcripción/metabolismo , Microambiente Tumoral
8.
Front Immunol ; 13: 914001, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36159780

RESUMEN

Accumulating evidence has demonstrated that the immune cells have an emerging role in controlling anti-tumor immune responses and tumor progression. The comprehensive role of mast cell in glioma has not been illustrated yet. In this study, 1,991 diffuse glioma samples were collected from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA). xCell algorithm was employed to define the mast cell-related genes. Based on mast cell-related genes, gliomas were divided into two clusters with distinct clinical and immunological characteristics. The survival probability of cluster 1 was significantly lower than that of cluster 2 in the TCGA dataset, three CGGA datasets, and the Xiangya cohort. Meanwhile, the hypoxic and metabolic pathways were active in cluster 1, which were beneficial to the proliferation of tumor cells. A potent prognostic model based on mast cell was constructed. Via machine learning, DRG2 was screened out as a characteristic gene, which was demonstrated to predict treatment response and predict survival outcome in the Xiangya cohort. In conclusion, mast cells could be used as a potential effective prognostic factor for gliomas.


Asunto(s)
Neoplasias Encefálicas , Glioma , Neoplasias Encefálicas/patología , Proteínas de Unión al GTP/genética , Glioma/patología , Humanos , Evasión Inmune , Mastocitos/patología , RNA-Seq
9.
Front Oncol ; 12: 1004700, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36313679

RESUMEN

Glioblastoma (GBM) is the most malignant type of glioma with the worst prognosis. Traditional therapies (surgery combined with radiotherapy and chemotherapy) have limited therapeutic effects. As a novel therapy emerging in recent years, immunotherapy is increasingly used in glioblastoma (GBM), so we expect to discover more effective immune targets. FGL2, a member of the thrombospondin family, plays an essential role in regulating the activity of immune cells and tumor cells in GBM. Elucidating the role of FGL2 in GBM can help improve immunotherapy efficacy and design treatment protocols. This review discusses the immunosuppressive role of FGL2 in the GBM tumor microenvironment and its ability to promote malignant tumor progression while considering FGL2-targeted therapeutic strategies. Also, we summarize the molecular mechanisms of FGL2 expression on various immune cell types and discuss the possibility of FGL2 and its related mechanisms as new GBM immunotherapy.

10.
Cells ; 10(3)2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33802060

RESUMEN

Glioblastoma is characterized by extensive necrotic areas with surrounding hypoxia. The cancer cell response to hypoxia in these areas is well-described; it involves a metabolic shift and an increase in stem cell-like characteristics. Less is known about the hypoxic response of tumor-associated astrocytes, a major component of the glioma tumor microenvironment. Here, we used primary human astrocytes and a genetically engineered glioma mouse model to investigate the response of this stromal cell type to hypoxia. We found that astrocytes became reactive in response to intermediate and severe hypoxia, similarly to irradiated and temozolomide-treated astrocytes. Hypoxic astrocytes displayed a potent hypoxia response that appeared to be driven primarily by hypoxia-inducible factor 2-alpha (HIF-2α). This response involved the activation of classical HIF target genes and the increased production of hypoxia-associated cytokines such as TGF-ß1, IL-3, angiogenin, VEGF-A, and IL-1 alpha. In vivo, astrocytes were present in proximity to perinecrotic areas surrounding HIF-2α expressing cells, suggesting that hypoxic astrocytes contribute to the glioma microenvironment. Extracellular matrix derived from hypoxic astrocytes increased the proliferation and drug efflux capability of glioma cells. Together, our findings suggest that hypoxic astrocytes are implicated in tumor growth and potentially stemness maintenance by remodeling the tumor microenvironment.


Asunto(s)
Astrocitos/metabolismo , Glioma/fisiopatología , Animales , Hipoxia de la Célula , Humanos , Ratones , Microambiente Tumoral
11.
Front Immunol ; 12: 694490, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34594324

RESUMEN

Tumor-infiltrating immune cells (TIICs) have become an important source of markers for predicting the clinical outcomes of cancer patients. However, measurements of cellular heterogeneity vary due to the frequently updated reference genomes and gene annotations. In this study, we systematically collected and evaluated the infiltration pattern of 65 immune cells. We constructed the Immune Cell Pair (ICP) score based on the cell pair algorithm in 3,715 samples and across 12 independent cancer types, among which, the ICP score from six cancer types was further validated in 2,228 GEO samples. An extensive tumorigenic and immunogenomic analysis was subsequently conducted. As a result, the ICP score showed a robust reliability and efficacy in predicting the survival of patients with gliomas, in pan-cancer samples, and six independent cancer types. Notably, the ICP score was correlated with the genomic alteration features in gliomas. Moreover, the ICP score exhibited a remarkable association with multiple immunomodulators that could potentially mediate immune escape. Finally, the ICP score predicted immunotherapeutic responses with a high sensitivity, allowing a useful tool for predicting the overall survival and guiding immunotherapy for cancer patients.


Asunto(s)
Algoritmos , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Perfilación de la Expresión Génica , Glioma/genética , Inmunogenética , Transcriptoma , Microambiente Tumoral , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/terapia , Bases de Datos Genéticas , Glioma/inmunología , Glioma/mortalidad , Glioma/terapia , Humanos , Inmunoterapia , Valor Predictivo de las Pruebas , Pronóstico , Escape del Tumor
12.
Front Immunol ; 12: 656541, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33959130

RESUMEN

Gliomas are primary malignant brain tumors. Monocytes have been proved to actively participate in tumor growth. Weighted gene co-expression network analysis was used to identify meaningful monocyte-related genes for clustering. Neural network and SVM were applied for validating clustering results. Somatic mutation and copy number variation were used for defining the features of identified clusters. Differentially expressed genes (DEGs) between the stratified groups after performing elastic regression and principal component analyses were used for the construction of risk scores. Monocytes were associated with glioma patients' survival and exhibited high predictive value. The prognostic value of risk score in glioma was validated by the abundant expression of immune checkpoint and metabolic profile. Additionally, high risk score was positively associated with the expression of immunogenic and antigen presenting factors, which indicated high immune infiltration. A prognostic model based on risk score demonstrated high accuracy rate of receiver operating characteristic curves. Compared with previous studies, our research dissected functional roles of monocytes from large-scale analysis. Findings of our analyses strongly support an immune modulatory and prognostic role of monocytes in glioma progression. Notably, monocyte could be an effective predictor for therapy responses of glioma patients.


Asunto(s)
Biomarcadores de Tumor , Glioma/inmunología , Glioma/mortalidad , Aprendizaje Automático , Monocitos/inmunología , Microambiente Tumoral/inmunología , Biología Computacional/métodos , Bases de Datos Genéticas , Perfilación de la Expresión Génica , Genómica/métodos , Glioma/metabolismo , Glioma/patología , Humanos , Inmunoterapia/métodos , Recuento de Leucocitos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Anotación de Secuencia Molecular , Monocitos/metabolismo , Monocitos/patología , Pronóstico , Curva ROC , Reproducibilidad de los Resultados , Transcriptoma , Resultado del Tratamiento
13.
Front Immunol ; 12: 627650, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868245

RESUMEN

Purpose: The extent of preoperative peritumoral edema in glioblastoma (GBM) has been negatively correlated with patient outcome. As several ongoing studies are investigating T-cell based immunotherapy in GBM, we conducted this study to assess whether peritumoral edema with potentially increased intracranial pressure, disrupted tissue homeostasis and reduced local blood flow has influence on immune infiltration and affects survival. Methods: A volumetric analysis of preoperative imaging (gadolinium enhanced T1 weighted MRI sequences for tumor size and T2 weighted sequences for extent of edema (including the infiltrative zone, gliosis etc.) was conducted in 144 patients using the Brainlab® software. Immunohistochemical staining was analyzed for lymphocytic- (CD 3+) and myelocytic (CD15+) tumor infiltration. A retrospective analysis of patient-, surgical-, and molecular characteristics was performed using medical records. Results: The edema to tumor ratio was neither associated with progression-free nor overall survival (p=0.90, p=0.74). However, GBM patients displaying IDH-1 wildtype had significantly higher edema to tumor ratio than patients displaying an IDH-1 mutation (p=0.01). Immunohistopathological analysis did not show significant differences in lymphocytic or myelocytic tumor infiltration (p=0.78, p=0.74) between these groups. Conclusion: In our cohort, edema to tumor ratio had no significant correlation with immune infiltration and outcome. However, patients with an IDH-1wildtype GBM had a significantly higher edema to tumor ratio compared to their IDH-1 mutated peer group. Further studies are necessary to elucidate the underlying mechanisms.


Asunto(s)
Biomarcadores de Tumor/genética , Edema Encefálico/genética , Neoplasias Encefálicas/genética , Glioblastoma/genética , Isocitrato Deshidrogenasa/genética , Linfocitos Infiltrantes de Tumor/inmunología , Mutación , Edema Encefálico/diagnóstico por imagen , Edema Encefálico/inmunología , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/cirugía , Femenino , Glioblastoma/diagnóstico por imagen , Glioblastoma/inmunología , Glioblastoma/cirugía , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Supervivencia sin Progresión , Estudios Retrospectivos , Carga Tumoral , Microambiente Tumoral
14.
Front Cell Dev Biol ; 9: 674995, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34381770

RESUMEN

Mesenchymal stem cells (MSCs) are promising tools for cancer therapy, but there is a risk of malignant transformation in their clinical application. Our previous work revealed that the paracrine protein S100B in the glioma microenvironment induces malignant transformation of MSCs and upregulates intracellular S100B, which could affect cell homeostasis by interfering with p53. The purpose of this study was to investigate whether extracellular S100B can be internalized by MSCs and the specific endocytic pathway involved in S100B internalization. By using real-time confocal microscopy and structured illumination microscopy (SIM), we visualized the uptake of fluorescently labeled S100B protein (S100B-Alexa488) and monitored the intracellular trafficking of internalized vesicles. The results showed that S100B-Alexa488 was efficiently internalized into MSCs in a time-dependent manner and transported through endolysosomal pathways. After that, we used chemical inhibitors and RNA interference approaches to investigate possible mechanisms involved in S100B-Alexa488 uptake. The internalization of S100B-Alexa488 was inhibited by pitstop-2 or dyngo-4a treatment or RNA-mediated silencing of clathrin or dynamin, and the lipid raft-mediated endocytosis inhibitors nystatin and MßCD. In conclusion, our findings show that clathrin and lipid rafts contribute to the internalization of S100B-Alexa488, which provides promising interventions for the safe application of MSCs in glioma therapy.

15.
Comput Struct Biotechnol J ; 19: 4603-4618, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34471502

RESUMEN

BACKGROUND: Gliomas are one of the most common types of primary tumors in central nervous system. Previous studies have found that macrophages actively participate in tumor growth. METHODS: Weighted gene co-expression network analysis was used to identify meaningful macrophage-related gene genes for clustering. Pamr, SVM, and neural network were applied for validating clustering results. Somatic mutation and methylation were used for defining the features of identified clusters. Differentially expressed genes (DEGs) between the stratified groups after performing elastic regression and principal component analyses were used for the construction of MScores. The expression of macrophage-specific genes were evaluated in tumor microenvironment based on single cell sequencing analysis. A total of 2365 samples from 15 glioma datasets and 5842 pan-cancer samples were used for external validation of MScore. RESULTS: Macrophages were identified to be negatively associated with the survival of glioma patients. Twenty-six macrophage-specific DEGs obtained by elastic regression and PCA were highly expressed in macrophages at single-cell level. The prognostic value of MScores in glioma was validated by the active proinflammatory and metabolic profile of infiltrating microenvironment and response to immunotherapies of samples with this signature. MScores managed to stratify patient survival probabilities in 15 external glioma datasets and pan-cancer datasets, which predicted worse survival outcome. Sequencing data and immunohistochemistry of Xiangya glioma cohort confirmed the prognostic value of MScores. A prognostic model based on MScores demonstrated high accuracy rate. CONCLUSION: Our findings strongly support a modulatory role of macrophages, especially M2 macrophages in glioma progression and warrants further experimental studies.

16.
Biomed Pharmacother ; 121: 109610, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31710894

RESUMEN

Bromopyruvate (3-BrPA) is a glycolysis inhibitor that has been reported to have a strong anti-tumour effect in many human tumours. Several studies have reported that 3-BrPA could inhibit glioma progression; however, its role on the interstitial cells in the glioma microenvironment has not been investigated. In previous studies, we found that in the glioma microenvironment, glioma stem cells can induce the malignant transformation of macrophages and dendritic cells. In this study, we focused on the effects of 3-BrPA on malignantly transformed macrophages and dendritic cells. First, we found that 3-BrPA inhibited the proliferation of malignantly transformed macrophages and dendritic cells in a dose-dependent and time-dependent manner. Further study indicated that 3-BrPA significantly decreased extracellular lactate and inhibited the clone formation, migration and invasion of malignantly transformed macrophages and dendritic cells. Using an online database and a series of experiments, we demonstrated that 3-BrPA inhibits the malignant progression of malignantly transformed macrophages and dendritic cells via the miR-449a/MCT1 axis. These findings built experimental basis for new approach against glioma.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Células Dendríticas/patología , Glioma/tratamiento farmacológico , Macrófagos/patología , MicroARNs/fisiología , Transportadores de Ácidos Monocarboxílicos/fisiología , Células Madre Neoplásicas/fisiología , Piruvatos/farmacología , Simportadores/fisiología , Microambiente Tumoral , Células Cultivadas , Glioma/metabolismo , Glioma/patología , Ácido Láctico/metabolismo , Transportadores de Ácidos Monocarboxílicos/antagonistas & inhibidores , Simportadores/antagonistas & inhibidores
17.
Cancer Biol Med ; 17(1): 154-168, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-32296583

RESUMEN

Objective: Neutrophil extracellular traps (NETs) produced by tumor-infiltrating neutrophils (TINs) are associated with poor prognosis in patients with several types of cancer. However, the mechanisms underlying the involvement of NETs in glioma progression remain largely unknown. This study aimed to elucidate the roles of NETs in biological processes that drive the crosstalk between glioma progression and the tumor microenvironment. Methods: Neutrophil infiltration and NETs formation were investigated in glioma tissue through immunohistochemistry, and their relationships with clinicopathological features and outcomes were statistically evaluated. The effects of NETs on glioma cell progression were studied in a co-culture system. In vivo and in vitro experiments validated the reactive oxygen species activity and cytokine production of TINs, as well as the ERK signaling pathway activation and the metastasis of gliomas. Results: Neutrophil infiltration and NETs formation were induced in high-grade glioma compared with low-grade glioma. NETs induced by TINs were determined to be an oncogenic marker of high-grade gliomas and to be involved in cell proliferation and invasion. NETs overproduction promoted glioma cell proliferation, migration, and invasion. Furthermore, HMGB1 was found to bind to RAGE and activate the NF-κB signaling pathway in vitro. In addition, NETs stimulated the NF-κB signaling pathway, thus promoting IL-8 secretion in glioblastoma. Subsequently, IL-8 recruited neutrophils which in turn mediated NETs formation via the PI3K/AKT/ROS axis in TINs. Conclusions: Our results suggest that NETs produced by TINs mediate the crosstalk between glioma progression and the tumor microenvironment by regulating the HMGB1/RAGE/IL-8 axis. Targeting NETs formation or IL-8 secretion may be an effective approach to inhibit glioma progression.


Asunto(s)
Neoplasias Encefálicas/inmunología , Trampas Extracelulares/inmunología , Glioma/inmunología , Neutrófilos/inmunología , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología , Adulto , Antígenos de Neoplasias/metabolismo , Encéfalo/patología , Encéfalo/cirugía , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/cirugía , Estudios de Casos y Controles , Línea Celular Tumoral , Movimiento Celular/inmunología , Proliferación Celular , Técnicas de Cocultivo , Conjuntos de Datos como Asunto , Progresión de la Enfermedad , Trampas Extracelulares/metabolismo , Femenino , Glioma/diagnóstico , Glioma/patología , Glioma/cirugía , Proteína HMGB1/metabolismo , Voluntarios Sanos , Humanos , Interleucina-8/metabolismo , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Clasificación del Tumor , Invasividad Neoplásica/inmunología , Neutrófilos/metabolismo
18.
Front Immunol ; 11: 178, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194542

RESUMEN

Background: Glioblastoma (GBM) is one of the most malignant and aggressive primary brain tumors. The incurability of glioblastoma is heavily influenced by the glioma microenvironment. FTY720, a potent immunosuppressant, has been reported to exert anti-tumor effects in glioblastoma. However, the impact of FTY720 on the glioma microenvironment remains unclear. Methods: We examined the effects of FTY720 on the distribution and polarization of glioma-associated microglia and macrophages (GAMs) in glioma-bearing rats using immunofluorescence staining. qRT-PCR and Western blotting were used to detect the expressions of CXCR4 and MAPK pathway-related signal molecules on microglia in the coculture system. The levels of inflammatory factors were tested via ELISA. Wound healing assay and Matrigel invasion assay were used to determine the migration and invasion of C6 glioma cells. Results: We discovered that FTY720 could inhibit the growth, migration, and invasion of glioma by targeting GAMs to impede their effect on glioma cells. Simultaneously, FTY720 could block the chemoattraction of GAMs by inhibiting MAPK-mediated secretion of IL-6 through increased internalization of CXCR4. Moreover, microglia and macrophages are polarized from pro-glioma to an anti-tumor phenotype. Conclusion: These results provide novel insights into the inhibitory effects of FTY720 on glioma by targeting GAMs-glioma interaction in the tumor microenvironment.


Asunto(s)
Antineoplásicos/administración & dosificación , Clorhidrato de Fingolimod/administración & dosificación , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Microglía/efectos de los fármacos , Receptores CXCR4/metabolismo , Microambiente Tumoral/efectos de los fármacos , Aloinjertos , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Glioblastoma/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Microglía/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores CXCR4/genética , Transducción de Señal/efectos de los fármacos
19.
Biomed Pharmacother ; 131: 110666, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32853911

RESUMEN

Gliomas are the most common and aggressive primary tumours of the central nervous system in adults. Bone marrow-derived mesenchymal stem cells (BMSCs) are an important component of the glioma microenvironment. Our previous study indicated that BMSCs in the glioma microenvironment could be induced to malignantly transform by glioma stem cells (GSCs). The malignant transformation of BMSCs is closely related to glioma progression; however, the underlying mechanism of this transformation has not been fully clarified. In this study, we found that compared with the levels in normal BMSCs, the levels of the long noncoding RNA FTX transcript XIST regulator (lncRNA-FTX) were increased in malignantly transformed BMSCs (tBMSCs), which was associated with the proliferation, migration and invasion of tBMSCs. Next, by using a luciferase reporter assay and an RNA pull-down assay, we found that lncRNA-FTX acted as a sponge for miR-186 in tBMSCs. Further research revealed that miR-186 could bind to the 3'-UTR (untranslated region) of c-Met, which acts as an oncogene in gliomas. Through functional assays, we showed that lncRNA-FTX could regulate c-Met expression in tBMSCs in a miR-186-dependent manner. Based on these data, we concluded that lncRNA-FTX plays a key role in the GSC-mediated malignant transformation of BMSCs in the glioma microenvironment, which is of great significance for further understanding the pathogenesis of glioma.


Asunto(s)
Glioma/metabolismo , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , Fenotipo , ARN Largo no Codificante/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Glioma/patología , Células Madre Mesenquimatosas/patología , Ratones , Ratones Desnudos , Microambiente Tumoral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
20.
Front Immunol ; 11: 578877, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329549

RESUMEN

Glioma is the most malignant primary tumor of the central nervous system and is characterized by an extremely low overall survival. Recent breakthroughs in cancer therapy using immune checkpoint blockade have attracted significant attention. However, despite representing the most promising (immunotherapy) treatment for cancer, the clinical application of immune checkpoint blockade in glioma patients remains challenging due to the "cold phenotype" of glioma and multiple factors inducing resistance, both intrinsic and acquired. Therefore, comprehensive understanding of the tumor microenvironment and the unique immunological status of the brain will be critical for the application of glioma immunotherapy. More sensitive biomarkers to monitor the immune response, as well as combining multiple immunotherapy strategies, may accelerate clinical progress and enable development of effective and safe treatments for glioma patients.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Glioma/inmunología , Glioma/metabolismo , Glioma/patología , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Terapia Molecular Dirigida , Transducción de Señal , Resultado del Tratamiento , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA