Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.269
Filtrar
1.
Zhongguo Fei Ai Za Zhi ; 27(5): 376-382, 2024 May 20.
Artículo en Chino | MEDLINE | ID: mdl-38880925

RESUMEN

Leptomeningeal metastasis (LM) is a lethal complication of malignant tumors, with an incidence rate of 3%-5% among patients with non-small cell lung cancer (NSCLC). LM poses significant challenges in diagnosis, has poor prognosis, limited treatment options, and lacks standardized criteria for evaluating therapeutic efficacy, making it a difficult aspect of NSCLC management. Circulating tumor DNA (ctDNA), shed from tumor cells and carrying cancer-related information, holds significant value in precision oncology. Cerebrospinal fluid (CSF), present in the subarachnoid space of the brain, the spinal cord, and the central canal, and in direct contact with meningeal tissues, serves as the fluid medium that best reflects the genetic characteristics of LM. In recent years, CSF ctDNA has become a focal point due to its multi-omics features, playing a crucial role in the management of central nervous system (CNS) metastatic tumors. Its applications span the entire continuum of care, including aiding in diagnosis, assessing treatment response, predicting prognosis, and analyzing resistance mechanisms. This article provides a concise overview of CSF ctDNA detection techniques and their clinical applications in patients with NSCLC-LM.
.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , ADN Tumoral Circulante , Neoplasias Pulmonares , Neoplasias Meníngeas , Humanos , Carcinoma de Pulmón de Células no Pequeñas/líquido cefalorraquídeo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , ADN Tumoral Circulante/líquido cefalorraquídeo , ADN Tumoral Circulante/genética , ADN Tumoral Circulante/sangre , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/líquido cefalorraquídeo , Neoplasias Pulmonares/genética , Neoplasias Meníngeas/secundario , Neoplasias Meníngeas/líquido cefalorraquídeo , Neoplasias Meníngeas/genética
2.
Cell Rep Med ; 5(6): 101606, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38866016

RESUMEN

Leptomeningeal disease (LMD) remains a rapidly lethal complication for late-stage melanoma patients. Here, we characterize the tumor microenvironment of LMD and patient-matched extra-cranial metastases using spatial transcriptomics in a small number of clinical specimens (nine tissues from two patients) with extensive in vitro and in vivo validation. The spatial landscape of melanoma LMD is characterized by a lack of immune infiltration and instead exhibits a higher level of stromal involvement. The tumor-stroma interactions at the leptomeninges activate tumor-promoting signaling, mediated through upregulation of SERPINA3. The meningeal stroma is required for melanoma cells to survive in the cerebrospinal fluid (CSF) and promotes MAPK inhibitor resistance. Knocking down SERPINA3 or inhibiting the downstream IGR1R/PI3K/AKT axis results in tumor cell death and re-sensitization to MAPK-targeting therapy. Our data provide a spatial atlas of melanoma LMD, identify the tumor-promoting role of meningeal stroma, and demonstrate a mechanism for overcoming microenvironment-mediated drug resistance in LMD.


Asunto(s)
Melanoma , Neoplasias Meníngeas , Células del Estroma , Microambiente Tumoral , Melanoma/genética , Melanoma/patología , Humanos , Microambiente Tumoral/genética , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Células del Estroma/metabolismo , Células del Estroma/patología , Animales , Línea Celular Tumoral , Ratones , Regulación Neoplásica de la Expresión Génica , Transcriptoma/genética , Perfilación de la Expresión Génica , Meninges/patología , Meninges/metabolismo , Resistencia a Antineoplásicos/genética , Transducción de Señal , Femenino
3.
J Neurooncol ; 168(3): 515-524, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38811523

RESUMEN

PURPOSE: Accurate classification of cancer subgroups is essential for precision medicine, tailoring treatments to individual patients based on their cancer subtypes. In recent years, advances in high-throughput sequencing technologies have enabled the generation of large-scale transcriptomic data from cancer samples. These data have provided opportunities for developing computational methods that can improve cancer subtyping and enable better personalized treatment strategies. METHODS: Here in this study, we evaluated different feature selection schemes in the context of meningioma classification. To integrate interpretable features from the bulk (n = 77 samples) and single-cell profiling (∼ 10 K cells), we developed an algorithm named CLIPPR which combines the top-performing single-cell models, RNA-inferred copy number variation (CNV) signals, and the initial bulk model to create a meta-model. RESULTS: While the scheme relying solely on bulk transcriptomic data showed good classification accuracy, it exhibited confusion between malignant and benign molecular classes in approximately ∼ 8% of meningioma samples. In contrast, models trained on features learned from meningioma single-cell data accurately resolved the sub-groups confused by bulk-transcriptomic data but showed limited overall accuracy. CLIPPR showed superior overall accuracy and resolved benign-malignant confusion as validated on n = 789 bulk meningioma samples gathered from multiple institutions. Finally, we showed the generalizability of our algorithm using our in-house single-cell (∼ 200 K cells) and bulk TCGA glioma data (n = 711 samples). CONCLUSION: Overall, our algorithm CLIPPR synergizes the resolution of single-cell data with the depth of bulk sequencing and enables improved cancer sub-group diagnoses and insights into their biology.


Asunto(s)
Algoritmos , Neoplasias Meníngeas , Meningioma , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Humanos , Análisis de la Célula Individual/métodos , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/clasificación , Meningioma/genética , Meningioma/patología , Meningioma/clasificación , Análisis de Secuencia de ARN/métodos , Variaciones en el Número de Copia de ADN , Biomarcadores de Tumor/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Transcriptoma , Perfilación de la Expresión Génica/métodos
4.
Cell Genom ; 4(6): 100566, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38788713

RESUMEN

Meningiomas, although mostly benign, can be recurrent and fatal. World Health Organization (WHO) grading of the tumor does not always identify high-risk meningioma, and better characterizations of their aggressive biology are needed. To approach this problem, we combined 13 bulk RNA sequencing (RNA-seq) datasets to create a dimension-reduced reference landscape of 1,298 meningiomas. The clinical and genomic metadata effectively correlated with landscape regions, which led to the identification of meningioma subtypes with specific biological signatures. The time to recurrence also correlated with the map location. Further, we developed an algorithm that maps new patients onto this landscape, where the nearest neighbors predict outcome. This study highlights the utility of combining bulk transcriptomic datasets to visualize the complexity of tumor populations. Further, we provide an interactive tool for understanding the disease and predicting patient outcomes. This resource is accessible via the online tool Oncoscape, where the scientific community can explore the meningioma landscape.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Transcriptoma , Meningioma/genética , Meningioma/patología , Humanos , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Masculino , Femenino , Persona de Mediana Edad , Regulación Neoplásica de la Expresión Génica , Algoritmos , Perfilación de la Expresión Génica/métodos
5.
Nat Genet ; 56(6): 1121-1133, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38760638

RESUMEN

Intratumor heterogeneity underlies cancer evolution and treatment resistance, but targetable mechanisms driving intratumor heterogeneity are poorly understood. Meningiomas are the most common primary intracranial tumors and are resistant to all medical therapies, and high-grade meningiomas have significant intratumor heterogeneity. Here we use spatial approaches to identify genomic, biochemical and cellular mechanisms linking intratumor heterogeneity to the molecular, temporal and spatial evolution of high-grade meningiomas. We show that divergent intratumor gene and protein expression programs distinguish high-grade meningiomas that are otherwise grouped together by current classification systems. Analyses of matched pairs of primary and recurrent meningiomas reveal spatial expansion of subclonal copy number variants associated with treatment resistance. Multiplexed sequential immunofluorescence and deconvolution of meningioma spatial transcriptomes using cell types from single-cell RNA sequencing show decreased immune infiltration, decreased MAPK signaling, increased PI3K-AKT signaling and increased cell proliferation, which are associated with meningioma recurrence. To translate these findings to preclinical models, we use CRISPR interference and lineage tracing approaches to identify combination therapies that target intratumor heterogeneity in meningioma cell co-cultures.


Asunto(s)
Heterogeneidad Genética , Neoplasias Meníngeas , Meningioma , Meningioma/genética , Meningioma/patología , Humanos , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Variaciones en el Número de Copia de ADN , Regulación Neoplásica de la Expresión Génica , Genómica/métodos , Análisis de la Célula Individual , Proliferación Celular/genética , Recurrencia Local de Neoplasia/genética , Transducción de Señal/genética , Línea Celular Tumoral , Transcriptoma
6.
J Mol Neurosci ; 74(2): 54, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760510

RESUMEN

This article discusses a rare case of coexistent meningiomas and Primary familial brain calcification (PFBC). PFBC is a neurodegenerative disease characterized by brain calcifications and a variety of neuropsychiatric symptoms and signs, with pathogenic variants in specific genes. The study explores the potential link between PFBC and meningiomas, highlighting shared features like intralesional calcifications and common genes such as MEA6. The article also revisits PFBC patients developing other brain tumors, particularly gliomas, emphasizing the intersection of oncogenes like PDGFB and PDGFRB in both calcifications and tumor progression. In recent investigations, attention has extended beyond brain tumors to breast cancer metastasis, unveiling a noteworthy connection. These findings suggest a broader connection between brain calcifications and tumors, encouraging a reevaluation of therapeutic approaches for PFBC.


Asunto(s)
Neoplasias Encefálicas , Calcinosis , Meningioma , Humanos , Calcinosis/genética , Calcinosis/patología , Meningioma/genética , Meningioma/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Femenino , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Encefalopatías/genética , Encefalopatías/patología , Encefalopatías/metabolismo
7.
Acta Neuropathol Commun ; 12(1): 74, 2024 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-38720399

RESUMEN

The combination of DNA methylation analysis with histopathological and genetic features allows for a more accurate risk stratification and classification of meningiomas. Nevertheless, the implications of this classification for patients with grade 2 meningiomas, a particularly heterogeneous tumor entity, are only partially understood. We correlate the outcomes of histopathologically confirmed grade 2 meningioma with an integrated molecular-morphologic risk stratification and determine its clinical implications. Grade 2 meningioma patients treated at our institution were re-classified using an integrated risk stratification involving DNA methylation array-based data, copy number assessment and TERT promoter mutation analyses. Grade 2 meningioma cases according to the WHO 2021 criteria treated between 2007 and 2021 (n = 100) were retrospectively analyzed. The median clinical and radiographic follow-up periods were 59.8 and 54.4 months. A total of 38 recurrences and 17 deaths were observed. The local control rates of the entire cohort after 2-, 4-, and 6-years were 84.3%, 68.5%, and 50.8%, with a median local control time of 77.2 months. The distribution of the integrated risk groups were as follows: 31 low, 54 intermediate, and 15 high risk cases. In the multivariable Cox regression analysis, integrated risk groups were significantly associated with the risk of local recurrence (hazard ratio (HR) intermediate: 9.91, HR high-risk: 7.29, p < 0.01). Gross total resections decreased the risk of local tumor progression (HR gross total resection: 0.19, p < 0.01). The comparison of 1p status and integrated risk groups (low vs. intermediate/high) revealed nearly identical local control rates within their respective subgroups. In summary, only around 50% of WHO 2021 grade 2 meningiomas have an intermediate risk profile. Integrated molecular risk stratification is crucial to guide the management of patients with grade 2 tumors and should be routinely applied to avoid over- and undertreatment, especially concerning the use of adjuvant radiotherapy.


Asunto(s)
Metilación de ADN , Neoplasias Meníngeas , Meningioma , Humanos , Meningioma/genética , Meningioma/patología , Meningioma/clasificación , Masculino , Femenino , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/clasificación , Persona de Mediana Edad , Anciano , Adulto , Estudios Retrospectivos , Clasificación del Tumor , Anciano de 80 o más Años , Telomerasa/genética , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/genética
8.
Genes Chromosomes Cancer ; 63(5): e23248, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38801095

RESUMEN

Meningiomas are non-glial tumors that are the most common primary brain tumors in adults. Although meningioma can possibly be cured with surgical excision, variations in atypical/anaplastic meningioma have a high recurrence rate and a poor prognosis. As a result, it is critical to develop novel therapeutic options for high-grade meningiomas. This review highlights the current histology of meningiomas, prevalent genetic and molecular changes, and the most extensively researched signaling pathways and therapies in meningiomas. It also reviews current clinical studies and novel meningioma treatments, including immunotherapy, microRNAs, cancer stem cell methods, and targeted interventions within the glycolysis pathway. Through the examination of the complex landscape of meningioma biology and the highlighting of promising therapeutic pathways, this review opens the way for future research efforts aimed at improving patient outcomes in this prevalent intracranial tumor entity.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Humanos , Meningioma/genética , Meningioma/patología , Meningioma/terapia , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/terapia , MicroARNs/genética , Inmunoterapia/métodos , Transducción de Señal
9.
Ups J Med Sci ; 1292024.
Artículo en Inglés | MEDLINE | ID: mdl-38571886

RESUMEN

Meningiomas, the most common primary brain tumors in adults, are often benign and curable by surgical resection. However, a subset is of higher grade, shows aggressive growth behavior as well as brain invasion, and often recurs even after several rounds of surgery. Increasing evidence suggests that tumor classification and grading primarily based on histopathology do not always accurately predict tumor aggressiveness and recurrence behavior. The underlying biology of aggressive treatment-resistant meningiomas and the impact of specific genetic aberrations present in these high-grade tumors is still only insufficiently understood. Therefore, an in-depth research into the biology of this tumor type is warranted. More recent studies based on large-scale molecular data such as whole exome/genome sequencing, DNA methylation sequencing, and RNA sequencing have provided new insights into the biology of meningiomas and have revealed new risk factors and prognostic subtypes. The most common genetic aberration in meningiomas is functional loss of NF2 and occurs in both low- and high-grade meningiomas, whereas NF2-wildtype meningiomas are enriched for recurrent mutations in TRAF7, KLF4, AKT1, PI3KCA, and SMO and are more frequently benign. Most meningioma mouse models are based on patient-derived xenografts and only recently have new genetically engineered mouse models of meningioma been developed that will aid in the systematic evaluation of specific mutations found in meningioma and their impact on tumor behavior. In this article, we review recent advances in the understanding of meningioma biology and classification and highlight the most common genetic mutations, as well as discuss new genetically engineered mouse models of meningioma.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Adulto , Humanos , Animales , Ratones , Meningioma/genética , Meningioma/patología , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/cirugía , Factor 4 Similar a Kruppel , Mutación , Pronóstico
10.
Neurosurg Rev ; 47(1): 136, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38561568

RESUMEN

This letter offers a nuanced evaluation of the recent study on single-cell transcriptome analysis of ECM-remodeling meningioma cells. While acknowledging the positive aspects, such as enhanced understanding of tumor heterogeneity and identification of potential therapeutic targets, it also highlights potential limitations, including challenges in data interpretation and validation.The focus on ECM-remodeling may inadvertently overshadow other critical aspects of tumor biology, necessitating a more holistic approach. The abstract concludes by emphasizing the importance of considering the broader context of tumor heterogeneity and microenvironmental influences in future research endeavors to improve clinical outcomes for patients with meningioma and other malignancies.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Humanos , Meningioma/genética , Meningioma/patología , Análisis de Expresión Génica de una Sola Célula , Matriz Extracelular/patología , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología
11.
Redox Biol ; 72: 103137, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38642502

RESUMEN

The oncogene Aurora kinase A (AURKA) has been implicated in various tumor, yet its role in meningioma remains unexplored. Recent studies have suggested a potential link between AURKA and ferroptosis, although the underlying mechanisms are unclear. This study presented evidence of AURKA upregulation in high grade meningioma and its ability to enhance malignant characteristics. We identified AURKA as a suppressor of erastin-induced ferroptosis in meningioma. Mechanistically, AURKA directly interacted with and phosphorylated kelch-like ECH-associated protein 1 (KEAP1), thereby activating nuclear factor erythroid 2 related factor 2 (NFE2L2/NRF2) and target genes transcription. Additionally, forkhead box protein M1 (FOXM1) facilitated the transcription of AURKA. Suppression of AURKA, in conjunction with erastin, yields significant enhancements in the prognosis of a murine model of meningioma. Our study elucidates an unidentified mechanism by which AURKA governs ferroptosis, and strongly suggests that the combination of AURKA inhibition and ferroptosis-inducing agents could potentially provide therapeutic benefits for meningioma treatment.


Asunto(s)
Aurora Quinasa A , Ferroptosis , Proteína Forkhead Box M1 , Meningioma , Factor 2 Relacionado con NF-E2 , Piperazinas , Ferroptosis/efectos de los fármacos , Ferroptosis/genética , Proteína Forkhead Box M1/metabolismo , Proteína Forkhead Box M1/genética , Aurora Quinasa A/metabolismo , Aurora Quinasa A/genética , Humanos , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Animales , Ratones , Meningioma/metabolismo , Meningioma/genética , Meningioma/patología , Piperazinas/farmacología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/genética , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/tratamiento farmacológico , Resistencia a Antineoplásicos/genética
12.
Zhonghua Bing Li Xue Za Zhi ; 53(5): 439-445, 2024 May 08.
Artículo en Chino | MEDLINE | ID: mdl-38678323

RESUMEN

Objective: To examine whether immunohistochemistry of methylthioadenosine phosphorylase (MTAP) and p16 could be used to predict the CDKN2A status in various brain tumors. Methods: A total of 118 cases of IDH-mutant astrocytomas, 16 IDH-wildtype glioblastoma, 17 polymorphic xanthoastrocytoma (PXA) and 20 meningiomas diagnosed at Xuanwu Hospital, Capital Medical University, Beijing, China from November 2017 to October 2023 were collected and analyzed. The CDKN2A status was detected by using fluorescence in situ hybridization or next-generation sequencing. Expression of MTAP and p16 proteins was detected with immunohistochemistry. The association of loss of MTAP/p16 expression with CDKN2A homozygous/heterozygous deletion was examined. Results: Among the 118 cases of IDH-mutant astrocytoma, 13 cases showed homozygous deletion of CDKN2A. All of them had no expression of MTAP while 9 cases had no expression of p16. Among the 16 cases of IDH wild-type glioblastoma, 6 cases showed homozygous deletion of CDKN2A. All 6 cases had no expression of MTAP, while 3 of these cases had no expression of p16 expression. Among the 17 PXA cases, 4 cases showed homozygous deletion of CDKN2A, and the expression of MTAP and p16 was also absent in these 4 cases. Among the 20 cases of meningiomas, 4 cases showed homozygous deletion of CDKN2A. Their expression of MTAP and p16 was also absent. Among the four types of brain tumors, MTAP was significantly correlated with CDKN2A homozygous deletion (P<0.05), with a sensitivity of 100%. However, it was only significantly correlated with the loss of heterozygosity (LOH) of CDKN2A in astrocytomas (P<0.001). P16 was associated with CDKN2A homozygous deletion in IDH-mutant astrocytoma and PXA (P<0.001), but not with the LOH of CDKN2A. Its sensitivity and specificity were lower than that of MTAP. Conclusions: MTAP could serve as a predictive surrogate for CDKN2A homozygous deletion in adult IDH-mutant astrocytoma, PXA, adult IDH-wildtype glioblastoma and meningioma. However, p16 could only be used in the first two tumor types, and its specificity and sensitivity are lower than that of MTAP.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Encefálicas , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Homocigoto , Purina-Nucleósido Fosforilasa , Humanos , Purina-Nucleósido Fosforilasa/genética , Purina-Nucleósido Fosforilasa/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética , Astrocitoma/genética , Astrocitoma/metabolismo , Meningioma/genética , Meningioma/metabolismo , Meningioma/patología , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Inmunohistoquímica , Hibridación Fluorescente in Situ , Eliminación de Gen , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/metabolismo , Mutación , Masculino , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Femenino , Adulto , Secuenciación de Nucleótidos de Alto Rendimiento
13.
Int J Mol Sci ; 25(8)2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38674001

RESUMEN

Medulloblastoma (MB) encompasses diverse subgroups, and leptomeningeal disease/metastasis (LMD) plays a substantial role in associated fatalities. Despite extensive exploration of canonical genes in MB, the molecular mechanisms underlying LMD and the involvement of the orthodenticle homeobox 2 (OTX2) gene, a key driver in aggressive MB Group 3, remain insufficiently understood. Recognizing OTX2's pivotal role, we investigated its potential as a catalyst for aggressive cellular behaviors, including migration, invasion, and metastasis. OTX2 overexpression heightened cell growth, motility, and polarization in Group 3 MB cells. Orthotopic implantation of OTX2-overexpressing cells in mice led to reduced median survival, accompanied by the development of spinal cord and brain metastases. Mechanistically, OTX2 acted as a transcriptional activator of the Mechanistic Target of Rapamycin (mTOR) gene's promoter and the mTORC2 signaling pathway, correlating with upregulated downstream genes that orchestrate cell motility and migration. Knockdown of mTOR mRNA mitigated OTX2-mediated enhancements in cell motility and polarization. Analysis of human MB tumor samples (N = 952) revealed a positive correlation between OTX2 and mTOR mRNA expression, emphasizing the clinical significance of OTX2's role in the mTORC2 pathway. Our results reveal that OTX2 governs the mTORC2 signaling pathway, instigating LMD in Group 3 MBs and offering insights into potential therapeutic avenues through mTORC2 inhibition.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Diana Mecanicista del Complejo 2 de la Rapamicina , Meduloblastoma , Neoplasias Meníngeas , Factores de Transcripción Otx , Animales , Femenino , Humanos , Masculino , Ratones , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Meduloblastoma/genética , Meduloblastoma/patología , Meduloblastoma/metabolismo , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/secundario , Factores de Transcripción Otx/metabolismo , Factores de Transcripción Otx/genética , Transducción de Señal
14.
Hum Pathol ; 148: 32-40, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38670237

RESUMEN

This study investigated the clinicopathological, immunohistochemical, and molecular features of primary leptomeningeal melanocytic neoplasms (LMNs). Twelve LMN cases were retrospectively reviewed. We performed Fluorescence in-situ hybridization (including a 4-probe FISH assay with CDKN2A and MYC assay) and Next-Generation sequencing analyses on available cases. Histologically, 2 tumours were classified as melanocytomas (MC), 2 as intermediate-grade melanocytomas (IMC), and 8 as leptomeningeal melanomas (LMM). Two rare cases of LMM were associated with large plaque-like blue nevus. One MC case was associated with Ota. Ten cases (83.3%) showed melanocytic cells with benign features diffusely proliferating within the meninges. The Ki-67 in three categories differed (MC 0-1%, IMC 0-3%, LMM 3-10%). 57.1% of LMM cases (4/7) were positive for FISH. Nine of 10 tumours harboured activating hotspot mutations in GNAQ, GNA11, or PLCB4. Additional mutations of EIF1AX, SF3B1, or BAP1 were found in 40%, 30%, and 10% of tumours, respectively. During the follow-up (median = 43 months), 5 LMM patients experienced recurrence and/or metastasis, 3 of them died of the disease and the other 2 are alive with the tumour. Our study is by far the first cohort of LMN cases tested by FISH. In addition to morphological indicators including necrosis and mitotic figures, using a combination of Ki-67 and FISH helps to differentiate between IMC and LMM, especially in LMM cases with less pleomorphic features. SF3B1 mutation is first described in 2 cases of plaque-type blue nevus associated with LMM. Patients with SF3B1 mutation might be related to poor prognosis in LMN.


Asunto(s)
Biomarcadores de Tumor , Inmunohistoquímica , Hibridación Fluorescente in Situ , Melanoma , Neoplasias Meníngeas , Mutación , Humanos , Masculino , Femenino , Persona de Mediana Edad , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Adulto , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/análisis , Melanoma/genética , Melanoma/patología , Estudios Retrospectivos , Anciano , Secuenciación de Nucleótidos de Alto Rendimiento , Adulto Joven , Adolescente , Análisis Mutacional de ADN
15.
J Neurooncol ; 168(3): 405-413, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38630387

RESUMEN

INTRODUCTION: Meningiomas are the most common primary brain and central nervous system tumors, accounting for approximately 40% of these tumors. The most important exams for the radiological study of meningiomas are computed tomography (CT) and magnetic resonance imaging (MRI). We aimed to analyze the radiological features of patients with meningioma related to the simultaneous presence of bilateral macronodular adrenocortical disease (BMAD), with or without pathogenic variants of ARMC5. METHODS: This study included 10 patients who were diagnosed with BMAD. All of them had a radiological diagnosis of expansive brain lesions suggestive of meningioma. All patients underwent brain MRI and a neuroradiolgist analyzed the following parameters: number, site and size of lesions; presence of calcification, edema and bone involvement. RESULTS AND DISCUSSION: Eight patients presented with germline variants of ARMC5; the other 2, did not. The most significant result was the incidence of multiple meningiomas, which was 50% in BMAD patients, whereas the average incidence described thus far is lower than 10%. Considering location, the 22 tumors in the BMAD patients were 5 convexity tumors (22.7%), and 17 skull base tumors (77.2%), the opposite proportion of patients without BMAD. A total of 40.9% of the tumors had calcification, 9% had cerebral edema and 40.9% had bone invasion due to hyperostosis. The literature describes meningioma calcification in 25% of patients, bone invasion by tumor hyperostosis in 20%, and cerebral edema in approximately 60%. CONCLUSION: Relevant results were found considering the rate of multiple meningiomas and tumor location. This finding reinforces the need for further research into the neurological effects caused by genetic variants of ARMC5 in patients with BMAD.


Asunto(s)
Imagen por Resonancia Magnética , Neoplasias Meníngeas , Meningioma , Humanos , Meningioma/genética , Meningioma/diagnóstico por imagen , Meningioma/patología , Femenino , Masculino , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/diagnóstico por imagen , Neoplasias Meníngeas/patología , Persona de Mediana Edad , Adulto , Anciano , Tomografía Computarizada por Rayos X , Proteínas del Dominio Armadillo
17.
Anticancer Drugs ; 35(6): 542-547, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38513197

RESUMEN

Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) are considered the first-line treatment for advanced or metastatic non-small cell lung cancer (NSCLC) patients harboring EGFR mutations. However, due to the rarity of cases, the response of EGFR-TKIs in patients harboring uncommon compound EGFR mutations still needs to be determined. Here, we demonstrated the case of a 47-year-old smoker diagnosed with leptomeningeal metastasis from NSCLC and had EGFR20 R776S, C797S, and EGFR21 L858R compound mutations. He was treated with furmonertinib combined with intrathecal pemetrexed chemotherapy following progression on osimertinib, which led to clinical improvement and successfully prolonged his survival by 3 months. Regrettably, the patient eventually died from heart disease. This report provides the first reported evidence for the use of furmonertinib and intrathecal pemetrexed chemotherapy in NSCLC patients harboring EGFR R776S/C797S/L858R mutations who progressed on previous EGFR-TKIs.


Asunto(s)
Acrilamidas , Compuestos de Anilina , Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma de Pulmón de Células no Pequeñas , Receptores ErbB , Neoplasias Pulmonares , Mutación , Pemetrexed , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Pemetrexed/administración & dosificación , Pemetrexed/uso terapéutico , Receptores ErbB/genética , Acrilamidas/administración & dosificación , Acrilamidas/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Compuestos de Anilina/administración & dosificación , Compuestos de Anilina/uso terapéutico , Masculino , Persona de Mediana Edad , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Inyecciones Espinales , Carcinomatosis Meníngea/tratamiento farmacológico , Carcinomatosis Meníngea/secundario , Carcinomatosis Meníngea/genética , Neoplasias Meníngeas/secundario , Neoplasias Meníngeas/tratamiento farmacológico , Neoplasias Meníngeas/genética , Indoles , Pirimidinas
18.
Neurosurg Rev ; 47(1): 118, 2024 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-38491247

RESUMEN

Meningiomas are the most common tumours that primarily arise in the central nervous system, but their intratumoural heterogeneity has not yet been thoroughly studied. We aimed to investigate the transcriptome characteristics and biological properties of ECM-remodeling meningioma cells. Single-cell RNA sequencing (ScRNA-seq) data from meningioma samples were acquired and used for analyses. We conducted comprehensive bioinformatics analyses, including screening for differentially expressed genes (DEGs), Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathway and Gene Ontology (GO) term enrichment analyses, Gene Set Enrichment Analysis (GSEA), protein-protein interaction (PPI) analysis, and copy number variation (CNV) analysis on single-cell sequencing data from meningiomas. Eighteen cell types, including six meningioma subtypes, were identified in the data. ECM-remodeling meningioma cells (MGCs) were mainly distributed in brain-tumour interface tissues. KEGG and GO enrichment analyses revealed that 908 DEGs were mainly related to cell adhesion, extracellular matrix organization, and ECM-receptor interaction. GSEA analysis demonstrated that homophilic cell adhesion via plasma membrane adhesion molecules was significantly enriched (NES = 2.375, P < 0.001). CNV analysis suggested that ECM-remodeling MGCs showed considerably lower average CNV scores. ECM-remodeling MGCs predominantly localized at the brain-tumour interface area and adhere stably to the basement membrane with a lower degree of malignancy. This study provides novel insights into the malignancy of meningiomas.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Humanos , Perfilación de la Expresión Génica , Meningioma/genética , Análisis de Expresión Génica de una Sola Célula , Variaciones en el Número de Copia de ADN , Neoplasias Meníngeas/genética
19.
BMC Cancer ; 24(1): 345, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38500077

RESUMEN

BACKGROUND: Meningioma, the most prevalent intracranial tumor, possesses a significant propensity for malignant transformation. Circular RNAs (circ-RNAs), a class of non-coding RNAs, have emerged as crucial players in tumorigenesis. This study explores the functional relevance of hsa_circ_0004872, a specific circ-RNA, in the context of meningioma. METHODS: Molecular structure and stability of hsa_circ_0004872 were elucidated through PCR identification. Meningioma cell proliferation and apoptosis were assessed using the CCK-8 assay and flow cytometry, respectively. Gene and protein expression were analyzed via qRT-PCR and western blot. Molecular interactions were confirmed through dual-luciferase reporter gene and RIP assays. RESULTS: Hsa_circ_0004872, derived from exons 2 to 4 of the host gene MAPK1, demonstrated enhanced stability compared to its host MAPK1. Clinical data described that hsa_circ_0004872 was reduced in meningioma tissues and cell lines, and negatively correlated to poor survival rate of meningioma patients. Overexpression of hsa_circ_0004872 exhibited inhibitory effects on cell proliferation and promotion of apoptosis in vitro. Subsequent investigations unveiled a direct interaction between hsa_circ_0004872 and miR-190a-3p, leading to the activation of the PI3K/AKT signaling pathway through targeting PTEN. Notably, miR-190a-3p silence accelerated the apoptosis and proliferation inhibition of meningioma cells by inactivating PTEN/PI3K/AKT signaling, while miR-190a-3p overexpression showed an opposite effect, which greatly reversed the anti-tumor effects of hsa_circ_0004872 overexpression. CONCLUSION: In summary, our findings highlighted the intricate role of hsa_circ_0004872 in meningioma, shedding light on the regulatory mechanisms involving circ-RNAs in tumor progression. This positions hsa_circ_0004872 as a potential key regulatory factor in meningioma with implications for future therapeutic interventions.


Asunto(s)
Neoplasias Meníngeas , Meningioma , MicroARNs , Humanos , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , Transformación Celular Neoplásica , Regulación Neoplásica de la Expresión Génica , Neoplasias Meníngeas/genética , Meningioma/genética , MicroARNs/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Fosfohidrolasa PTEN/genética , Transducción de Señal/genética
20.
J Neurooncol ; 167(3): 455-465, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38446374

RESUMEN

PURPOSE: Meningiomas are the most common type of brain tumors and are generally benign, but malignant atypical meningiomas and anaplastic meningiomas frequently recur with poor prognosis. The metabolism of meningiomas is little known, so few effective treatment options other than surgery and radiation are available, and the targets for treatment of recurrence are not well defined. The Aim of this paper is to find the therapeutic target. METHODS: The effects of bone morphogenetic protein (BMP) signal inhibitor (K02288) and upstream regulator Gremlin2 (GREM2) on meningioma's growth and senescence were examined. In brief, we examined as follows: 1) Proliferation assay by inhibiting BMP signaling. 2) Comprehensive analysis of forced expression GREM2.3) Correlation between GREM2 mRNA expression and proliferation marker in 87 of our clinical samples. 4) Enrichment analysis between GREM2 high/low expressed groups using RNA-seq data (42 cases) from the public database GREIN. 5) Changes in metabolites and senescence markers associated with BMP signal suppression. RESULTS: Inhibitors of BMP receptor (BMPR1A) and forced expression of GREM2 shifted tryptophan metabolism from kynurenine/quinolinic acid production to serotonin production in malignant meningiomas, reduced NAD + /NADH production, decreased gene cluster expression involved in oxidative phosphorylation, and caused decrease in ATP. Finally, malignant meningiomas underwent cellular senescence, decreased proliferation, and eventually formed psammoma bodies. Reanalyzed RNA-seq data of clinical samples obtained from GREIN showed that increased expression of GREM2 decreased the expression of genes involved in oxidative phosphorylation, similar to our experimental results. CONCLUSIONS: The GREM2-BMPR1A-tryptophan metabolic pathway in meningiomas is a potential new therapeutic target.


Asunto(s)
Proteínas Morfogenéticas Óseas , Calcinosis , Neoplasias Meníngeas , Meningioma , Transducción de Señal , Humanos , Meningioma/metabolismo , Meningioma/patología , Meningioma/genética , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/genética , Calcinosis/patología , Calcinosis/metabolismo , Calcinosis/genética , Proliferación Celular , Senescencia Celular , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA