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












Base de datos
Intervalo de año de publicación
1.
iScience ; 27(5): 109698, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38655198

RESUMEN

Mounting evidence supports the role of neuroinflammation in radiation-induced brain injury (RIBI), a chronic disease characterized by delayed and progressive neurological impairment. Asparagine endopeptidase (AEP), also known as legumain (LGMN), participates in multiple malignancies and neurodegenerative diseases and may potentially be involved in RIBI. Here, we found AEP expression was substantially elevated in the cortex and hippocampus of wild-type (Lgmn+/+) mice following whole-brain irradiation. Lgmn knockout (Lgmn-/-) alleviated neurological impairment caused by whole-brain irradiation by suppressing neuronal senescence. Bulk RNA and metabolomic sequencing revealed AEP's involvement in the antigen processing and presentation pathway and neuroinflammation. This was further confirmed by co-culturing Lgmn+/+ primary neurons with the conditioned media derived from irradiated Lgmn+/+ or Lgmn-/- primary microglia. Furthermore, esomeprazole inhibited the enzymatic activity of AEP and RIBI. These findings identified AEP as a critical factor of neuroinflammation in RIBI, highlighting the prospect of targeting AEP as a therapeutic approach.

2.
J Clin Invest ; 134(3)2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-37988165

RESUMEN

Oxygen and nutrient deprivation are common features of solid tumors. Although abnormal alternative splicing (AS) has been found to be an important driving force in tumor pathogenesis and progression, the regulatory mechanisms of AS that underly the adaptation of cancer cells to harsh microenvironments remain unclear. Here, we found that hypoxia- and nutrient deprivation-induced asparagine endopeptidase (AEP) specifically cleaved DDX3X in a HIF1A-dependent manner. This cleavage yields truncated carboxyl-terminal DDX3X (tDDX3X-C), which translocates and aggregates in the nucleus. Unlike intact DDX3X, nuclear tDDX3X-C complexes with an array of splicing factors and induces AS events of many pre-mRNAs; for example, enhanced exon skipping (ES) in exon 2 of the classic tumor suppressor PRDM2 leads to a frameshift mutation of PRDM2. Intriguingly, the isoform ARRB1-Δexon 13 binds to glycolytic enzymes and regulates glycolysis. By utilizing in vitro assays, glioblastoma organoids, and animal models, we revealed that AEP/tDDX3X-C promoted tumor malignancy via these isoforms. More importantly, high AEP/tDDX3X-C/ARRB1-Δexon 13 in cancerous tissues was tightly associated with poor patient prognosis. Overall, our discovery of the effect of AEP-cleaved DDX3X switching on alternative RNA splicing events identifies a mechanism in which cancer cells adapt to oxygen and nutrient shortages and provides potential diagnostic and/or therapeutic targets.


Asunto(s)
Empalme Alternativo , Glioblastoma , Animales , Humanos , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Glioblastoma/patología , Oxígeno/metabolismo , Isoformas de Proteínas/metabolismo , Empalme del ARN , Factores de Empalme de ARN/metabolismo , Microambiente Tumoral
3.
Mol Biotechnol ; 2023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37751129

RESUMEN

Investigations of protein-protein interactions (PPIs) are of paramount importance for comprehending cellular processes within biological systems. The bimolecular fluorescence complementation (BiFC) assay presents a convenient methodology for visualizing PPIs within live cells. While a range of fluorescent proteins have been introduced into the BiFC system, there is a growing demand for new fluorescent proteins to accommodate the expanding requirements of researchers. This study describes the introduction of Tagged blue fluorescent protein 2 (TagBFP2) into the BiFC assay to verify the interaction between two proteins, with Enhanced yellow fluorescent protein (EYFP) employed as a positive control. Both fluorescent proteins demonstrated optimal performance in this study. Compared to EYFP, the BiFC system utilizing TagBFP2 yielded a higher signal-to-noise ratio, which facilitated differentiation of the signal of PPIs from noise and enabled employment of other fluorescent proteins within the BiFC assay. Notably, the utilization of a fluorescent secondary antibody in immunofluorescence applications or the tagging of an interest protein with a fluorescent protein occupied the green or yellow channel. Overall, the present article introduces a BiFC assay that is highly straightforward, reliable, and replicable, with the ability to be completed within 1 week. This method requires neither expensive instrumentation nor technical skills of a high order.

4.
J Appl Clin Med Phys ; 23(11): e13758, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36107021

RESUMEN

INTRODUCTION: To explore and evaluate the performance of MRI-based brain tumor super-resolution generative adversarial network (MRBT-SR-GAN) for improving the MRI image resolution in brain tumors. METHODS: A total of 237 patients from December 2018 and April 2020 with T2-fluid attenuated inversion recovery (FLAIR) MR images (one image per patient) were included in the present research to form the super-resolution MR dataset. The MRBT-SR-GAN was modified from the enhanced super-resolution generative adversarial networks (ESRGAN) architecture, which could effectively recover high-resolution MRI images while retaining the quality of the images. The T2-FLAIR images from the brain tumor segmentation (BRATS) dataset were used to evaluate the performance of MRBT-SR-GAN contributed to the BRATS task. RESULTS: The super-resolution T2-FLAIR images yielded a 0.062 dice ratio improvement from 0.724 to 0.786 compared with the original low-resolution T2-FLAIR images, indicating the robustness of MRBT-SR-GAN in providing more substantial supervision for intensity consistency and texture recovery of the MRI images. The MRBT-SR-GAN was also modified and generalized to perform slice interpolation and other tasks. CONCLUSIONS: MRBT-SR-GAN exhibited great potential in the early detection and accurate evaluation of the recurrence and prognosis of brain tumors, which could be employed in brain tumor surgery planning and navigation. In addition, this technique renders precise radiotherapy possible. The design paradigm of the MRBT-SR-GAN neural network may be applied for medical image super-resolution in other diseases with different modalities as well.


Asunto(s)
Neoplasias Encefálicas , Aprendizaje Profundo , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Imagen por Resonancia Magnética/métodos , Redes Neurales de la Computación , Neoplasias Encefálicas/diagnóstico por imagen
6.
J Exp Clin Cancer Res ; 41(1): 209, 2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-35765111

RESUMEN

BACKGROUND: Abnormal proliferation and migration of cells are hallmarks of cancer initiation and malignancy. Asparagine endopeptidase (AEP) has specific substrate cleavage ability and plays a pro-cancer role in a variety of cancers. However, the underlying mechanism of AEP in cancer proliferation and migration still remains unclear. METHODS: Co-immunoprecipitation and following mass spectrometry were used to identify the substrate of AEP. Western blotting was applied to measure the expression of proteins. Single cell/nuclear-sequences were done to detect the heterogeneous expression of Tmod3 in tumor tissues. CCK-8 assay, flow cytometry assays, colony formation assay, Transwell assay and scratch wound-healing assay were performed as cellular functional experiments. Mouse intracranial xenograft tumors were studied in in vivo experiments. RESULTS: Here we showed that AEP cleaved a ubiquitous cytoskeleton regulatory protein, tropomodulin-3 (Tmod3) at asparagine 157 (N157) and produced two functional truncations (tTmod3-N and tTmod3-C). Truncated Tmod3 was detected in diverse tumors and was found to be associated with poor prognosis of high-grade glioma. Functional studies showed that tTmod3-N and tTmod3-C enhanced cancer cell migration and proliferation, respectively. Animal models further revealed the tumor-promoting effects of AEP truncated Tmod3 in vivo. Mechanistically, tTmod3-N was enriched in the cell cortex and competitively inhibited the pointed-end capping effect of wild-type Tmod3 on filamentous actin (F-actin), leading to actin remodeling. tTmod3-C translocated to the nucleus, where it interacted with Staphylococcal Nuclease And Tudor Domain Containing 1 (SND1), facilitating the transcription of Ras Homolog Family Member A/Cyclin Dependent Kinases (RhoA/CDKs). CONCLUSION: The newly identified AEP-Tmod3 protease signaling axis is a novel "dual-regulation" mechanism of tumor cell proliferation and migration. Our work provides new clues to the underlying mechanisms of cancer proliferation and invasive progression and evidence for targeting AEP or Tmod3 for therapy.


Asunto(s)
Actinas , Neoplasias Encefálicas , Cisteína Endopeptidasas , Endonucleasas , Glioma , Tropomodulina , Proteína de Unión al GTP rhoA , Actinas/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Quinasas Ciclina-Dependientes/metabolismo , Cisteína Endopeptidasas/metabolismo , Proteínas del Citoesqueleto , Endonucleasas/metabolismo , Glioma/metabolismo , Glioma/patología , Xenoinjertos , Humanos , Ratones , Transducción de Señal , Tropomodulina/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
7.
Front Oncol ; 11: 740413, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34778058

RESUMEN

OBJECTIVE: The aim of this study was to establish a nomogram model for predicting the risk of short-term recurrence in glioma patients. METHODS: The clinical data of recurrent glioma patients were summarized and analyzed in this study. Univariate and multivariate logistic regression analyses were performed to analyze the correlation between clinical data and the risk of short-term recurrence after operation. A nomogram was established based on the multivariate logistic regression model results. RESULTS: A total of 175 patients with recurrent glioma were enrolled, with 53 patients in the short-term recurrence (STR) group (recurrent time ≤6 months) and 122 patients in the long-term recurrence (LTR) group (recurrent time ≥36 months). Univariate analysis revealed that age at diagnosis, Karnofsky performance scores (KPSs), tumor location, glioma grade, glioma type, extent of resection (EOR), adjuvant chemotherapy (ad-CT), concurrent chemotherapy (co-CT), and isocitrate dehydrogenase (IDH) status were significantly associated with the short-term glioma recurrence. Multivariate analyses revealed that age at diagnosis, KPS, glioma grade, EOR, and IDH were independent risk factors for short-term glioma recurrence. A risk nomogram for the short-term recurrence of glioma was established, with the concordance index (C-index) of 0.971. The findings of calibration and receiver operating characteristic (ROC) curves showed that our nomogram model had good performance and discrimination to estimate short-term recurrence probability. CONCLUSION: This nomogram model provides reliable information about the risk of short-term glioma recurrence for oncologists and neurosurgeons. This model can predict the short-term recurrence probability and give assistance to decide the interval of follow-up or formulate individualized treatment strategies based on the predicted results. A free online prediction risk tool for this nomogram is provided: https://rj2021.shinyapps.io/Nomogram_ recurrence-risk/.

8.
Cancer Lett ; 522: 225-237, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34582975

RESUMEN

Glioblastoma (GBM) is one of the most devastating cancers and is characterized by rapid cell proliferation and aggressive invasiveness. Legumain (LGMN), a substrate-specific protease, is associated with poor progression of GBM. Circular RNAs (circRNAs) are aberrantly expressed in various cancers and play crucial roles in tumor progression; however, the functional roles of circRNAs originating from LGMN remain largely unknown in GBM. Herein, we found that hsa_circ_0033009 (circLGMN) was the most abundantly expressed circRNA derived from LGMN. CircLGMN was upregulated in high-grade glioma (HGG), and high expression of circLGMN was associated with poor prognosis in patients with glioma. CircLGMN overexpression promoted GBM cell proliferation and enhanced cell invasion. Mechanistically, circLGMN acts as a sponge for miR-127-3p, and prevents miR-127-3p-mediated degradation of LGMN mRNA, ultimately leading to increased LGMN protein expression. Treatment with miR-127-3p mimic suppressed proliferation and reduced invasion of GBM cells overexpressing circLGMN. Moreover, circLGMN overexpression promoted GBM malignancy in vivo, while miR-127-3p overexpression alleviated this effect. Taken together, circLGMN is a novel tumor-promoting circRNA that acts by sponging miR-127-3p, which ultimately leads to LGMN upregulation. Thus, targeting the circLGMN/miR-127-3p/LGMN axis might be a promising strategy for GBM treatment. More importantly, the discovery of the self-regulatory mechanism of LGMN expression by circLGMN, will facilitate further research on LGMN.


Asunto(s)
Cisteína Endopeptidasas/genética , Glioblastoma/genética , MicroARNs/genética , ARN Circular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/patología , Humanos
9.
Front Oncol ; 11: 801776, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35004326

RESUMEN

Glioblastoma is a rare yet lethal type of tumor that poses a crucible for the medical profession, owing to its rapid proliferation and invasion resulting in poor prognosis. Circular RNAs (circRNAs), a subclass of regulatory RNAs, are implicated in the regulation of cancerous progression. This study aims to investigate the roles and underlying mechanism of circPIK3C2A in regulating proliferation and invasion of glioblastoma. qRT-PCR assays showed that the expression level of circPIK3C2A was aberrantly higher in glioblastoma cell lines, in comparison with that in normal glia cells. The ectopic expression of circPIK3C2A promoted the proliferation, invasion and clonal formation of glioblastoma cells, while circPIK3C2A loss-of-function exerted exactly the opposite biological effects on the cells. The construction of subcutaneous xenograft tumor model in nude mice indicated that circPIK3C2A loss-of-function effectively diminished tumor load in vivo and prolonged the survival time of tumor-bearing animals. Luciferase reporter assay confirmed the interaction among circPIK3C2A/miR-877-5p and FOXM1. CircPIK3C2A function as competitive endogenous RNA via sponging miR-877-5p through certain binding sites, thereby modulating the expression of FOXM1. Our results collectively indicate that circPIK3C2A functions as ceRNA by mediating miR-877-5p/FOXM1 axis, providing a novel perspective of applying CircPIK3C2A in the clinical intervention of glioblastoma in the future.

10.
Cancer Lett ; 490: 111-123, 2020 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-32711096

RESUMEN

Pseudogenes, which are long noncoding RNAs that originate from protein-coding genes, have been suggested to play important roles in disease. Although studies have revealed high expression of legumain (LGMN) in many types of tumors, the regulation of LGMN remains largely unknown. Here, we found that a novel LGMN pseudogene (LGMNP1) was upregulated in glioblastoma (GBM) tissues and high LGMNP1 expression in GBM cells enhanced proliferation and invasion. Biochemical analysis showed that cytoplasmic LGMNP1 functionally targeted miR-495-3p in a manner involving an RNA-induced silencing complex. Dual-luciferase reporter assays demonstrated that LGMN was a target of miR-495-3p, and LGMN was upregulated and positively correlated with LGMNP1 in GBM. Moreover, miR-495-3p was downregulated and negatively correlated with LGMNP1 in GBM tissues. Notably, the tumor-promoting effects of LGMNP1 upregulation could be alleviated by miR-495-3p mimics. Furthermore, GBM cells overexpressing LGMNP1 exhibited more aggressive tumor progression and elevated LGMN expression in vivo. Thus, our data illustrate that LGMNP1 exerts its oncogenic activity, at least in part, as a competitive endogenous RNA (ceRNA) that elevates LGMN expression by sponging miR-495-3p. CeRNA-mediated miRNA sequestration might be a novel therapeutic strategy in GBM.


Asunto(s)
Neoplasias Encefálicas/patología , Cisteína Endopeptidasas/genética , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/patología , MicroARNs/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Cisteína Endopeptidasas/metabolismo , Progresión de la Enfermedad , Glioblastoma/genética , Glioblastoma/metabolismo , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Seudogenes/genética , ARN Largo no Codificante/genética
11.
J Natl Cancer Inst ; 112(4): 343-355, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31400201

RESUMEN

BACKGROUND: Isocitrate dehydrogenase wild-type (WT) glioblastoma (GBM) accounts for 90% of all GBMs, yet only 27% of isocitrate dehydrogenase WT-GBMs have p53 mutations. However, the tumor surveillance function of WT-p53 in GBM is subverted by mechanisms that are not fully understood. METHODS: We investigated the proteolytic inactivation of WT-p53 by asparaginyl endopeptidase (AEP) and its effects on GBM progression in cancer cells, murine models, and patients' specimens using biochemical and functional assays. The sera of healthy donors (n = 48) and GBM patients (n = 20) were examined by enzyme-linked immunosorbent assay. Furthermore, effects of AEP inhibitors on GBM progression were evaluated in murine models (n = 6-8 per group). The statistical significance between groups was determined using two-tailed Student t tests. RESULTS: We demonstrate that AEP binds to and directly cleaves WT-p53, resulting in the inhibition of WT-p53-mediated tumor suppressor function in both tumor cells and stromal cells via extracellular vesicle communication. High expression of uncleavable p53-N311A-mutant rescue AEP-induced tumorigenesis, proliferation, and anti-apoptotic abilities. Knock down or pharmacological inhibition of AEP reduced tumorigenesis and prolonged survival in murine models. However, overexpression of AEP promoted tumorigenesis and shortened the survival time. Moreover, high AEP levels in GBM tissues were associated with a poor prognosis of GBM patients (n = 83; hazard ratio = 3.94, 95% confidence interval = 1.87 to 8.28; P < .001). A correlation was found between high plasma AEP levels and a larger tumor size in GBM patients (r = 0.6, P = .03), which decreased dramatically after surgery. CONCLUSIONS: Our results indicate that AEP promotes GBM progression via inactivation of WT-p53 and may serve as a prognostic and therapeutic target for GBM.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Glioblastoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Inhibidores de Cisteína Proteinasa/farmacología , Progresión de la Enfermedad , Glioblastoma/enzimología , Glioblastoma/patología , Xenoinjertos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Transgénicos , Células del Estroma/enzimología , Células del Estroma/metabolismo , Células del Estroma/patología
12.
Mol Ther Nucleic Acids ; 18: 388-399, 2019 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-31648104

RESUMEN

Zinc fingers and homeoboxes 1 (ZHX1) is a transcription repressor that has been implicated in the tumorigenesis and progression of diverse tumors. The functional role and regulating mechanism of ZHX1 has not been elucidated in glioblastoma (GBM). Previous reports have suggested that a large number of non-coding RNAs play a vital role in glioma initiation and progression. This study aimed to investigate the functional role and co-regulatory mechanisms of the metastasis-associated lung adenocarcinoma transcript-1 (MALAT1)/ microRNA-199a (miR-199a)/ZHX1 axis in GBM. We analyzed the expression of the MALAT1/miR-199a/ZHX1 axis and its correlation with patients' overall survival using two different glioma gene-expression datasets. A series of in vitro and in vivo studies including dual luciferase reporter assay, fluorescence in situ hybridization (FISH), RNA immunoprecipitation, and pull-down experiments were completed to elucidate the biological significance of the MALAT1/miR-199a/ZHX1 axis in promoting glioma proliferation and progression. Elevated ZHX1 expression correlated with poor prognosis in GBM patients, and in vitro studies demonstrated that ZHX1 attenuated GBM cell apoptosis by downregulation of pro-apoptotic protein (Bax) and upregulation of anti-apoptotic protein (Bcl-2). Furthermore, knockdown of MALAT1 inhibited GBM proliferation and progression in vitro and reduced tumor volume and prolonged survival in an orthotopic GBM murine model. Finally, we demonstrated that MALAT1 promoted ZHX1 expression via acting as a competing endogenous RNA by sponging miR-199a. The MALAT1/miR-199a/ZHX1 axis promotes GBM cell proliferation and progression in vitro and in vivo, and its expression negatively correlates with GBM patient survival. Blocking the MALAT1/miR-199a/ZHX1 axis can serve as a novel therapeutic strategy for treating GBM.

13.
Neuro Oncol ; 21(10): 1284-1296, 2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31504797

RESUMEN

BACKGROUND: Circular RNAs (circRNAs), a newly discovered type of endogenous noncoding RNA, have been proposed to mediate the progression of diverse types of tumors. Systematic studies of circRNAs have just begun, and the physiological roles of circRNAs remain largely unknown. Here, we focused on elucidating the potential role and molecular mechanism of circular forkhead box O3 (circFOXO3) in glioblastoma (GBM) progression. METHODS: First, we analyzed circFOXO3 alterations in GBM and noncancerous tissues through real-time quantitative reverse transcription PCR (qRT-PCR). Next, we used loss- and gain-of-function approaches to evaluate the effect of circFOXO3 on GBM cell proliferation and invasion. Mechanistically, fluorescent in situ hybridization, RNA pull-down, dual luciferase reporter, and RNA immunoprecipitation assays were performed to confirm the interaction between circFOXO3 and miR-138-5p/miR-432-5p in GBM. An animal model was used to verify the in vitro experimental findings. RESULTS: CircFOXO3 expression was significantly higher in GBM tissues than in noncancerous tissues. GBM cell proliferation and invasion were reduced by circFOXO3 knockdown and enhanced by circFOXO3 overexpression. Further biochemical analysis showed that circFOXO3 exerted its pro-tumorigenic activity by acting as a competing endogenous RNA (ceRNA) to increase expression of nuclear factor of activated T cells 5 (NFAT5) via sponging both miR-138-5p and miR-432-5p. Notably, tumor inhibition by circFOXO3 downregulation could be reversed by miR-138-5p/miR-432-5p inhibitors in GBM cells. Moreover, GBM cells with lower circFOXO3 expression developed less aggressive tumors in vivo. CONCLUSIONS: Our data demonstrate that circFOXO3 can exert regulatory functions in GBM and that ceRNA-mediated microRNA sequestration might be a potential strategy for GBM therapy.


Asunto(s)
Neoplasias Encefálicas/patología , Proteína Forkhead Box O3/genética , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/patología , Factores de Transcripción/genética , Animales , Neoplasias Encefálicas/genética , ADN Circular/genética , Progresión de la Enfermedad , Glioblastoma/genética , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , MicroARNs/genética , Factores de Transcripción/metabolismo
14.
Int J Biol Sci ; 15(4): 738-748, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30906206

RESUMEN

Ubiquitin-specific protease 17 (USP17), a novel member of deubiquitinase, is reported to play essential roles in several solid tumors. However, the expression and function of USP17 in breast cancer tumorigenesis remains ambiguity. Here we found that the mRNA level of USP17 was lower in breast cancer tissues than normal tissues. Meanwhile, higher USP17 level was detected in normal epithelial cell MCF-10A and a less-malignant cell MCF-7 than malignant cell line MDA-MB-231. Inhibition of USP17 in MCF7 cells enhanced tumorigenesis and tumor growth while overexpression of USP17 in malignant MDA-MB-231 cells reduced its tumorigenesis and growth ability in vitro and in vivo. Further study revealed that USP17 interacted with and deubiquitinated Asparaginyl endopeptidase (AEP), resulting in decreased protein levels of AEP. Moreover, knockdown of AEP inhibited breast cancer tumorigenesis and growth in vitro and in vivo through the inactivation of ERK signaling. Taken together, our works indicate that USP17 deubiquitinates AEP, down-regulates its protein level, and inhibits breast cancer tumorigenesis through disturbing ERK signaling. Thus, our data suggests that USP17 is a potential tumor suppressor in breast cancer and AEP is a promising target in breast cancer therapy.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Neoplasias de la Mama/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Cisteína Endopeptidasas/metabolismo , Femenino , Humanos , Células MCF-7 , Ubiquitinación/fisiología
15.
Biochem Biophys Res Commun ; 509(2): 617-623, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30606477

RESUMEN

Long non-coding RNAs (lncRNAs) play vital roles in the pathobiology of glioblastoma multiforme (GBM). Though radiotherapy remains the most effective component of multiple therapies for patients with GBM, lncRNAs conferring GBM radioresistance are less unknown. Here, the present study identified that the antisense transcript of hypoxia-inducible factor-1α (AHIF) was upregulated in GBM cells after radiotherapy. The deregulation of AHIF affected GBM cell clonogenic formation, DNA repair and apoptosis. Notably, knockdown of AHIF inhibited tumorigenesis after radiotherapy in vivo. Further biochemical analysis identified that AHIF regulated proteins associated with apoptosis after radiotherapy. Thus, the present data illustrate that suppression of AHIF increases radiosensitivity in GBM cells, which may be a potential diagnostic and therapeutic target for GBM patients.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/radioterapia , Glioblastoma/genética , Glioblastoma/radioterapia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , ARN Largo no Codificante/genética , Regulación hacia Arriba , Animales , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Ratones , Tolerancia a Radiación , Regulación hacia Arriba/efectos de la radiación
16.
Int J Oncol ; 54(1): 261-270, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30387845

RESUMEN

Glioblastoma multiforme (GBM) has the highest mortality rate among patients with brain tumors, and radiotherapy forms an important part of its treatment. Thus, there is an urgent requirement to elucidate the mechanisms conferring GBM progression and radioresistance. In the present study, it was identified that antisense transcript of hypoxia­inducible factor­1α (AHIF) was significantly upregulated in GBM cancerous tissues, as well as in radioresistant GBM cells. The expression of AHIF was also upregulated in response to radiation. Knockdown of AHIF in GBM cells decreased viability and invasive capacities, and increased the proportion of apoptotic cells. By contrast, overexpression of AHIF in GBM cells increased viability and invasive capacities, and decreased the proportion of apoptotic cells. Furthermore, exosomes derived from AHIF­knockdown GBM cells inhibited viability, invasion and radioresistance, whereas exosomes derived from AHIF­overexpressing GBM cells promoted viability, invasion and radioresistance. Further biochemical analysis identified that AHIF regulates factors associated with migration and angiogenesis in exosomes. To the best of our knowledge, the present study is the first to establish that AHIF promotes glioblastoma progression and radioresistance via exosomes, which suggests that AHIF is a potential therapeutic target for GBM.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Exosomas/genética , Glioblastoma/metabolismo , ARN Largo no Codificante/genética , Tolerancia a Radiación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/radioterapia , Línea Celular Tumoral , Supervivencia Celular , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/radioterapia , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Regulación hacia Arriba , Adulto Joven
17.
J Craniofac Surg ; 28(2): e168-e170, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27792104

RESUMEN

Neurocutaneous melanosis (NCM) is a rare congenital syndrome characterized by the presence of multiple congenital melanocytic nevi and the proliferation of melanocytes in the central nervous system. The authors present a 9-year-old Chinese boy whose clinical manifestations are intermittent headache for 2 months and persistent abdominal pain for 10 days. 3D-reconstruction computed tomography angiography image, digital subtraction angiography, and magnetic resonance imaging plus angiography (MRI+MRA) examinations results suggested that cavernoma at left frontal lobe potentially associated with hemorrhage. In addition, miliary abnormal signals were widely scattered on MRA image so that other malignant metastatic diseases cannot be ruled out. GI physical examination had not any abnormal findings, antispasmodic drugs were ineffective but antiepilepsy drugs were effective to abdominal pain. In surgery, no cavernoma was noticed but an accumulation of densely melanocytic mass located at the lesion on radiology images. The lesions spread along with perivascular of sylvian veins and leptomeningeal. Pathology investigation demonstrated brain metastatic malignant melanoma associated with hemosiderosis. The lesion of brain parenchyma was totally removed but the spread lesions could not be treated with surgery. Adjuvant radiotherapy was performed but failed to control the malignant development, still the patient died in 3 months postinitial operation. The authors conclude that abdominal pain was a manifestation of epilepsy related to the frontal lobe lesion. Neurocutaneous melanosis is a rare disease, brain metastases result in abdominal pain is rare even more, and it is worth the attention of clinicians.


Asunto(s)
Dolor Abdominal/etiología , Neoplasias Encefálicas/patología , Hemangioma Cavernoso/etiología , Melanoma/patología , Melanosis/complicaciones , Síndromes Neurocutáneos/complicaciones , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/etiología , Niño , Cefalea/etiología , Hemangioma Cavernoso/diagnóstico por imagen , Hemangioma Cavernoso/cirugía , Humanos , Masculino , Melanoma/diagnóstico , Melanoma/etiología , Melanosis/diagnóstico por imagen , Melanosis/cirugía , Síndromes Neurocutáneos/diagnóstico por imagen , Síndromes Neurocutáneos/cirugía
18.
Folia Neuropathol ; 53(3): 236-44, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26443314

RESUMEN

MicroRNAs (miRNAs) are small noncoding RNAs that negatively regulate protein biosynthesis and participate in the pathogenesis of various tumours. Previous studies have shown that miR-210 is highly expressed in different types of human cancers, including glioblastoma multiforme (GBM). However, the role that miR-210 plays in GBM remains unclear. Here, we detected the expression and examined the function of miRNA-210 in GBM cells. Furthermore, we investigated the possible molecular mechanisms by which miRNA-210 mediates cell proliferation and apoptosis. Fifteen GBM and five normal brain tissues, in addition to the U87MG and U251 GBM cell lines, were analysed in this study. We found that miR-210 was upregulated in GBM tissues and cell lines when compared to normal brain tissue. Cell counting and flow cytometric assay results demonstrated that upregulation of miR-210 induced cell proliferation and decreased cell apoptosis, respectively. In addition, downregulation of miR-210 inhibited cell proliferation and induced apoptosis. We also detected a miR-210 target, regulator of differentiation 1 (ROD1), which is involved in GBM progression. Knockdown of ROD1 reversed the growth arrest and apoptosis that were originally induced by miR-210 inhibition. We propose that miR-210 regulates cell proliferation and apoptosis in GBM cells by targeting ROD1. Our findings may provide a new potential therapeutic target for the treatment of GBM.


Asunto(s)
Apoptosis , Neoplasias Encefálicas/patología , Proliferación Celular , Glioblastoma/patología , MicroARNs/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Anciano , Apoptosis/genética , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Proteína de Unión al Tracto de Polipirimidina/genética , Transfección
19.
Int J Clin Exp Med ; 8(1): 456-63, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25785017

RESUMEN

MicroRNA-128 is down-regulated in glioma tissues, which regulates cell proliferation, self-renewal, apoptosis, angiogenesis and differentiation. This study aims at investigating the diagnostic value of serum miR-128 in human glioma. Real-time quantitative reverse transcriptase polymerase chain reaction was used to detect the expression levels of miR-128 in serum samples from 151 glioma patients, 59 postoperative patients, 52 meningioma patients and 53 normal donors. To analyze the association of miR-128 expression with clinicopathological parameters in serum samples and matched tissues, matched 151 glioma tissues were collected in the study. Receiver operating characteristic analysis (ROC) was utilized to evaluate the value of serum miR-128 as a biomarker for the early diagnosis of glioma. Results revealed that miR-128 expression was significantly decreased in glioma preoperative serum compared with normal controls and meningioma serum samples (both P < 0.001). ROC analyses showed that serum miR-128 levels were reliable in distinguishing patients with glioma from normal controls and meningioma, with the area under the curve (AUC) values of 0.9095 and 0.8283, respectively. In addition, the AUC value for discriminating glioma II-IV from I was 0.7362. Importantly, serum miR-128 expression was significantly elevated after surgery (P < 0.001), although it didn't reach to normal levels (P < 0.001). Furthermore, low miR-128 levels in serum and tissue were markedly correlated with high pathological grade and low Karnofsky Performance Status score (KPS). These findings proved that serum miR-128 could be a sensitive and specific biomarker of glioma.

20.
Med Oncol ; 31(4): 911, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24623016

RESUMEN

MicroRNA-155 is highly expressed in many malignant tumors with poor prognosis, which regulates cell apoptosis, proliferation, invasion, metastasis, tumor angiogenesis, and metabolism. This study aims at investigating the clinical significance of miR-155 expression in human gliomas. Real-time quantitative PCR was used to detect the expression levels of miR-155 in 131 glioma and 16 normal brain tissues. The association of miR-155 expression with clinicopathological factors and prognosis of glioma patients were analyzed. The expression levels of miR-155 were significantly higher in glioma tissues than that in normal brain tissues (P < 0.001), which was associated with high pathological grade (P < 0.001) and low Karnofsky Performance Status score (P = 0.022). As a result of Kaplan-Meier survival and Cox regression analyses, overall survival (OS) rates and progression-free survival were significantly poorer in high-expression group relative to low-expression group (both P < 0.001). Furthermore, miR-155 expression was significantly associated with poor OS (P < 0.001) and PFS (P = 0.001) in glioma patients who had high pathological grades (III-IV) as calculated by subgroup analyses. These findings reveal that miR-155 expression might be an independent prognostic factor and a therapeutic target for human glioma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , MicroARNs/metabolismo , Adolescente , Adulto , Anciano , Apoptosis , Encéfalo/patología , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/patología , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Glioma/diagnóstico , Glioma/patología , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Pronóstico , Modelos de Riesgos Proporcionales , Resultado del Tratamiento , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...