Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Invest New Drugs ; 41(5): 768-776, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37603207

RESUMEN

BACKGROUND: Rearranged during transfection (RET) fusion-positive occurs in approximately 2% of non-small cell lung cancer (NSCLC). This mutation often predicts metastasis risk and poor prognosis, and current mainstream therapies provide limited patient benefit. Selective RET inhibitors Pralsetinib and Selpercatinib are targeted drugs approved by the US Food and Drug Administration for treating RET-mutated tumors. The phase I/II clinical trial results of their treatment of NSCLC have been published. However, the clinical effect of selective RET inhibitors on RET fusion-positive NSCLC remains controversial. Purpose Meta-analysis was performed to investigate the efficacy and safety of selective RET inhibitors in treating RET fusion-positive NSCLC. Methods Qualified literature was searched in Pubmed, Cochrane Library, Embase, and Web of Science. Outcomes included objective response rate (ORR), median progression-free survival (mPFS), disease control rate (DCR), intracranial ORR, and adverse events. Stata 15.1 software was used to analyze the data. Results A total of 8 studies were included in this meta-analysis. The combined results showed that the ORR of patients treated with selective RET inhibitors was 67% (95% confidence interval:0.64 to 0.70, P < 0.01), DCR was 92% (95%CI: 0.91-0.94, P < 0.01), the mPFS was 16.09 months (95%CI: 11.66-20.52, P < 0.01). In treated patients with RET mutation, the intracranial ORR was 86% (95%CI:0.74 ~ 0.96, P < 0.01). ORR in untreated patients was more effective than untreated patients [HR = 0.44 (95%CI: 0.35-0.56, P < 0.01)]. The major adverse events (grade 3-4) are neutropenia (13%) and anaemia (13%). Conclusions Selective RET inhibitors Pralsetinib and Selpercatinib have shown a good effect on RET fusion-positive NSCLC, with a low incidence of adverse events.

2.
Cancer Cell Int ; 23(1): 132, 2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37407973

RESUMEN

Glioma is the most common and aggressive primary malignant brain tumor. Circular RNAs (circRNAs) and RNA-binding proteins (RBPs) have been verified to mediate diverse biological behaviors in various human cancers. Therefore, the aim of this study was to explore a novel circRNA termed circGNB1 and elucidate relative molecular mechanism in functional phenotypes, which might be a potential prognostic biomarker and therapeutic approach for glioma. CircGNB1 was upregulated in glioma and closely associated with the low poor prognosis. Functional assays demonstrated that circGNB1 overexpression promoted glioma stem cells (GSCs) viability proliferation, invasion, and neurosphere formation. Mechanistically, circGNB1 upregulated the expression of oncogene XPR1 via sponging miR-515-5p and miR-582-3p. The following experiments proved XPR1 could promote the malignant phenotype of GSCs via upregulating IL6 expression and activating JAK2/STAT3 signaling. Moreover, the RNA binding protein IGF2BP3 could bind to and maintain the stability of circGNB1, thus promoting the effects of circGNB1 on GSCs. Our study reveals that circGNB1 plays a crucial role in promoting tumorigenesis and malignant progression in glioma, which provides a promising cancer biomarker.

3.
J Pathol ; 249(1): 26-38, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30953361

RESUMEN

Mesenchymal glioblastoma (GBM) is the most aggressive subtype of GBM. Our previous study found that neurotrophic factor prosaposin (PSAP) is highly expressed and secreted in glioma and can promote the growth of glioma. The role of PSAP in mesenchymal GBM is still unclear. In this study, bioinformatic analysis, western blotting and RT-qPCR were used to detect the expression of PSAP in different GBM subtypes. Human glioma cell lines and patient-derived glioma stem cells were studied in vitro and in vivo, revealing that mesenchymal GBM expressed and secreted the highest level of PSAP among four subtypes of GBM, and PSAP could promote GBM invasion and epithelial-mesenchymal transition (EMT)-like processes in vivo and in vitro. Bioinformatic analysis and western blotting showed that PSAP mainly played a regulatory role in GBM invasion and EMT-like processes via the TGF-ß1/Smad signaling pathway. In conclusion, the overexpression and secretion of PSAP may be an important factor causing the high invasiveness of mesenchymal GBM. PSAP is therefore a potential target for the treatment of mesenchymal GBM. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Transición Epitelial-Mesenquimal , Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Saposinas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular , Femenino , Glioblastoma/genética , Glioblastoma/patología , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Células Madre Neoplásicas/patología , Fosforilación , Saposinas/genética , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/genética , Células Tumorales Cultivadas
4.
Cancer Immunol Immunother ; 68(7): 1157-1169, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31197461

RESUMEN

Chordoma is difficult to eradicate due to high local recurrence rates. The immune microenvironment is closely associated with tumor prognosis; however, its role in skull base chordoma is unknown. The expression of Galectin-9 (Gal9) and tumor-infiltrating lymphocyte (TIL) markers was assessed by immunohistochemistry. Kaplan-Meier and multivariate Cox analyses were used to assessing local recurrence-free survival (LRFS) and overall survival (OS) of patients. MiR-455-5p was identified as a regulator of Gal9 expression. Immunopositivity for Gal9 was associated with tumor invasion (p = 0.019), Karnofsky performance status (KPS) score (p = 0.017), and total TIL count (p < 0.001); downregulation of miR-455-5p was correlated with tumor invasion (p = 0.017) and poor prognosis; and the T-cell immunoglobulin and mucin-domain 3 (TIM3)+ TIL count was associated with chordoma invasion (p = 0.010) and KPS score (p = 0.037). Furthermore, multivariate analysis indicated that only TIM3+ TIL density was an independent prognostic factor for LRFS (p = 0.010) and OS (p = 0.016). These results can be used to predict clinical outcome and provide a basis for immune therapy in skull base chordoma patients.


Asunto(s)
Cordoma/patología , Galectinas/genética , Linfocitos Infiltrantes de Tumor/inmunología , MicroARNs/metabolismo , Neoplasias de la Base del Cráneo/patología , Adolescente , Adulto , Anciano , Niño , Cordoma/genética , Cordoma/inmunología , Cordoma/mortalidad , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Galectinas/inmunología , Galectinas/metabolismo , Regulación Neoplásica de la Expresión Génica/inmunología , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Humanos , Recuento de Linfocitos , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/genética , Invasividad Neoplásica/inmunología , Pronóstico , Estudios Retrospectivos , Neoplasias de la Base del Cráneo/inmunología , Neoplasias de la Base del Cráneo/mortalidad , Análisis de Supervivencia , Adulto Joven
5.
Crit Rev Microbiol ; 45(2): 239-251, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30776938

RESUMEN

Epidemiological studies have shown that Helicobacter pylori (HP) infection is a risk factor for gastric cancer (GC). HP infection may induce the release of pro-inflammatory mediators, and abnormally increase the level of reactive oxygen species (ROS), nitric oxide (NO), and cytokines in mucosal epithelial cells of the stomach. However, the specific mechanism underlying the pathogenesis of HP-associated GC is still poorly understood. Recent studies have revealed that abnormal microRNA expression may affect the proliferation, differentiation, and apoptosis of mucosal epithelial cells of the stomach to further influence GC occurrence, development, and metastasis. Herein, we summarize the role of abnormal microRNAs in the regulation of HP-associated GC progression. Abnormal microRNA expression in HP-positive GC may be a biomarker for GC diagnosis, occurrence, and development as well as its targeted treatment and prognosis.


Asunto(s)
Infecciones por Helicobacter/genética , Helicobacter pylori/fisiología , MicroARNs/genética , Neoplasias Gástricas/genética , Animales , Apoptosis , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/fisiopatología , Humanos , MicroARNs/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/fisiopatología
6.
BMC Cancer ; 19(1): 594, 2019 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-31208357

RESUMEN

BACKGROUND: Secondary central nervous system involvement of non-Hodgkin's lymphoma (NHL) is rare and with poor prognosis, the most common pathological type is diffuse large B cell lymphoma (DLBCL). Although it can occur in any part of central nervous system, it rarely directly infiltrates the spinal cord or cauda equina. CASE PRESENTATION: We present the case of 64-year-old immunocompetent man with a worsening pain of waist and left lower extremity, accompanied by numbness and paresis of bilateral lower extremity for 20 days. His previous medical history included a resection of painless mass in the left groin in another hospital 7 months ago, and the pathological diagnosis was non-Hodgkin small B cell lymphoma. Gd-enhanced MRI and F-18 FDG PET-CT scan demonstrated multiple infiltrations in the cauda equina. During the operation, we removed as many as 11 subdural-extramedullary bean-size lesions involving multiple nerve roots. The paralysis of his left leg recovered rapidly after the operation. During the follow-up period of more than one year, he underwent standard R-CHOP chemical therapy, no evidence of recurrence was noted until the 13th month, the patient died because of intracranial relapse. CONCLUSIONS: Imaging examination is important in the diagnosis of multiple secondary cauda equina non-Hodgkin's lymphoma, and we highlight the significance of gadolinium-enhanced MRI and F-18 FDG-PET/CT in preoperative diagnosis as well as the previous history.


Asunto(s)
Cauda Equina/patología , Linfoma de Células B/patología , Neoplasias del Sistema Nervioso Periférico/secundario , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cauda Equina/diagnóstico por imagen , Medios de Contraste , Ciclofosfamida/uso terapéutico , Doxorrubicina/uso terapéutico , Encefalocele/mortalidad , Resultado Fatal , Estudios de Seguimiento , Gadolinio/química , Humanos , Linfoma de Células B/cirugía , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Neoplasias del Sistema Nervioso Periférico/diagnóstico por imagen , Neoplasias del Sistema Nervioso Periférico/tratamiento farmacológico , Neoplasias del Sistema Nervioso Periférico/cirugía , Tomografía Computarizada por Tomografía de Emisión de Positrones , Prednisona/uso terapéutico , Rituximab/uso terapéutico , Negativa del Paciente al Tratamiento , Vincristina/uso terapéutico
7.
J Neurooncol ; 132(2): 313-321, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28074324

RESUMEN

Giant pituitary adenomas (diameter >4 cm) are a challenge to treat, and there is no consensus on the optimal surgical strategy. We report here our experience in surgical management of these lesions. Adult patients with giant pituitary adenomas (n = 62; 54 non-functioning and eight hormone-secreting adenomas) who underwent surgical resection at our hospital from 2009 to 2015 were retrospectively reviewed. Surgical and clinical outcomes were analyzed. Single transsphenoidal and transcranial approaches were used in 43 (69.4%) and four (6.5%) patients, respectively. A combined transsphenoidal and transcranial approach was used in 13 patients (20.9%) and in two patients (3.2%), a transcranial procedure was followed 3 months later by a transsphenoidal approach. Greater than 90% resection was achieved in 47 cases (75.8%). During a mean follow-up period of 46.9 months, 49 patients (79%) showed improved visual impairment scores, while none experienced visual deterioration. There was no post-operative hemorrhage or mortality. A total of 27 patients (43.5%) received adjuvant medical and/or radiation therapy. At last follow-up, eight patients (12.9%) had recurrence. For giant pituitary adenoma, the transsphenoidal and transcranial approaches should be combined flexibly based on the characteristics of the tumor. In certain cases, a simultaneous combined approach can maximize tumor extirpation and lower the risk of swelling and bleeding of the residual tumor.


Asunto(s)
Adenoma/cirugía , Enanismo Hipofisario/cirugía , Procedimientos Neuroquirúrgicos/métodos , Nariz/cirugía , Cráneo/cirugía , Resultado del Tratamiento , Adenoma/diagnóstico por imagen , Adulto , Anciano , Enanismo Hipofisario/diagnóstico por imagen , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
8.
J Craniofac Surg ; 28(6): e554-e557, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28708652

RESUMEN

Skull base chordoma is a rare tumor arising from embryonic remnants of the notochord with invasive potential. Due to the destruction of osseous landmarks and invasion of surrounding structures, surgical resection is challenging. The authors explored the clinical value of a multimodal neuronavigation system in skull base chordoma resection using a neuroendoscope. Between January 2012 and January 2016, the authors utilized neuroendoscopy to excise skull base chordoma in 93 patients. The authors performed 45 operations assisted by multimodal neuronavigation (neuronavigation group) and 48 without intraoperative imaging guidance (control group). In the control group, 35 patients (73%) underwent gross total resection. In the neuronavigation group, all patients underwent gross total resection without radiographically identified bleeding. Only 1 patient (2%) in the neuronavigation group showed a temporary reduction in vision, which improved after symptomatic treatment. In contrast, there were 4 patients (8%) with postoperative complication, including 2 patients with intracranial hematoma and 2 with neurological deficits. Complication rates were higher than the neuronavigation group. In the follow-up period, 2 patients in the control group with subtotal resection had recurrence within 24 months, but without extracranial metastases. The multimodal neuronavigation system could contribute intraoperative real-time guidance for spatial relationships between lesions and adjacent neurovascular structures, as well as eroded and distorted anatomical landmarks through multiple image fusion and 3-dimensional reconstruction. It significantly improves surgical outcome and provides a new insight into the management of skull base chordomas.


Asunto(s)
Cordoma/cirugía , Neuroendoscopía/métodos , Neoplasias de la Base del Cráneo/cirugía , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Imagen Multimodal , Recurrencia Local de Neoplasia/cirugía , Neuroendoscopios , Neuroendoscopía/instrumentación , Neuronavegación/instrumentación , Neuronavegación/métodos , Base del Cráneo/cirugía , Tomografía Computarizada por Rayos X , Adulto Joven
9.
Acta Neurochir (Wien) ; 158(7): 1355-62, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27165299

RESUMEN

BACKGROUND: Susceptibility-weighted imaging (SWI) exploits susceptibility differences between tissues to enhance contrast in magnetic resonance imaging to enable the visualization of small blood vessels that are difficult to detect by other contrast agents. This study explored the value of SWI-based planning for neuronavigation-guided deep brain biopsies to reduce the incidence of post-surgical complications. METHODS: The cohort of 84 patients was divided into 41 biopsies performed aided by SWI (SWI group) and 43 biopsies based on conventional T1w-Gd-based imaging (T1w-Gd group). Biopsy targets were determined using magnetic resonance spectroscopy (MRS) before the operation, and the safest trajectory was selected based on preoperative images of blood vessels. RESULTS: Within 24 h of surgery, there was no radiographically identified bleeding, no blood extravasation and no clinical intracranial hypertension in the SWI group. Only one patient (2.5 %) with basal ganglia lymphoma developed transient hemiparesis after biopsy, who later recovered after undergoing symptomatic treatment. Complication rates in the SWI group were lower than in the T1w-Gd group, where a 7 % morbidity rate was encountered with one patient developing a permanent neurological deficit and two showing biopsy-associated hemorrhages. SWI imaging yielded a better visualization of subcortical vessels and deep-seated brain structures. CONCLUSIONS: SWI-based imaging revealed significantly better visualization of small-caliber vasculature that was not detectable on conventional T1w-Gd imaging, minimizing damage to the brain and reducing postoperative complications. Furthermore, MRS can contribute significantly to target selection to improve the yield of image-guided biopsies.


Asunto(s)
Neoplasias Encefálicas/cirugía , Encéfalo/cirugía , Imagen por Resonancia Magnética/métodos , Procedimientos Quirúrgicos Mínimamente Invasivos/métodos , Neuronavegación/métodos , Adulto , Anciano , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Femenino , Humanos , Biopsia Guiada por Imagen/efectos adversos , Biopsia Guiada por Imagen/métodos , Imagen por Resonancia Magnética/efectos adversos , Masculino , Persona de Mediana Edad , Procedimientos Quirúrgicos Mínimamente Invasivos/efectos adversos , Neuronavegación/efectos adversos , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/prevención & control
10.
J Biol Chem ; 288(3): 1634-42, 2013 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-23192351

RESUMEN

The forkhead box M1 (FoxM1) is a key transcription factor regulating multiple aspects of cell biology. Prior studies have shown that FoxM1 is overexpressed in a variety of human tumors, including brain tumor, and plays a critical role in cancer development and progression. In this study we found that FoxM1 was up-regulated by heat shock factor 1 (HSF1) under heat shock stress condition in multiple cell lines. Knockdown of HSF1 with HSF1 siRNA or inhibition of HSF1 with a HSF1 inhibitor abrogated heat shock-induced expression of FoxM1. Genetic deletion of HSF1 in mouse embryo fibroblast cells also abolished heat shock stress-induced FoxM1 expression. Moreover, we showed that HSF1 directly bound to FoxM1 promoter and increased FoxM1 promoter activity. Furthermore, we demonstrated that FoxM1 was required for the G(2)-M phase progression through regulating Cdc2, Cdc20, and Cdc25B under a mild heat shock stress but enhanced cell survival under lethal heat shock stress condition. Finally, in human glioblastoma specimens, FoxM1 overexpression correlated with elevated HSF1 expression. Our results indicate that FoxM1 is regulated by HSF1 and is critical for HSF1-mediated heat shock response. We demonstrated a novel mechanism of stress resistance controlled by HSF1 and a new HSF-FoxM1 connection that mediates cellular thermotolerance.


Asunto(s)
Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Respuesta al Choque Térmico/genética , Factores de Transcripción/genética , Animales , Línea Celular Tumoral , Supervivencia Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Fibroblastos/citología , Fibroblastos/metabolismo , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Técnicas de Silenciamiento del Gen , Glioblastoma/metabolismo , Glioblastoma/patología , Factores de Transcripción del Choque Térmico , Calor , Humanos , Ratones , Regiones Promotoras Genéticas , Unión Proteica , ARN Interferente Pequeño/genética , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo
11.
Biochem Pharmacol ; 226: 116380, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38945276

RESUMEN

GBM is the most threatening form of brain tumor. The advancement of GBM is propelled by the growth, infiltration, and movement of cancer cells. Understanding the underlying mechanisms and identifying new therapeutic agents are crucial for effective GBM treatment. Our research focused on examining the withhold influence of Enhydrin on the destructive activity of GBM cells, both in laboratory settings and within living organisms. By employing network pharmacology and bioinformatics analysis, we have determined that Jun serves as the gene of interest, and EMT as the critical signaling pathway. Mechanistically, Enhydrin inhibits the activity of the target gene Jun to increase the expression of Smad7, which is infinitively regulated by the transcription factor Jun, and as the inhibitory transcription factor, Smad7 can down-regulate TGF-ß1 and the subsequent Smad2/3 signaling pathway. Consequently, this whole process greatly hinders the EMT mechanism of GBM, leading to the notable decline in cell proliferation, invasion, and migration. In summary, our research shows that Enhydrin hinders EMT by focusing on the Jun/Smad7/TGF-ß1 signaling pathway, presenting a promising target for treating GBM. Moreover, Enhydrin demonstrates encouraging prospects as a new medication for GBM treatment.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Transducción de Señal , Proteína smad7 , Factor de Crecimiento Transformador beta1 , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Humanos , Proteína smad7/metabolismo , Proteína smad7/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Línea Celular Tumoral , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Ratones , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/fisiología , Ratones Desnudos , Fenotipo , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos
12.
Oncogene ; 43(36): 2679-2695, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39098847

RESUMEN

Exciting breakthroughs have been achieved in the field of glioblastoma with therapeutic interventions targeting specific ferroptosis targets. Nonetheless, the precise mechanisms through which circRNAs regulate the ferroptosis pathway have yet to be fully elucidated. Here we have identified a novel circRNA, circVPS8, which is highly expressed in glioblastoma. Our findings demonstrated that circVPS8 enhances glioma stem cells' viability, proliferation, sphere-forming ability, and stemness. Additionally, it inhibits ferroptosis in GSCs. In vivo, experiments further supported the promotion of glioblastoma growth by circVPS8. Mechanistically, circVPS8 acts as a scaffold, binding to both MKRN1 and SOX15, thus facilitating the ubiquitination of MKRN1 and subsequent degradation of SOX15. Due to competitive binding, the ubiquitination ability of MKRN1 towards HNF4A is reduced, leading to elevated HNF4A expression. Increased HNF4A expression, along with decreased SOX15 expression, synergistically inhibits ferroptosis in glioblastoma. Overall, our study highlights circVPS8 as a promising therapeutic target and provides valuable insights for clinically targeted therapy of glioblastoma.


Asunto(s)
Ferroptosis , Glioma , Factor Nuclear 4 del Hepatocito , Células Madre Neoplásicas , ARN Circular , Animales , Humanos , Ratones , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Ferroptosis/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Glioblastoma/genética , Glioblastoma/metabolismo , Glioma/patología , Glioma/genética , Glioma/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fenotipo , ARN Circular/genética , Factores de Transcripción SOXC/genética , Factores de Transcripción SOXC/metabolismo
13.
Phytomedicine ; 130: 155611, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-38776737

RESUMEN

BACKGROUND: Glioblastoma is the most malignant and prevalent primary human brain tumor, and the immunological microenvironment controlled by glioma stem cells is one of the essential elements contributing to its malignancy. The use of medications to ameliorate the tumor microenvironment may give a new approach for glioma treatment. METHODS: Glioma stem cells were separated from clinical patient-derived glioma samples for molecular research. Other studies, including CCK8, EdU, Transwell, and others, supported luteolin's ability to treat glioma progenitor cells. Network pharmacology and molecular docking models were used to study the drug target, and qRT-PCR, WB, and IF were used to evaluate the molecular mechanism. Intracranial xenografts were examined using HE and IHC, while macrophage polarization was examined using FC. RESULTS: We originally discovered that luteolin inhibits glioma stem cells. IL6 released by glioma stem cells is blocked during medication action and inhibits glioma stem cell proliferation and invasion via the IL6/STAT3 signaling pathway. Additionally, luteolin inhibits the secretion of TGFß1, affects the polarization function of macrophages in the microenvironment, inhibits the polarization of M2 macrophages in TAM, and further inhibits various functions of glioma stem cells by affecting the IL6/STAT3 signaling pathway, luteolin crosstalk TGFß1/SMAD3 signaling pathway, and so on. CONCLUSIONS: Through the suppression of the immunological microenvironment and inhibition of the IL6/STAT3 signaling pathway, our study determined the inhibitory effect of luteolin on glioma stem cells. This medication's dual inhibitory action, which has a significant negative impact on the glioma stem cells' malignant process, makes it both a viable anti-glioma medication and a candidate for targeted glioma microenvironment therapy.


Asunto(s)
Neoplasias Encefálicas , Proliferación Celular , Glioblastoma , Luteolina , Células Madre Neoplásicas , Factor de Transcripción STAT3 , Microambiente Tumoral , Luteolina/farmacología , Microambiente Tumoral/efectos de los fármacos , Humanos , Glioblastoma/tratamiento farmacológico , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Interleucina-6/metabolismo , Línea Celular Tumoral , Macrófagos/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Ratones , Simulación del Acoplamiento Molecular , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína smad3/metabolismo
14.
Phytomedicine ; 129: 155631, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38640858

RESUMEN

BACKGROUND: The utilization of Chinese medicine as an adjunctive therapy for cancer has recently gained significant attention. Ferroptosis, a newly regulated cell death process depending on the ferrous ions, has been proved to be participated in glioma stem cells inactivation. PURPOSE: We aim to study whether ginsenoside Rg5 exerted inhibitory effects on crucial aspects of glioma stem cells, including cell viability, tumor initiation, invasion, self-renewal ability, neurosphere formation, and stemness. METHODS: Through comprehensive sequencing analysis, we identified a compelling association between ginsenoside Rg5 and the ferroptosis pathway, which was further validated through subsequent experiments demonstrating its ability to activate this pathway. RESULTS: To elucidate the precise molecular targets affected by ginsenoside Rg5 in gliomas, we conducted an intersection analysis between differentially expressed genes obtained from sequencing and a database-predicted list of transcription factors and potential targets of ginsenoside Rg5. This rigorous approach led us to unequivocally confirm NR3C1 (Nuclear Receptor Subfamily 3 Group C Member 1) as a direct target of ginsenoside Rg5, a finding consistently supported by subsequent experimental investigations. Moreover, we uncovered NR3C1's capacity to transcriptionally regulate ferroptosis -related genes HSPB1 and NCOA4. Strikingly, ginsenoside Rg5 induced notable alterations in the expression levels of both HSPB1 (Heat Shock Protein Family B Member 1) and NCOA4 (Nuclear Receptor Coactivator 4). Finally, our intracranial xenograft assays served to reaffirm the inhibitory effect of ginsenoside Rg5 on the malignant progression of glioblastoma. CONCLUSION: These collective findings strongly suggest that ginsenoside Rg5 hampers glioblastoma progression by activating ferroptosis through NR3C1, which subsequently modulates HSPB1 and NCOA4. Importantly, this novel therapeutic direction holds promise for advancing the treatment of glioblastoma.


Asunto(s)
Ferroptosis , Ginsenósidos , Glioblastoma , Ginsenósidos/farmacología , Ferroptosis/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Humanos , Animales , Línea Celular Tumoral , Coactivadores de Receptor Nuclear/metabolismo , Ratones , Ratones Desnudos , Chaperonas Moleculares/metabolismo , Proteínas de Choque Térmico/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Antineoplásicos Fitogénicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico
15.
Biomed Pharmacother ; 176: 116847, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38823277

RESUMEN

Luteolin, a monomeric substance, is a natural product of the Brucea javanica (BJ) plant. Brucea javanica oil emulsion injection (BJOEI) is a proprietary Chinese medicine purified from BJ that is widely used clinically as an anti-tumor treatment. Although a growing body of research suggests that luteolin and BJOEI have anti-tumor effects, the molecular mechanism of action has not been fully elucidated. In this study, through molecular docking technology, we found that luteolin can interact directly with GPSM2 and regulate the FoxO signaling pathway through GPSM2. In addition, the inhibitory effect of luteolin on colon adenocarcinoma (COAD) cells was found to be offset by knockdown of GPSM2. In contrast, the anti-proliferative effects of luteolin could be notably reversed by overexpression of GPSM2. The results reveal that GPSM2 is crucial in luteolin-mediated anti-proliferative effects. The mediation of anti-proliferative effects by GPSM2 has also been indirectly demonstrated in RKO and SW480 xenograft mice models. In addition, we verified that BJOEI inhibits the progression of COAD by mediating GPSM2 and regulating the FoxO signaling pathway. We also found that BJOEI achieved a better anti-tumor effect when combined with fluorouracil injection. Collectively, our data show that the anti-tumor effects of BJOEI and luteolin on COAD are GPSM2-dependent and downregulating the expression of GPSM2 to regulate the FoxO signaling pathway may be an effective way to treat COAD.


Asunto(s)
Adenocarcinoma , Proliferación Celular , Neoplasias del Colon , Fluorouracilo , Luteolina , Ratones Desnudos , Luteolina/farmacología , Humanos , Animales , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Neoplasias del Colon/metabolismo , Fluorouracilo/farmacología , Línea Celular Tumoral , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Adenocarcinoma/metabolismo , Proliferación Celular/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos BALB C , Transducción de Señal/efectos de los fármacos , Ratones , Productos Biológicos/farmacología , Productos Biológicos/aislamiento & purificación , Productos Biológicos/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Simulación del Acoplamiento Molecular
16.
Aging (Albany NY) ; 15(16): 8237-8257, 2023 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-37606960

RESUMEN

BACKGROUND: Since December 2019, Coronavirus disease 2019 (COVID-19) induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in significant morbidity and mortality worldwide. There is an increased risk of ischemic stroke (IS) associated with COVID-19. However, few studies have been reported to explain the potential correlation between COVID-19 and IS. METHODS: We investigated the relationship and relevant mechanisms between COVID-19 and IS using single-cell RNA sequencing and multiple bioinformatics approaches. RESULTS: By intersecting differentially expressed genes and WGCNA critical module genes, we obtained 73 COVID-19-related IS genes. According to the KEGG pathway analysis, the COVID-19-related IS disease genes were significantly enriched in the hematopoietic cell lineage pathway, ribosome pathway, COVID-19 pathway and primary immunodeficiency pathway. Finally, three genes associated with immunity (B4GALT5, CRISPLD2, F5) and two genes associated with ferroptosis (ACSL1, CREB5) were identified up-regulated in COVID-19-related IS. Significantly, it was found that all five genes were highly expressed in monocytes by single cell RNA sequencing. CONCLUSION: We believe these genes (B4GALT5, CRISPLD2, F5, ACSL1, CREB5) may regulate the immune response and ferroptosis of multiple immune cells, mainly including monocytes, which may contribute to the development of COVID-19-related IS. In addition, these genes may be potential targets for the treatment of COVID-19-related IS.


Asunto(s)
COVID-19 , Ferroptosis , Accidente Cerebrovascular Isquémico , Humanos , SARS-CoV-2 , Biomarcadores , Biología Computacional , Análisis de Secuencia de ARN
17.
Cell Death Dis ; 14(1): 23, 2023 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-36635261

RESUMEN

Glioma is the most aggressive and common malignant neoplasms in human brain tumors. Numerous studies have showed that glioma stem cells (GSCs)drive the malignant progression of gliomas. Recent studies have revealed that circRNAs can maintain stemness and promote malignant progression of glioma stem cells. We used bioinformatics analysis to identify circRNAs and potential RNA-binding proteins (RBPs) in glioma. qRT-PCR, western blotting, RNA FISH, RNA pull-down, RNA immunoprecipitation assay, ChIP, immunohistochemistry, and immunofluorescence methods were used to quantified the expression of circNCAPG, U2AF65, RREB1 and TGF-ß1, and the underlying mechanisms between them. MTS, EDU, neurosphere formation, limiting dilution neurosphere formation and transwell assays examined the proliferation and invasive capability of GSCs, respectively. We identified a novel circRNA named circNCAPG was overexpressed and indicated the poor prognosis in glioma patients. Upregulating circNCAPG promoted the malignant progression of GSCs. RNA binding protein U2AF65 could stabilize circNCAPG by direct binding. Mechanically, circNCAPG interacted with and stabilized RREB1, as well as stimulated RREB1 nuclear translocation to activate TGF-ß1 signaling pathway. Furthermore, RREB1 transcriptionally upregulated U2AF65 expression to improve the stability of circNCAPG in GSCs, which established a feedback loop involving U2AF65, circNCAPG and RREB1. Since circRNA is more stable than mRNA and can execute its function continuously, targeting circNCAPG in glioma may be a novel promising therapeutic.


Asunto(s)
Neoplasias Encefálicas , Glioma , MicroARNs , ARN Circular , Humanos , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/genética , Proteínas de Unión al ADN/metabolismo , Retroalimentación , Regulación Neoplásica de la Expresión Génica , Glioma/patología , MicroARNs/genética , Células Madre Neoplásicas/metabolismo , ARN Circular/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Empalme U2AF/genética , Factor de Empalme U2AF/metabolismo
18.
Oncogene ; 41(26): 3461-3473, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35637250

RESUMEN

Glioblastoma multiforme (GBM) is the most lethal primary tumor with active neovascularization in the central nervous system. Studying the novel molecular mechanisms of GBM angiogenesis is very important. The glioblastoma-associated microglia (GAM) M2 polarization was constructed, and microglia-derived exosomes (MDEs) were isolated to co-culture with human brain microvessel endothelial cells (hBMECs). CircRNA sequence and molecular biological experiments were used to detect the expression levels and regulation functions among circKIF18A, FOXC2, ITGB3, CXCR4, DLL4 and the PI3K/AKT signaling. The functional effects of silencing or overexpression of these molecules were evaluated in hBMECs viability, invasion, and tube formation in vitro and tumorigenicity in vivo. M2 microglia polarization is positively correlated with microvessels' density in GBM patients. M2 GAM can promote the angiogenesis of GBM via transporting exosomal circKIF18A into hBMECs. Mechanistically, circKIF18A can bind to, maintain the stability and nuclear translocation of FOXC2 in hBMECs. Furtherly, as a transcription factor, FOXC2 can directly bind to the promoter of ITGB3, CXCR4, and DLL4 and upregulate their expressions. Besides, FOXC2 can also activate the PI3K/AKT signaling and promote the angiogenesis of GBM. Our study identified a novel molecular mechanism for M2 GAM-derived exosomal circKIF18A participating in GBM angiogenesis via targeting FOXC2. This may provide a novel treatment target to improve the outcomes for anti-angiogenic therapies in GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Células Endoteliales/metabolismo , Factores de Transcripción Forkhead/metabolismo , Glioblastoma/patología , Humanos , Microglía/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo
19.
Cell Death Dis ; 13(7): 645, 2022 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-35871061

RESUMEN

Glioblastoma multiforme (GBM) is the most lethal type of craniocerebral gliomas. Glioma stem cells (GSCs) are fundamental reasons for the malignancy and recurrence of GBM. Revealing the critical mechanism within GSCs' self-renewal ability is essential. Our study found a novel circular RNA (circRPPH1) that was up-regulated in GSCs and correlated with poor survival. The effect of circRPPH1 on the malignant phenotype and self-renewal of GSCs was detected in vitro and in vivo. Mechanistically, UPF1 can bind to circRPPH1 and maintain its stability. Therefore, more existing circRPPH1 can interact with transcription factor ATF3 to further transcribe UPF1 and Nestin expression. It formed a feedback loop to keep a stable stream for stemness biomarker Nestin to strengthen tumorigenesis of GSCs continually. Besides, ATF3 can activate the TGF-ß signaling to drive GSCs for tumorigenesis. Knocking down the expression of circRPPH1 significantly inhibited the proliferation and clonogenicity of GSCs both in vitro and in vivo. The overexpression of circRPPH1 enhanced the self-renewal of GSCs. Our findings suggest that UPF1/circRPPH1/ATF3 maintains the potential self-renewal of GSCs through interacting with RNA-binding protein and activating the TGF-ß signal pathway. Breaking the feedback loop against self-renewing GSCs may represent a novel therapeutic target in GBM treatment.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Factor de Transcripción Activador 3 , Neoplasias Encefálicas/patología , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular/genética , Transformación Celular Neoplásica/metabolismo , Retroalimentación , Glioblastoma/patología , Glioma/genética , Humanos , Células Madre Neoplásicas/metabolismo , Nestina/metabolismo , Fenotipo , ARN Helicasas/genética , ARN Helicasas/metabolismo , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
20.
Front Oncol ; 11: 699933, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34408982

RESUMEN

Glioblastoma (GBM) is a common and refractory subtype of high-grade glioma with a poor prognosis. The epithelial-mesenchymal transition (EMT) is an important cause of enhanced glioblastoma invasiveness and tumor recurrence. Our previous study found that retinoic acid receptor-related orphan receptor A (RORA) is a nuclear receptor and plays an important role in inhibiting proliferation and tumorigenesis of glioma. We further confirmed RORA was downregulated in GBM. Thus, we determined whether RORA was involved in the migration, invasion, and EMT of GBM. Human GBM cell lines, U87 and T98G, and patient-derived glioma stem cells (GSCs), GSC2C and GSC4D, were used for in vitro and in vivo experiments. The expressions of RORA, CASC2, and EIF4A3 in GBM cells and GSCs were detected by RT-qPCR and western blotting. The biological effects of RORA, CASC2, and EIF4A3 on GBM migration, invasion, and EMT were evaluated using the migration assay, transwell assay, immunofluorescence staining, and xenograft experiments. We found that RORA inhibited the migration, invasion, and EMT of GBM. CASC2 could bind to, maintain the stability, and promote the nuclear translocation of RORA protein. EIF4A3 could downregulate CASC2 expression via inducing its cleavage, while RORA transcriptionally inhibited EIF4A3 expression, which formed a feedback loop among EIF4A3/CASC2/RORA. Moreover, gene set enrichment analysis (GSEA) and in vitro and in vivo experiments showed RORA inhibited the aggressiveness of GBM by negatively regulating the TGF-ß1/Smad signaling pathway. Therefore, The EIF4A3/CASC2/RORA feedback loop regulated TGF-ß1/Smad signaling pathway might become a promising therapeutic strategy for GBM treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA