Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Carcinog ; 63(7): 1235-1247, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38517048

RESUMEN

Glioblastoma (GBM) is the most aggressive brain tumor type with worse clinical outcome due to the hallmarks of strong invasiveness, high rate of recurrence, and therapeutic resistance to temozolomide (TMZ), the first-line drug for GBM, representing a major challenge for successful GBM therapeutics. Understanding the underlying mechanisms that drive GBM progression will shed novel insight into therapeutic strategies. Receptor-type tyrosine-protein phosphatase S (PTPRS) is a frequently mutated gene in human cancers, including GBM. Its role in GBM has not yet been clarified. Here, inactivating PTPRS mutation or deficiency was frequently found in GBM, and deficiency in PTPRS significantly induced defects in the G2M checkpoint and limited GBM cells proliferation, leading to potent resistance to TMZ treatment in vitro and in vivo. Surprisingly, loss of PTPRS triggered an unexpected mesenchymal phenotype that markedly enhances the migratory capabilities of GBM cells through upregulating numerous matrix metalloproteinases via MAPK-MEK-ERK signaling. Therefore, this work provides a therapeutic window for precisely excluding PTPRS-mutated patients who do not respond to TMZ.


Asunto(s)
Antineoplásicos Alquilantes , Neoplasias Encefálicas , Proliferación Celular , Resistencia a Antineoplásicos , Glioblastoma , Temozolomida , Temozolomida/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ratones , Movimiento Celular/efectos de los fármacos , Mutación , Ensayos Antitumor por Modelo de Xenoinjerto , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo
2.
Mol Divers ; 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39012564

RESUMEN

Prolactinoma was the most common functional pituitary neuroendocrine tumor tissue type, which was caused by excessive proliferation of pituitary prolactin (PRL) cells. Drug therapy of dopamine receptor agonists was generally considered as the prior treatment for prolactinoma patients. However, there were still prolactinoma patients who were resistant to dopamine agonists. Studies have been reported that paeoniflorin can inhibit the secretion of PRL in prolactinoma cells lacking dopamine D2 receptor (D2R) expression, and paeoniflorin can be metabolized into albiflorin by intestinal flora in rats. The effect of albiflorin on prolactinoma has not been reported yet. In this study, network pharmacology was used to analyze the mechanism of paeoniflorin and its metabolite albiflorin as multi-target therapy for prolactinoma, and the experimental verification was carried out. In order to clarify the complex relationship among paeoniflorin, albiflorin and prolactinoma, we constructed a component-target-disease network, and further constructed interaction network, MMP9, EGFR, FGF2, FGFR1 and LGALS3 were screened as the core targets. Kyoto encyclopedia of genes and genomes (KEGG) analysis showed that paeoniflorin and albiflorin may be involved in various pathways in the treatment of prolactinoma, included relaxin signaling pathway and PI3K-Akt signaling pathway. Molecular docking analysis showed that paeoniflorin and albiflorin had good binding activity with MMP9. Western blotting results showed that paeoniflorin and albiflorin could significantly reduce the expression of MMP9, and ELISA results showed that paeoniflorin and albiflorin could significantly reduce the concentration of PRL in GH3 cells, and the reduce degree of albiflorin was stronger than paeoniflorin at 50 µM, which indicated that albiflorin might be a potential drug to treat prolactinoma, which can regulate prolactinoma through MMP9 and reduce the concentration of PRL. Our study provided a new therapeutic strategy for prolactinoma.

3.
Cell Biol Int ; 46(11): 1927-1936, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35971755

RESUMEN

Microglia is the major cellular component of glioma mass that promotes glioma growth, invasion, and chemoresistance by releasing inflammatory factors. Sterile alpha and HEAT/Armadillo motif (SARM), a member of the Toll-interleukin-1 receptor (TIR) domain-containing adaptor family, is primarily expressed in the central nervous system. However, the role of SARM in glioma is still undefined. In the present work, we examined the function of SARM in microglial polarization and glioma progression. Our results showed that forced the expression of SARM in GL261 glioma cells inhibited tumor growth, and reduced interleukin (IL)-6 secretion in conditioned media. Silencing of SARM in microglia cells inhibited IL-4-induced M2 polarization, enhanced lipopolysaccharide -induced M1 microglial polarization. Furthermore, overexpression of SARM increased the migration of microglia cells upon TGFß stimulation. These data suggested that SARM is involved in neuro-inflammation and microglia activation. In summary, this study provides novel insight into the mechanisms of microglial polarization.


Asunto(s)
Glioma , Microglía , Medios de Cultivo Condicionados , Glioma/metabolismo , Humanos , Interleucina-4/metabolismo , Lipopolisacáridos/farmacología , Microglía/metabolismo , Receptores de Interleucina-1/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
4.
Cell Commun Signal ; 18(1): 22, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32046730

RESUMEN

BACKGROUND: Glioblastoma and Alzheimer's disease (AD) are the most common and devastating diseases in the central nervous system. The dysfunction of Presenilin1 is the main reason for AD pathogenesis. However, the molecular function of Presenilin1 and its relative mechanism in glioblastoma remain unclear. METHODS: Expression of presenilin1 in glioma was determined by IHC. CCK-8, colony formation, Flow cytometry, Edu staining were utilized to evaluate functions of presenilin1 on glioblastoma proliferation. The mechanism of above process was assessed by Western blotting and cell immunofluorescence. Mouse transplanting glioblastoma model and micro-MRI detection were used to verified presenilin1 function in vivo. RESULTS: In this study, we found that all grades of glioma maintained relatively low Presenilin1 expression and that the expression of Presenilin1 in high-grade glioma was significantly lower than that in low-grade glioma. Moreover, the Presenilin1 level had a positive correlation with glioma and glioblastoma patient prognosis. Next, we determined that Presenilin1 inhibited the growth and proliferation of glioblastoma cells by downregulating CDK6, C-myc and Cyclin D1 to arrest the cell cycle at the G1/S phase. Mechanistically, Presenilin1 promoted the direct phosphorylation of ß-catenin at the 45 site and indirect phosphorylation at the 33/37/41 site, then decreased the stabilized part of ß-catenin and hindered its translocation from the cytoplasm to the nucleus. Furthermore, we found that Presenilin1 downregulation clearly accelerated the growth of subcutaneous glioblastoma, and Presenilin1 overexpression significantly repressed the subcutaneous and intracranial transplantation of glioblastoma by hindering ß-catenin-dependent cell proliferation. CONCLUSION: Our data implicate the antiproliferative effect of Presenilin1 in glioblastoma by suppressing Wnt/ß-catenin signaling, which may provide a novel therapeutic agent for glioblastoma. Video Abstract.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Glioblastoma/metabolismo , Glioblastoma/patología , Presenilina-1/metabolismo , Vía de Señalización Wnt , Animales , Línea Celular Tumoral , Proliferación Celular , Ciclina D1/metabolismo , Regulación hacia Abajo , Fase G1 , Humanos , Ratones Desnudos , Fosforilación , Pronóstico , Proteolisis , Proteínas Proto-Oncogénicas c-myc/metabolismo , Fase S , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/metabolismo
5.
Neurochem Res ; 44(7): 1690-1702, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31004260

RESUMEN

Trigeminal neuralgia (TN) is a type of chronic neuropathic pain that is caused by peripheral nerve lesions that result from various conditions, including the compression of vessels, tumors and viral infections. MicroRNAs (miRs) are increasingly recognized as potential regulators of neuropathic pain. Previous evidence has demonstrated that miR-195 is involved in neuropathic pain, but the mechanism remains unclear. To investigate the pathophysiological role of miR-195 and Shh signaling in TN, persistent facial pain was induced by infraorbital nerve chronic constriction injury (CCI-IoN), and facial pain responses were evaluated by Von Frey hairs. qPCR and Western blotting were used to determine the relative expression of miR-195 and Patched1, the major receptor of the Sonic Hedgehog (Shh) signaling pathway, in the caudal brain stem at distinct time points after CCI-IoN. Here, we found that the expression of miR-195 was increased in a rat model of CCI-IoN. In contrast, the expression of Patched1 decreased significantly. Luciferase assays confirmed the binding of miR-195 to Patched1. In addition, the overexpression of miR-195 by an intracerebroventricular (i.c.v) administration of LV-miR-195 aggravated facial pain development, and this was reversed by upregulating the expression of Patched1. These results suggest that miR-195 is involved in the development of TN by targeting Patched1 in the Shh signaling pathway, thus regulating extracellular glutamate.


Asunto(s)
Proteínas Hedgehog/metabolismo , MicroARNs/fisiología , Receptor Patched-1/metabolismo , Transducción de Señal/fisiología , Neuralgia del Trigémino/fisiopatología , Animales , Regulación hacia Abajo , Ácido Glutámico/líquido cefalorraquídeo , Ácido Glutámico/metabolismo , Infusiones Intraventriculares , Lentivirus/genética , Masculino , MicroARNs/administración & dosificación , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Receptor Patched-1/genética , Ratas Sprague-Dawley
6.
BMC Neurol ; 19(1): 217, 2019 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-31481028

RESUMEN

BACKGROUND: Microvascular decompression (MVD) is a type of neurosurgery used to treat trigeminal neuralgia (TN) caused by the vertebrobasilar contact/compression. The surgery is not risk-free, however; it may cause recurrent facial pain or other side-effects. The objective of this study was to assess the long-term pain relief and the complications of MVD surgery for the vertebrobasilar compression treatment. METHODS: Twenty-three patients with TN compressed by the vertebra-basilar artery (VBA) were treated with MVD. Teflon felt was placed between the brain stem and the offending artery to mobilize the artery towards the skull base and the clivus. The Barrow Neurological Institute (BNI) Pain Intensity Scale score was used to assess pre- and post-surgical pains. RESULTS: Of 23 patients with pre-operative BNI IV to V, 19 patients (83%) were pain-free after surgery. Four patients experienced transient partial pain relief with BNI II-III, and 3 of them (13%) were completely pain-free within 3 months. The success rate was 96%. Three patients (13%) had pain recurrences, and one received a second MVD surgery for pain relief during the period of follow-up. Four patients suffered from TN hypesthesia, and only 2 patients (8.6%) had permanent facial hypesthesia, while one patient (4.3%) developed a gradual hearing loss after surgery. CONCLUSIONS: While our success rate of immediate pain relief after surgery was comparable with some reports, the percentage of patients who had pain recurrences was lower, and cases who had permanent facial hypesthesia or developed a gradual hearing loss were fewer after MVD surgery. Our rate of transient complications was higher, and the postoperative pain relief seemed unusually delayed. Our study indicates that MVD is an effective, reliable, and safe neurosurgery for treatment of TN compressed by the VBA albeit our small sample size. Failure of treatment and recurrence of the disease as well as complications could be minimized by preventing displacement of the Teflon implant and extraneous Teflon touching the trigeminal nerves.


Asunto(s)
Dolor Facial/etiología , Hipoestesia/etiología , Cirugía para Descompresión Microvascular/métodos , Neuralgia del Trigémino/cirugía , Anciano , Arteria Basilar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Dimensión del Dolor , Recurrencia , Resultado del Tratamiento , Nervio Trigémino
7.
Future Oncol ; 15(32): 3723-3738, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31650850

RESUMEN

Aim: Apocynum venetum polyphenol (AVP) was used in in vitro glioma cells culture to prove the growth inhibitory effect of AVP on human U87 glioma cells via NF-κB pathway. Materials & methods: The MTT assay, DAPI morphology, quantitative PCR and western blot experiments were used for determination in vitro. Results & conclusion: AVP can also induce U87 cancer cells apoptosis illustrated by DAPI morphology. AVP could enhance the mRNA and protein expression of IκB-α, TNF-α, TRAIL, caspase-3 and caspase-9 in U87 cancer cells and reduce those of NF-κBp65, cIAP-1, cIAP-2, TGF-ß2, CyclinD1, VEGF and IL-8. After ammonium pyrrolidine dithiocarbamate (PDTC) treatment, the NF-κBp65 expression was reduced in U87 cells, and AVP could raise these effects. The results of HPLC indicate that AVP mainly contains six constituents. The growth inhibitory effects of AVP on U87 glioma cells are predominantly from these natural active constituents.


Asunto(s)
Antineoplásicos/farmacología , Apocynum/química , Apoptosis/efectos de los fármacos , Glioma/patología , Polifenoles/farmacología , Factor de Transcripción ReIA/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/metabolismo , Humanos , Polifenoles/química , Polifenoles/aislamiento & purificación , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/genética
8.
Med Sci Monit ; 25: 7652-7659, 2019 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-31604907

RESUMEN

BACKGROUND As a kind of benign tumor, pituitary adenomas have attracted increasing attention from researchers. The plasmacytoma variant translocation 1 (PVT1) is a molecule in the lncRNA family protein that has been proven to play critical roles in many cancers; however, no study has explored the special biological roles of PVT1 in pituitary adenoma. MATERIAL AND METHODS The qRT-PCR assay was conducted to evaluate PVT1 expressions in various cell lines and tissues. Loss of function assays were carried out to detect the influence of silenced PVT1 on the proliferation, migration, and epithelial-mesenchymal transition (EMT) of pituitary adenoma cells. Western blotting was used to identify correlation between ß-catenin and PVT1. RESULTS The PVT1 expressions were significantly enhanced in tissues of pituitary adenoma and cancer cells. Cell migration and proliferation were inhibited when the PVT1 gene was knocked down. Knockdown of PVT1 repressed the migration and EMT of pituitary adenoma cells. The PVT1 downregulation obviously blocked Wnt/ß-catenin signaling pathway activity. PVT1 aggravated progression of pituitary adenoma through initiating the Wnt/ß-catenin signaling pathway. CONCLUSIONS PVT1 exerts an oncogenic role through activating Wnt/ß-catenin signaling in pituitary adenoma cells. The present results may provide a potential therapeutic target or approach for treating pituitary adenomas.


Asunto(s)
Adenoma/genética , Movimiento Celular/genética , Ciclina D1/metabolismo , Transición Epitelial-Mesenquimal/genética , Neoplasias Hipofisarias/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Largo no Codificante/metabolismo , beta Catenina/metabolismo , Adenoma/patología , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hipofisarias/patología , ARN Largo no Codificante/genética , ARN Interferente Pequeño/metabolismo , Vía de Señalización Wnt
10.
Int J Neurosci ; 126(1): 1-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26000816

RESUMEN

The control of malignant glioma cell cycle by microRNAs (miRNAs) is well established. The deregulation of miRNAs in glioma may contribute to tumor proliferation by directly targeting the critical cell-cycle regulators. Tumor suppressive miRNAs inhibit cell cycle through repressing the expression of positive cell-cycle regulators. However, oncogenic miRNAs promote the cell-cycle progression by targeting cell-cycle negative regulators. Recent studies have identified that transcription factors had involved in the expression of miRNAs. Transcription factors and miRNAs are implicated in regulatory network of glioma cell cycle, the deregulation of these transcription factors might be a cause of the deregulation of miRNAs. Abnormal versions of miRNAs have been implicated in the cell cycle of glioma. Based on those, miRNAs are excellent biomarker candidates and potential targets for therapeutic intervention in glioma.


Asunto(s)
Ciclo Celular/genética , Neoplasias del Sistema Nervioso Central/genética , Glioma/genética , MicroARNs/genética , ARN Neoplásico/genética , Biomarcadores de Tumor , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Neoplasias del Sistema Nervioso Central/patología , Replicación del ADN , ADN de Neoplasias/biosíntesis , ADN de Neoplasias/genética , Fase G1/genética , Glioma/patología , Humanos , FN-kappa B/fisiología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas c-myc/fisiología , Fase S/genética , Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/genética
11.
Mol Cancer ; 14: 21, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25644759

RESUMEN

BACKGROUND: The poor prognosis and minimally successful treatments of malignant glioma indicate a challenge to identify new therapeutic targets which impact glioma progression. Neurotensin (NTS) and its high affinity receptor (NTSR1) overexpression induces neoplastic growth and predicts the poor prognosis in various malignancies. Whether NTS can promote the glioma progression and its prognostic significance for glioma patients remains unclear. METHODS: NTS precursor (ProNTS), NTS and NTSR1 expression levels in glioma were detected by immunobloting Elisa and immunohistochemistry assay. The prognostic analysis was conducted from internet by R2 microarray platform. Glioma cell proliferation was evaluated by CCK8 and BrdU incorporation assay. Wound healing model and Matrigel transwell assay were utilized to test cellular migration and invasion. The orthotopic glioma implantations were established to analyze the role of NTS and NTSR1 in glioma progression in vivo. RESULTS: Positive correlations were shown between the expression levels of NTS and NTSR1 with the pathological grade of gliomas. The high expression levels of NTS and NTSR1 indicate a worse prognosis in glioma patients. The proliferation and invasiveness of glioma cells could be enhanced by NTS stimulation and impaired by the inhibition of NTSR1. NTS stimulated Erk1/2 phosphorylation in glioma cells, which could be reversed by SR48692 or NTSR1-siRNA. In vivo experiments showed that SR48692 significantly prolonged the survival length of glioma-bearing mice and inhibited glioma cell invasiveness. CONCLUSION: NTS promotes the proliferation and invasion of glioma via the activation of NTSR1. High expression levels of NTS and NTSR1 predict a poor prognosis in glioma patients.


Asunto(s)
Movimiento Celular/genética , Glioma/genética , Glioma/patología , Invasividad Neoplásica/genética , Neurotensina/genética , Receptores de Neurotensina/genética , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Glioma/tratamiento farmacológico , Humanos , Ratones , Fosforilación/efectos de los fármacos , Fosforilación/genética , Pronóstico , Precursores de Proteínas/genética , Pirazoles/farmacología , Quinolinas/farmacología
12.
Neural Regen Res ; 19(5): 1072-1077, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-37862210

RESUMEN

MIcroglia/macrophage-mediated erythrophagocytosis plays a crucial role in hematoma clearance after intracerebral hemorrhage. Dynamic cytoskeletal changes accompany phagocytosis. However, whether and how these changes are associated with microglia/macrophage-mediated erythrophagocytosis remain unclear. In this study, we investigated the function of acetylated α-tubulin, a stabilized microtubule form, in microglia/macrophage erythrophagocytosis after intracerebral hemorrhage both in vitro and in vivo. We first assessed the function of acetylated α-tubulin in erythrophagocytosis using primary DiO GFP-labeled red blood cells co-cultured with the BV2 microglia or RAW264.7 macrophage cell lines. Acetylated α-tubulin expression was significantly decreased in BV2 and RAW264.7 cells during erythrophagocytosis. Moreover, silencing α-tubulin acetyltransferase 1 (ATAT1), a newly discovered α-tubulin acetyltransferase, decreased Ac-α-tub levels and enhanced the erythrophagocytosis by BV2 and RAW264.7 cells. Consistent with these findings, in ATAT1-/- mice, we observed increased ionized calcium binding adapter molecule 1 (Iba1) and Perls-positive microglia/macrophage phagocytes of red blood cells in peri-hematoma and reduced hematoma volume in mice with intracerebral hemorrhage. Additionally, knocking out ATAT1 alleviated neuronal apoptosis and pro-inflammatory cytokines and increased anti-inflammatory cytokines around the hematoma, ultimately improving neurological recovery of mice after intracerebral hemorrhage. These findings suggest that ATAT1 deficiency accelerates erythrophagocytosis by microglia/macrophages and hematoma absorption after intracerebral hemorrhage. These results provide novel insights into the mechanisms of hematoma clearance and suggest ATAT1 as a potential target for the treatment of intracerebral hemorrhage.

13.
Exp Neurol ; 374: 114691, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38224942

RESUMEN

Blood-brain barrier (BBB) impairment and glutamate release are two pathophysiological features of traumatic brain injury (TBI), contributing to secondary brain damage and neuroinflammation. However, our knowledge of BBB integrity damage and dysfunction are still limited due to the diverse and fluctuating expression of glutamate receptors after trauma. Here, we confirmed the downregulation of metabotropic glutamate receptor 5 (mGluR5) on microvascular endothelial cell within the acute phase of TBI, and the recovered mGluR5 levels on BBB was positively associated with blood perfusion and neurological recovery. In whole body mGluR5-knockout mice, BBB dysfunction and neurological deficiency were exacerbated after TBI compared with wild type mice. In terms of mechanism, the amino acid sequence 201-259 of cytoskeletal protein Alpha-actinin-1 (ACTN1) interacted with mGluR5, facilitating mGluR5 translocation from cytoplasmic compartment to plasma membrane in endothelial cells. Activation of plasma membrane mGluR5 triggers the PLC/PKCµ/c-Jun signaling pathway, leading to increased expression of the tight junction-actin cytoskeleton connecting protein zonula occludens-1 (ZO-1). Our findings uncover a novel mechanism mediated by membrane and cytoplasmic mGluR5 in endothelial cell integrity maintenance and repair, providing the potential therapeutic target for TBI treatment targeting at mGluR5 and mGluR5/ACTN1 complex in BBB.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Animales , Ratones , Barrera Hematoencefálica/metabolismo , Lesiones Encefálicas/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Células Endoteliales/metabolismo , Ratones Noqueados , Receptor del Glutamato Metabotropico 5/metabolismo
14.
Int J Med Sci ; 10(4): 399-407, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23471193

RESUMEN

OBJECTIVE: To investigate the relationship between hypoxia and in vitro "stemness" of cancer stem cells (CSCs). METHODS: U87 cells, U251 cells and primary glioma cells (n=3) experienced hypoxia. Transmission electron microscopy was done to detect the ultrastructure of these cancer cells; MTT assay to detect the cell growth; flow cytometry to detect cell cycle and CD133 expression; Transwell chamber assay was carried out to detect the cell migration; colony-forming assay to detect the colony-forming efficiency; real-time quantitative PCR and Western blot were carried out to detect the mRNA and protein expression of markers of stem cells and their differentiation, respectively. RESULTS: Hypoxia maintained the undifferentiated state of primary glioma cells, slowed down the growth of glioma cells which were in a relatively quiescent stage, increased the colony forming efficiency and migration of glioma cells, and elevated the expression of markers of stem cells, but the expression of markers for stem cell differentiation was reduced after hypoxia treatment. CONCLUSION: Hypoxia may induce the "dedifferentiation" of differentiated glioma cells which then acquire the stemness.


Asunto(s)
Neoplasias Encefálicas/patología , Desdiferenciación Celular/genética , Hipoxia de la Célula/genética , Células Madre Neoplásicas/citología , Oxígeno/farmacología , Antígeno AC133 , Antígenos CD , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Desdiferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/metabolismo , Glioma/patología , Glicoproteínas , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Técnicas In Vitro , Invasividad Neoplásica/genética , Células Madre Neoplásicas/metabolismo , Péptidos
15.
Front Cell Infect Microbiol ; 13: 1296491, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38274731

RESUMEN

Streptomyces, the largest genus in the Streptomycetaceae family and a prolific producer of antibacterial drugs, is a saprophytic soil organism that rarely causes invasive infections. Here we report a case of necrotic pneumonia caused by Streptomyces albireticuli in a 75-year-old man who presented with progressive chest tightness and dyspnea. Streptomyces albireticuli was isolated from his bronchoalveolar lavage fluid and identified through whole-genome sequencing (WGS) and phylogenetic analysis. The patient responded satisfactorily to clarithromycin therapy. The findings of this study may enhance our vigilance in identifying visceral infections caused by Streptomyces.


Asunto(s)
Quistes , Enfermedades Pulmonares , Neumonía , Streptomyces , Masculino , Humanos , Anciano , Filogenia , Streptomyces/genética , Pulmón
16.
Front Neurol ; 14: 1247549, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38313405

RESUMEN

Background: Endovascular or surgical treatment of wide-neck, large basilar apex aneurysms is challenging. We present a novel concept for the treatment of complex basilar apex aneurysms using flow-diverter devices combined with the flow-T stenting-assisted coiling technique. Assess the efficacy and safety profile of the technique in this complex aneurysm. Case description: A patient with multiple unruptured intracranial aneurysms underwent staged treatment. A large basilar apex aneurysm was treated with a flow-diverter stent combined with a flow-T stenting-assisted coiling technique in the first stage, and a giant supraclinoid aneurysm was treated with a flow-diverter stent applied in the second stage. Clinical presentations, technical details, intra- and perioperative complications, and clinical and angiographic outcomes were recorded, with a 9-month follow-up. Results: The patient achieved full neurologic recovery postoperatively. Cerebral angiography performed postoperatively showed revascularization, good laminar flow, and no in-stent or adjacent stenosis. Conclusion: Flow-diverter stents combined with flow-T stenting-assisted coiling for the treatment of giant basilar apex aneurysms is a feasible technique with efficacy demonstrated at a 9-month follow-up. Staged endovascular treatment of multiple intracranial aneurysms may be a safe and viable option.

17.
J Biol Chem ; 286(42): 36694-9, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-21896488

RESUMEN

Tumor tolerance plays a critical role in tumor growth and escape from immune surveillance. The mechanism of tumor tolerance development is not fully understood. Regulatory T cells (Tregs) play a critical role in tumor tolerance. TIM4 (T cell immunoglobulin- and mucin domain-containing molecule-4) is involved in immune regulation. We investigated the role of TIM4 in the induction of Tregs in tumors. Surgically removed glioma tissue and peripheral blood samples were obtained from 25 glioma patients. Immune cells were isolated from the tissue and blood samples. Confocal microscopy was employed to detect macrophages phagocytosing apoptotic T cells. The generation of tumor-specific Tregs and the immune suppression function of Tregs were observed in cell culture models. High levels of TIM4 were detected in glioma-derived macrophages. Phosphatidylserine (PS) was detected in glioma-derived T cells; naïve T cells expressed low levels of PS that could be up-regulated by hypoxia. Glioma-derived macrophages phagocytosed PS-expressing T cells, gaining the tolerogenic properties, which could induce tumor-specific Tregs; the latter could suppress tumor-specific CD8(+) T cells. We conclude that macrophage-derived TIM4 plays an important role in the induction of Tregs in gliomas, which may play an important role in tumor tolerance.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/inmunología , Glioma/inmunología , Tolerancia Inmunológica , Macrófagos/inmunología , Proteínas de la Membrana/inmunología , Linfocitos T Reguladores/inmunología , Hipoxia de la Célula/inmunología , Femenino , Glioma/metabolismo , Glioma/patología , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Proteínas de la Membrana/biosíntesis , Fagocitosis/inmunología , Fosfatidilserinas/biosíntesis , Fosfatidilserinas/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Regulación hacia Arriba/inmunología
18.
Mol Cell Biochem ; 364(1-2): 235-41, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22350753

RESUMEN

Accumulated evidence suggests a major role for the activation of the Sonic Hedgehog (SHH) signaling pathway in the development of neural crest stem cells that give rise to the sympathetic nervous system. We therefore investigated the involvement of SHH signaling in the pathogenesis of neuroblastoma (NB), a common childhood malignant tumor of the sympathetic nervous system. Inhibition of SHH signaling by cyclopamine induced apoptosis and blocked proliferation in all major types of NB cells, and abrogated the tumorigenicity of NB cells. Our study has revealed a molecular mechanism for the persistent activation of the SHH pathway which promotes the development of NB, and suggests a new approach for the treatment of this childhood malignant tumor.


Asunto(s)
Proteínas Hedgehog/metabolismo , Neoplasias de Tejido Nervioso/metabolismo , Neuroblastoma/metabolismo , Transducción de Señal , Apoptosis/efectos de los fármacos , Pruebas de Carcinogenicidad , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/genética , Humanos , Terapia Molecular Dirigida , Neoplasias de Tejido Nervioso/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Cresta Neural/metabolismo , Cresta Neural/patología , Neuroblastoma/patología , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/patología , Alcaloides de Veratrum/farmacología
19.
Neurol Sci ; 33(5): 1057-62, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22234522

RESUMEN

Eukaryotic initiation factors 3 (EIF3) complex is essential for initiation of protein synthesis for both cells and virus. It consists of 13 subunits (EIF3A to M), among which EIF3B serves as a major scaffolding subunit. However, its functions in human glioblastoma have not been explored yet. Here, we showed that EIF3B was expressed in human glioblastoma (Grade I-IV) and human glioblastoma cell lines (U251, U87, A172 and U373). Loss of function analysis was performed on U87 cells using lentivirus-mediated siRNA against EIF3B. EIF3B-shRNA expressing lentivirus could effectively infect U87 glioma cells and downregulate EIF3B expression. Knockdown of EIF3B expression significantly inhibited proliferation of U87 cells. Further study showed that the proliferation inhibitory effect was associated with accumulation in G0/G1-phase cell number and an increased rate of apoptosis. In conclusion, EIF3B promotes the proliferation of U87 cells and may play an important role in human glioblastoma development.


Asunto(s)
Apoptosis , Proliferación Celular , Factor 3 de Iniciación Eucariótica/genética , Glioblastoma/genética , Línea Celular Tumoral , Factor 3 de Iniciación Eucariótica/metabolismo , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Comput Math Methods Med ; 2022: 3436631, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35912147

RESUMEN

Objective: To develop and authenticate a risk stratification framework and nomogram for ascertaining cancer-specific survival (CSS) among the pediatric brainstem gliomas. Methods: For patients less than 12 years, according to Surveillance, Epidemiology, and End Results (SEER), information from 1998 to 2016 is found in their databases. The survival outcomes, treatments, and demographic clinicopathologic conditions are scrutinized per the database validation, and training cohorts are divided and validated using multivariate Cox regression analysis. A nomogram was designed, and predominantly, the risk stratification conceptualization engaged selected tenets according to the multivariate analysis. The model's authenticity was substantiated through C-index measure and calibration curves. Results: There are 806 pediatric concerns of histologically concluded brainstem glioma in the research. According to multivariate analysis, age, grade, radiotherapy, and race (with P value < 0.05) depicted independent prognostic variations of the pediatric gliomas. The nomogram's C-index was approximately 0.75 and an accompanied predictive capability for CSS. Conclusion: The nomogram constructed in this glioma's context is the primary predictor of using risk stratification. A combination of nomograms with the risk stratification mechanism assists clinicians in monitoring high-risk individuals and engage targeted accessory treatment.


Asunto(s)
Neoplasias Encefálicas/mortalidad , Tronco Encefálico/patología , Glioma/mortalidad , Neoplasias Encefálicas/terapia , Niño , Preescolar , Estudios de Cohortes , Glioma/terapia , Humanos , Lactante , Análisis Multivariante , Nomogramas , Pronóstico , Análisis de Regresión , Medición de Riesgo/métodos , Programa de VERF
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA