Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 793
Filtrar
1.
Neurobiol Dis ; 196: 106521, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38697575

RESUMEN

BACKGROUND: Lesion network mapping (LNM) is a popular framework to assess clinical syndromes following brain injury. The classical approach involves embedding lesions from patients into a normative functional connectome and using the corresponding functional maps as proxies for disconnections. However, previous studies indicated limited predictive power of this approach in behavioral deficits. We hypothesized similarly low predictiveness for overall survival (OS) in glioblastoma (GBM). METHODS: A retrospective dataset of patients with GBM was included (n = 99). Lesion masks were registered in the normative space to compute disconnectivity maps. The brain functional normative connectome consisted in data from 173 healthy subjects obtained from the Human Connectome Project. A modified version of the LNM was then applied to core regions of GBM masks. Linear regression, classification, and principal component (PCA) analyses were conducted to explore the relationship between disconnectivity and OS. OS was considered both as continuous and categorical (low, intermediate, and high survival) variable. RESULTS: The results revealed no significant associations between OS and network disconnection strength when analyzed at both voxel-wise and classification levels. Moreover, patients stratified into different OS groups did not exhibit significant differences in network connectivity patterns. The spatial similarity among the first PCA of network maps for each OS group suggested a lack of distinctive network patterns associated with survival duration. CONCLUSIONS: Compared with indirect structural measures, functional indirect mapping does not provide significant predictive power for OS in patients with GBM. These findings are consistent with previous research that demonstrated the limitations of indirect functional measures in predicting clinical outcomes, underscoring the need for more comprehensive methodologies and a deeper understanding of the factors influencing clinical outcomes in this challenging disease.


Asunto(s)
Neoplasias Encefálicas , Conectoma , Glioblastoma , Imagen por Resonancia Magnética , Humanos , Glioblastoma/mortalidad , Glioblastoma/diagnóstico por imagen , Glioblastoma/fisiopatología , Masculino , Femenino , Neoplasias Encefálicas/fisiopatología , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/diagnóstico por imagen , Persona de Mediana Edad , Conectoma/métodos , Estudios Retrospectivos , Adulto , Anciano , Imagen por Resonancia Magnética/métodos , Encéfalo/fisiopatología , Encéfalo/diagnóstico por imagen , Red Nerviosa/diagnóstico por imagen , Red Nerviosa/fisiopatología
2.
Artículo en Inglés | MEDLINE | ID: mdl-38083627

RESUMEN

Glioblastoma (GBM) is the most aggressive high-grade brain cancer with a median survival time of <15 months. Due to GBMs fast and infiltrative growth patient prognosis is poor with recurrence after treatment common. Investigating GBMs ability to communicate, specifically via Ca2+ signaling, within its functional tumour networks may unlock new therapeutics to reduce the rapid infiltration and growth which currently makes treatment ineffective. This work aims to produce patterned networks of GBM cells such that the Ca2+ communication at a network level can be repeatedly and reliably investigated.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Sistemas Microfisiológicos , Humanos , Encéfalo/patología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/fisiopatología , Glioblastoma/patología , Glioblastoma/fisiopatología , Silicio
3.
Nature ; 619(7971): 844-850, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37380778

RESUMEN

The tumour microenvironment plays an essential role in malignancy, and neurons have emerged as a key component of the tumour microenvironment that promotes tumourigenesis across a host of cancers1,2. Recent studies on glioblastoma (GBM) highlight bidirectional signalling between tumours and neurons that propagates a vicious cycle of proliferation, synaptic integration and brain hyperactivity3-8; however, the identity of neuronal subtypes and tumour subpopulations driving this phenomenon is incompletely understood. Here we show that callosal projection neurons located in the hemisphere contralateral to primary GBM tumours promote progression and widespread infiltration. Using this platform to examine GBM infiltration, we identified an activity-dependent infiltrating population present at the leading edge of mouse and human tumours that is enriched for axon guidance genes. High-throughput, in vivo screening of these genes identified SEMA4F as a key regulator of tumourigenesis and activity-dependent progression. Furthermore, SEMA4F promotes the activity-dependent infiltrating population and propagates bidirectional signalling with neurons by remodelling tumour-adjacent synapses towards brain network hyperactivity. Collectively our studies demonstrate that subsets of neurons in locations remote to primary GBM promote malignant progression, and also show new mechanisms of glioma progression that are regulated by neuronal activity.


Asunto(s)
Neoplasias Encefálicas , Carcinogénesis , Glioma , Neuronas , Microambiente Tumoral , Humanos , Encéfalo/patología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/fisiopatología , Carcinogénesis/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Glioblastoma/patología , Glioblastoma/fisiopatología , Glioma/patología , Glioma/fisiopatología , Neuronas/patología , Proliferación Celular , Sinapsis , Progresión de la Enfermedad , Animales , Ratones , Axones , Cuerpo Calloso/patología , Vías Nerviosas
4.
J Neurosurg Sci ; 67(4): 422-430, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33297605

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) is the most common and deadly glioma subtype. Early growth response 1 (EGR1) participates in the progression of several cancer types, but the expression and function of EGR1 in GBM was rarely investigated. METHODS: The expressions of EGR1 in GBM were detected with qRT-PCR and immunohistochemistry in 12 pairs of fresh GBM tissues and 116 paraffin-embedded specimens. The patients were divided into high and low EGR1 groups according to the IHC score of EGR1, and the prognostic significances of different groups were evaluated with univariate and multivariate analyses. With in-vitro experiments, we assessed the role of EGR1 in the proliferation and invasion of GBM cells. RESULTS: In our study, EGR1 was up-regulated in GBM tissues compared with tumor-adjacent normal tissues. High expression of EGR1 or HMGB1 were unfavorable prognostic biomarkers of GBM. Coexpression of EGR1 and HMGB1 could predict the prognosis of GBM more sensitively. EGR1 facilitated the proliferation and invasion of GBM cells. Moreover, EGR1 promoted the invasion, instead of proliferation, of GBM cells by elevating the expression of HMGB1. CONCLUSIONS: ERG1 was a prognostic biomarker of GBM, and ERG1 and HMGB1 synergistically could predict the GBM prognosis more precisely. ERG1 could promote GBM cell invasion by inducing HMGB1 expression.


Asunto(s)
Neoplasias Encefálicas , Proteína 1 de la Respuesta de Crecimiento Precoz , Glioblastoma , Proteína HMGB1 , Invasividad Neoplásica , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Glioblastoma/diagnóstico , Glioblastoma/genética , Glioblastoma/fisiopatología , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/fisiopatología , Perfilación de la Expresión Génica , Humanos , Análisis Multivariante , Proliferación Celular/genética , Invasividad Neoplásica/genética , Regulación hacia Arriba/genética , Línea Celular Tumoral , Masculino , Femenino , Persona de Mediana Edad , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Biomarcadores de Tumor/genética , Mutación
5.
Exp Brain Res ; 240(12): 3183-3192, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36260096

RESUMEN

Body representation disorders are complex, varied, striking, and very disabling in most cases. Deficits of body representation have been described after lesions to multimodal and sensorimotor cortical areas. A few studies have reported the effects of tumors on the representation of the body, but little is known about the changes after tumor resection. Moreover, the impact of brain lesions on the hand size representation has been investigated in few clinical cases. Hands are of special importance, as no other body part has the ability for movement and interaction with the environment that the hands have, and we use them for a multitude of daily activities. Studies with clinical population can add further knowledge into the way hands are represented. Here, we report a single case study of a patient (AM) who was an expert bodybuilder and underwent a surgery to remove a glioblastoma in the left posterior prefrontal and precentral cortex at the level of the hand's motor region. Pre- (20 days) and post- (4 months) surgery assessment did not show any motor or cognitive impairments. A hand localization task was used, before and after surgery (12 months), to measure possible changes of the metric representation of his right hand. Results showed a post-surgery modulation of the typically distorted hand representation, with an overall accuracy improvement, especially on width dimension. These findings support the direct involvement of sensorimotor areas in the implicit representation of the body size and its relevance on defining specific size representation dimensions.


Asunto(s)
Imagen Corporal , Neoplasias Encefálicas , Glioblastoma , Mano , Procedimientos Neuroquirúrgicos , Corteza Sensoriomotora , Humanos , Imagen Corporal/psicología , Neoplasias Encefálicas/fisiopatología , Neoplasias Encefálicas/psicología , Neoplasias Encefálicas/cirugía , Mano/fisiopatología , Movimiento/fisiología , Corteza Sensoriomotora/fisiopatología , Glioblastoma/fisiopatología , Glioblastoma/psicología , Glioblastoma/cirugía , Procedimientos Neuroquirúrgicos/efectos adversos , Procedimientos Neuroquirúrgicos/psicología , Tamaño Corporal
6.
Sci Data ; 9(1): 453, 2022 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35906241

RESUMEN

Glioblastoma is the most common aggressive adult brain tumor. Numerous studies have reported results from either private institutional data or publicly available datasets. However, current public datasets are limited in terms of: a) number of subjects, b) lack of consistent acquisition protocol, c) data quality, or d) accompanying clinical, demographic, and molecular information. Toward alleviating these limitations, we contribute the "University of Pennsylvania Glioblastoma Imaging, Genomics, and Radiomics" (UPenn-GBM) dataset, which describes the currently largest publicly available comprehensive collection of 630 patients diagnosed with de novo glioblastoma. The UPenn-GBM dataset includes (a) advanced multi-parametric magnetic resonance imaging scans acquired during routine clinical practice, at the University of Pennsylvania Health System, (b) accompanying clinical, demographic, and molecular information, (d) perfusion and diffusion derivative volumes, (e) computationally-derived and manually-revised expert annotations of tumor sub-regions, as well as (f) quantitative imaging (also known as radiomic) features corresponding to each of these regions. This collection describes our contribution towards repeatable, reproducible, and comparative quantitative studies leading to new predictive, prognostic, and diagnostic assessments.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Adulto , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/fisiopatología , Genómica , Glioblastoma/diagnóstico por imagen , Glioblastoma/genética , Glioblastoma/fisiopatología , Humanos , Imagen por Resonancia Magnética , Pronóstico
7.
Sci Rep ; 12(1): 2417, 2022 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-35165339

RESUMEN

Programmed death ligand 1 (PD-L1) plays a key role in glioblastoma multiforme (GBM) immunosuppression, vitality, proliferation, and migration, and is therefore a promising target for treating GBM. CRISPR/Cas9-mediated genomic editing can delete both cell surface and intracellular PD-L1. This systemic deliverable genomic PD-L1 deletion system can be used as an effective anti-GBM therapy by inhibiting tumor growth and migration, and overcoming immunosuppression. To target PD-L1 for CRISPR/Cas9 gene editing, we first identified two single guide RNA (sgRNA) sequences located on PD-L1 exon 3. The first sgRNA recognizes the forward strand of human PD-L1 near the beginning of exon 3 that allows editing by Cas9 at approximately base pair 82 (g82). The second sgRNA recognizes the forward strand of exon 3 that directs cutting at base pair 165 (g165). A homology-directed repair template (HDR) combined with the dual-sgRNAs was used to improve PD-L1 knockout specificity and efficiency. sgRNAs g82 and g165 were cloned into the multiplex CRISPR/Cas9 assembly system and co-transfected with the HDR template in human U87 GBM cells (g82/165 + HDR). T7E1 analysis suggests that the dual-sgRNA CRISPR/Cas9 strategy with a repair template was capable of editing the genomic level of PD-L1. This was further confirmed by examining PD-L1 protein levels by western blot and immunofluorescence assays. Western blot analysis showed that the dual-sgRNAs with the repair template caused a 64% reduction of PD-L1 protein levels in U87 cells, while immunostaining showed a significant reduction of intracellular PD-L1. PD-L1 deletion inhibited proliferation, growth, invasion and migration of U87 cells, indicating intracellular PD-L1 is necessary for tumor progression. Importantly, U87 cells treated with g82/165 + HDR polarized tumor-associated macrophages (TAM) toward an M1 phenotype, as indicated by an increase in TNF-α and a decrease in IL-4 secretions. This was further confirmed with flow cytometry that showed an increase in the M1 markers Ly6C + and CD80 +, and a decrease in the M2 marker CD206 + both in vitro and in vivo. Utilizing dual-sgRNAs and an HDR template with the CRISPR/Cas9 gene-editing system is a promising avenue for the treatment of GBM.


Asunto(s)
Antígeno B7-H1/genética , Polaridad Celular , Glioblastoma/genética , Glioblastoma/fisiopatología , Macrófagos Asociados a Tumores/citología , Antígeno B7-H1/metabolismo , Sistemas CRISPR-Cas , Línea Celular Tumoral , Proliferación Celular , Exones , Edición Génica , Técnicas de Silenciamiento del Gen , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Interleucina-4/genética , Interleucina-4/metabolismo , Invasividad Neoplásica , ARN Guía de Kinetoplastida , Macrófagos Asociados a Tumores/metabolismo
8.
Sci Rep ; 12(1): 2341, 2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35149717

RESUMEN

The growth of glioblastoma (GBM), one of the deadliest adult cancers, is fuelled by a subpopulation of stem/progenitor cells, which are thought to be the source of resistance and relapse after treatment. Re-engagement of a latent capacity of these cells to re-enter a trajectory resulting in cell differentiation is a potential new therapeutic approach for this devastating disease. ASCL1, a proneural transcription factor, plays a key role in normal brain development and is also expressed in a subset of GBM cells, but fails to engage a full differentiation programme in this context. Here, we investigated the barriers to ASCL1-driven differentiation in GBM stem cells. We see that ASCL1 is highly phosphorylated in GBM stem cells where its expression is compatible with cell proliferation. However, overexpression of a form of ASCL1 that cannot be phosphorylated on Serine-Proline sites drives GBM cells down a neuronal lineage and out of cell cycle more efficiently than its wild-type counterpart, an effect further enhanced by deletion of the inhibitor of differentiation ID2, indicating mechanisms to reverse the block to GBM cell differentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/fisiopatología , Glioblastoma/metabolismo , Glioblastoma/fisiopatología , Proteína 2 Inhibidora de la Diferenciación/genética , Células Madre Neoplásicas/metabolismo , Secuencias de Aminoácidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Neoplasias Encefálicas/genética , Ciclo Celular , Diferenciación Celular , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Humanos , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Células Madre Neoplásicas/citología , Fosforilación
9.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35163279

RESUMEN

Glioblastoma (GBM) is a devastating type of brain tumor, and current therapeutic treatments, including surgery, chemotherapy, and radiation, are palliative at best. The design of effective and targeted chemotherapeutic strategies for the treatment of GBM require a thorough analysis of specific signaling pathways to identify those serving as drivers of GBM progression and invasion. The Wnt/ß-catenin and PI3K/Akt/mTOR (PAM) signaling pathways are key regulators of important biological functions that include cell proliferation, epithelial-mesenchymal transition (EMT), metabolism, and angiogenesis. Targeting specific regulatory components of the Wnt/ß-catenin and PAM pathways has the potential to disrupt critical brain tumor cell functions to achieve critical advancements in alternative GBM treatment strategies to enhance the survival rate of GBM patients. In this review, we emphasize the importance of the Wnt/ß-catenin and PAM pathways for GBM invasion into brain tissue and explore their potential as therapeutic targets.


Asunto(s)
Glioblastoma/metabolismo , Transducción de Señal/fisiología , Apoptosis/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/fisiología , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/fisiopatología , Humanos , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo
10.
Int J Mol Sci ; 23(2)2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-35054765

RESUMEN

Elevation of intracellular cAMP levels has been implicated in glioma cell proliferation inhibition, differentiation, and apoptosis. Inhibition of phosphodiesterase is a way to elevate intracellular cAMP levels. The present study aimed to investigate the anti-glioma potential of dipyridamole, an inhibitor of phosphodiesterase. Upon treatment with dipyridamole, human U87 glioma cells decreased cell viability, clonogenic colonization, migration, and invasion, along with Noxa upregulation, Endoplasmic Reticulum (ER) stress, impaired autophagic flux, Yes-associated Protein 1 (YAP1) phosphorylation, and YAP1 reduction. Pharmacological and genetic studies revealed the ability of dipyridamole to initiate Noxa-guided apoptosis through ER stress. Additionally, the current study further identified the biochemical role of YAP1 in communicating with ER stress and autophagy under situations of dipyridamole treatment. YAP1 promoted autophagy and protected glioma cells from dipyridamole-induced apoptotic cell death. Dipyridamole impaired autophagic flux and rendered glioma cells more vulnerable to apoptotic cell death through ER stress-inhibitable YAP1/autophagy axis. The overall cellular changes caused by dipyridamole appeared to ensure a successful completion of apoptosis. Dipyridamole also duplicated the biochemical changes and apoptosis in glioma T98G cells. Since dipyridamole has additional biochemical and pharmacological properties, further research centered on the anti-glioma mechanisms of dipyridamole is still needed.


Asunto(s)
Apoptosis , Autofagia , Dipiridamol/farmacología , Estrés del Retículo Endoplásmico , Glioblastoma/tratamiento farmacológico , Antineoplásicos/farmacología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/fisiopatología , Humanos , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Señalizadoras YAP/genética , Proteínas Señalizadoras YAP/metabolismo
11.
Int J Mol Sci ; 23(2)2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-35054779

RESUMEN

The tumor microenvironment is a highly dynamic accumulation of resident and infiltrating tumor cells, responsible for growth and invasion. The authors focused on the leading-edge concepts regarding the glioblastoma microenvironment. Due to the fact that the modern trend in the research and treatment of glioblastoma is represented by multiple approaches that target not only the primary tumor but also the neighboring tissue, the study of the microenvironment in the peritumoral tissue is an appealing direction for current and future therapies.


Asunto(s)
Glioblastoma/fisiopatología , Microambiente Tumoral , Humanos
12.
Mol Cancer Res ; 20(2): 319-331, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34654721

RESUMEN

Glioblastoma multiforme (GBM), classified as World Health Organization grade IV astrocytoma, is the deadliest adult cancer of the central nervous system. An important contributing factor to poor survival rates in GBM is extensive invasion, which decreases the efficacy of resection and subsequent adjuvant therapies. These treatments could be markedly improved with increased resolution of the genetic and molecular initiators and effectors of invasion. Connexin 43 (Cx43) is the principal astrocytic gap junction (GJ) protein. Despite the heterogeneity of GBM, a subpopulation of cells in almost all GBM tumors express Cx43. Functional GJs between GBM cells and astrocytes at the tumor edge are of critical interest for understanding invasion. In this study, we find that both in vitro and in ex vivo slice cultures, GBM is substantially less invasive when placed in a Cx43-deficient astrocyte environment. Furthermore, when Cx43 is deleted in GBM, the invasive phenotype is recovered. These data strongly suggest that there are opposing roles for Cx43 in GBM migration. We find that Cx43 is localized to the tumor edge in our ex vivo model, suggesting that GBM-astrocyte GJ communication at the tumor border is a driving force for invasion. Finally, we find that by a Cx43-dependent mechanism, but likely not direct channel-mediated diffusion, miRNAs associated with cell-matrix adhesion are transferred from GBM to astrocytes and miR-19b promotes invasion, revealing a role for post-transcriptional manipulation of astrocytes in fostering an invasion-permissive peritumoral niche. IMPLICATIONS: Cx43-mediated communication, specifically miRNA transfer, profoundly impacts glioblastoma invasion and may enable further therapeutic insight.


Asunto(s)
Astrocitos/metabolismo , Conexina 43/metabolismo , Uniones Comunicantes/patología , Glioblastoma/fisiopatología , Humanos , Invasividad Neoplásica
13.
Neurosci Lett ; 766: 136351, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34793898

RESUMEN

GBM is the most life-threatening neurological disease with annual incidence of âˆ¼ 5 cases per 100,000 people and a median survival of less than 15 months. Seizures are the first clinical symptoms in 40%-45% of patients with GBM and its epileptogenic mechanisms are poorly understood, largely due to the challenge to develop a clinically-relevant animal model and the unknown latent period. In this study, we used continuous video-EEG monitoring to detect the earliest interictal and ictal events in a CRISPR- IUE GBM rat model that shares pathological and clinical features with those observed in human patients. To our best knowledge, we showed for the first time that interictal epileptiform discharges emerged during early postnatal weeks and the first ictal event occurred during the fourth postnatal week. We also showed GBM animals showed independent bi-hemispheric epileptogenic events, suggesting a widespread circuitry dysregulation. Together, our work identified the temporal- and spatial frame of epileptogenic network in a highly clinically-relevant GBM animal model, paving ways for mechanistic studies at molecular, cellular and circuitry levels.


Asunto(s)
Neoplasias Encefálicas/fisiopatología , Cerebro/fisiopatología , Modelos Animales de Enfermedad , Glioblastoma/fisiopatología , Convulsiones/fisiopatología , Animales , Neoplasias Encefálicas/complicaciones , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Electroencefalografía/métodos , Electroporación , Epilepsia/etiología , Epilepsia/fisiopatología , Femenino , Masculino , Ratas , Ratas Sprague-Dawley , Convulsiones/etiología , Grabación en Video
14.
Int J Oncol ; 60(1)2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34970698

RESUMEN

Tumor­treating fields (TTFields) are emerging cancer therapies based on alternating low­intensity electric fields that interfere with dividing cells and induce cancer cell apoptosis. However, to date, there is limited knowledge of their effects on normal cells, as well as the effects of different duty cycles on outcomes. The present study evaluated the effects of TTFields with different duty cycles on glioma spheroid cells and normal brain organoids. A customized TTFields system was developed to perform in vitro experiments with varying duty cycles. Three duty cycles were applied to three types of glioma spheroid cells and brain organoids. The efficacy and safety of the TTFields were evaluated by analyzing the cell cycle of glioma cells, and markers of neural stem cells (NSCs) and astrocytes in brain organoids. The application of the TTFields at the 75 and 100% duty cycle markedly inhibited the proliferation of the U87 and U373 compared with the control. FACS analysis revealed that the higher the duty cycle of the applied fields, the greater the increase in apoptosis detected. Exposure to a higher duty cycle resulted in a greater decrease in NSC markers and a greater increase in glial fibrillary acidic protein expression in normal brain organoids. These results suggest that TTFields at the 75 and 100% duty cycle induced cancer cell death, and that the neurotoxicity of the TTFields at 75% was less prominent than that at 100%. Although clinical studies with endpoints related to safety and efficacy need to be performed before this strategy may be adopted clinically, the findings of the present study provide meaningful evidence for the further advancement of TTFields in the treatment of various types of cancer.


Asunto(s)
Apoptosis , Encéfalo/fisiopatología , Glioblastoma/terapia , Magnetoterapia/normas , Organoides , Glioblastoma/fisiopatología , Humanos , Magnetoterapia/métodos , Magnetoterapia/estadística & datos numéricos
15.
Nat Commun ; 12(1): 7155, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34887423

RESUMEN

Pyroptosis induced by the N-terminal gasdermin domain (GSDMNT) holds great potential for anti-tumor therapy. However, due to the extreme cytoxicity of GSDMNT, it is challenging to efficiently produce and deliver GSDMNT into tumor cells. Here, we report the development of two strategies to package recombinant adeno-associated virus (rAAV) expressing GSDMNT: 1) drive the expression of GSDMNT by a mammal specific promoter and package the virus in Sf9 insect cells to avoid its expression; 2) co-infect rAAV-Cre to revert and express the double-floxed inverted GSDMNT. We demonstrate that these rAAVs can induce pyroptosis and prolong survival in preclinical cancer models. The oncolytic-viruses induce pyroptosis and evoke a robust immune-response. In a glioblastoma model, rAAVs temporarily open the blood-brain barrier and recruit tumor infiltrating lymphocytes into the brain. The oncolytic effect is further improved in combination with anti-PD-L1. Together, our strategies efficiently produce and deliver GSDMNT into tumor cells and successfully induce pyroptosis, which can be exploited for anti-tumor therapy.


Asunto(s)
Neoplasias de la Mama/terapia , Dependovirus/genética , Glioblastoma/fisiopatología , Glioblastoma/terapia , Proteínas de Neoplasias/genética , Piroptosis , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/fisiopatología , Línea Celular Tumoral , Dependovirus/fisiología , Femenino , Glioblastoma/genética , Glioblastoma/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/inmunología , Viroterapia Oncolítica , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Ratas , Ratas Wistar , Células Sf9 , Empaquetamiento del Genoma Viral
16.
Int J Mol Sci ; 22(24)2021 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-34948346

RESUMEN

Glioblastoma (GBM), a grade IV brain tumor, is known for its heterogenicity and its resistance to the current treatment regimen. Over the last few decades, a significant amount of new molecular and genetic findings has been reported regarding factors contributing to GBM's development into a lethal phenotype and its overall poor prognosis. MicroRNA (miRNAs) are small non-coding sequences of RNA that regulate and influence the expression of multiple genes. Many research findings have highlighted the importance of miRNAs in facilitating and controlling normal biological functions, including cell differentiation, proliferation, and apoptosis. Furthermore, miRNAs' ability to initiate and promote cancer development, directly or indirectly, has been shown in many types of cancer. There is a clear association between alteration in miRNAs expression in GBM's ability to escape apoptosis, proliferation, and resistance to treatment. Further, miRNAs regulate the already altered pathways in GBM, including P53, RB, and PI3K-AKT pathways. Furthermore, miRNAs also contribute to autophagy at multiple stages. In this review, we summarize the functions of miRNAs in GBM pathways linked to dysregulation of cell cycle control, apoptosis and resistance to treatment, and the possible use of miRNAs in clinical settings as treatment and prediction biomarkers.


Asunto(s)
Apoptosis , Ciclo Celular , Glioblastoma/metabolismo , MicroARNs/metabolismo , Autofagia , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/fisiopatología , Humanos , Transducción de Señal
17.
Clin Epigenetics ; 13(1): 201, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732244

RESUMEN

BACKGROUND: Glioblastoma (GBM) is a malignant human brain tumor that has an extremely poor prognosis. Classic mutations such as IDH (isocitrate dehydrogenase) mutations, EGFR (epidermal growth factor receptor) alternations, and MGMT (O6-methylguanine-methyltransferase) promoter hypermethylation have been used to stratify patients and provide prognostic significance. Epigenetic perturbations have been demonstrated in glioblastoma tumorigenesis. Despite the genetic markers used in the management of glioblastoma patients, new biomarkers that could predict patient survival independent of known biomarkers remain to be identified. METHODS: ATAC-seq (assay for transposase accessible chromatin followed by sequencing) and RNA-seq have been used to profile chromatin accessible regions using glioblastoma patient samples with short-survival versus long-survival. Cell viability, cell cycle, and Western blot analysis were used to characterize the cellular phenotypes and identify signaling pathways. RESULTS: Analysis of chromatin accessibility by ATAC-seq coupled with RNA-seq methods identified the GSTM1 (glutathione S-transferase mu-1) gene, which featured higher chromatin accessibility in GBM tumors with short survival. GSTM1 was confirmed to be a significant prognostic marker to predict survival using a different GBM patient cohort. Knockdown of GSTM1 decreased cell viability, caused cell cycle arrest, and decreased the phosphorylation levels of the NF-kB (nuclear factor kappa B) p65 subunit and STAT3 (signal transducer and activator of transcription 3) (pSer727). CONCLUSIONS: This report demonstrates the use of ATAC-seq coupled with RNA-seq to identify GSTM1 as a prognostic marker of GBM patient survival. Activation of phosphorylation levels of NF-kB p65 and STAT3 (pSer727) by GSTM1 is shown. Analysis of chromatin accessibility in patient samples could generate an independent biomarker that can be used to predict patient survival.


Asunto(s)
Cromatina/metabolismo , Glioblastoma/diagnóstico , Glutatión Transferasa/análisis , Biomarcadores/análisis , Biomarcadores/sangre , Cromatina/genética , Glioblastoma/epidemiología , Glioblastoma/fisiopatología , Glutatión Transferasa/sangre , Humanos , Estimación de Kaplan-Meier , Pronóstico
18.
Nat Commun ; 12(1): 6130, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34675201

RESUMEN

Epigenetic mechanisms which play an essential role in normal developmental processes, such as self-renewal and fate specification of neural stem cells (NSC) are also responsible for some of the changes in the glioblastoma (GBM) genome. Here we develop a strategy to compare the epigenetic and transcriptional make-up of primary GBM cells (GIC) with patient-matched expanded potential stem cell (EPSC)-derived NSC (iNSC). Using a comparative analysis of the transcriptome of syngeneic GIC/iNSC pairs, we identify a glycosaminoglycan (GAG)-mediated mechanism of recruitment of regulatory T cells (Tregs) in GBM. Integrated analysis of the transcriptome and DNA methylome of GBM cells identifies druggable target genes and patient-specific prediction of drug response in primary GIC cultures, which is validated in 3D and in vivo models. Taken together, we provide a proof of principle that this experimental pipeline has the potential to identify patient-specific disease mechanisms and druggable targets in GBM.


Asunto(s)
Neoplasias Encefálicas/genética , Glioblastoma/genética , Células Madre Neoplásicas/metabolismo , Células-Madre Neurales/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/fisiopatología , Diferenciación Celular , Metilación de ADN , Epigénesis Genética , Epigenómica , Glioblastoma/metabolismo , Glioblastoma/fisiopatología , Humanos , Ratones , Transcripción Genética
19.
Int J Mol Sci ; 22(19)2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34638796

RESUMEN

Diosmin, a natural flavone glycoside acquired through dehydrogenation of the analogous flavanone glycoside hesperidin, is plentiful in many citrus fruits. Glioblastoma multiforme (GBM) is the most malignant primary brain tumor; the average survival time of GBM patients is less than 18 months after standard treatment. The present study demonstrated that diosmin, which is able to cross the blood-brain barrier, inhibited GBM cell growth in vitro and in vivo. Diosmin also impeded migration and invasion by GBM8401and LN229 GBM cells by suppressing epithelial-mesenchymal transition, as indicated by increased expression of E-cadherin and decreased expression of Snail and Twist. Diosmin also suppressed autophagic flux, as indicated by increased expression of LC3-II and p62, and induced cell cycle arrest at G1 phase. Importantly, diosmin did not exert serious cytotoxic effects toward control SVG-p12 astrocytes, though it did reduce astrocyte viability at high concentrations. These findings provide potentially helpful support to the development of new therapies for the treatment of GBM.


Asunto(s)
Autofagia/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Diosmina/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/fisiopatología , Línea Celular Tumoral , Diosmina/uso terapéutico , Femenino , Glioblastoma/fisiopatología , Humanos , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Int J Mol Sci ; 22(15)2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34360836

RESUMEN

Lysyl oxidase-like 3 (LOXL3), belonging to the lysyl oxidase family, is responsible for the crosslinking in collagen or elastin. The cellular localization of LOXL3 is in the extracellular space by reason of its canonical function. In tumors, the presence of LOXL3 has been associated with genomic stability, cell proliferation, and metastasis. In silico analysis has shown that glioblastoma was among tumors with the highest LOXL3 expression levels. LOXL3 silencing of U87MG cells by siRNA led to the spreading of the tumor cell surface, and the transcriptome analysis of these cells revealed an upregulation of genes coding for extracellular matrix, cell adhesion, and cytoskeleton components, convergent to an increase in cell adhesion and a decrease in cell invasion observed in functional assays. Significant correlations of LOXL3 expression with genes coding for tubulins were observed in the mesenchymal subtype in the TCGA RNA-seq dataset of glioblastoma (GBM). Conversely, genes involved in endocytosis and lysosome formation, along with MAPK-binding proteins related to focal adhesion turnover, were downregulated, which may corroborate the observed decrease in cell viability and increase in the rate of cell death. Invasiveness is a major determinant of the recurrence and poor outcome of GBM patients, and downregulation of LOXL3 may contribute to halting the tumor cell invasion.


Asunto(s)
Aminoácido Oxidorreductasas/metabolismo , Adhesión Celular , Regulación Neoplásica de la Expresión Génica , Glioblastoma/enzimología , Aminoácido Oxidorreductasas/genética , Aminoácido Oxidorreductasas/fisiología , Línea Celular Tumoral , Proliferación Celular , Simulación por Computador , Citoesqueleto/metabolismo , Endocitosis , Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica , Glioblastoma/genética , Glioblastoma/patología , Glioblastoma/fisiopatología , Humanos , Lisosomas/fisiología , Invasividad Neoplásica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA