Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
CNS Neurosci Ther ; 28(12): 2319-2330, 2022 12.
Article in English | MEDLINE | ID: mdl-36184801

ABSTRACT

AIMS: We aimed to investigate the role of receptor-interacting protein 2 (RIP2) in regulation of stemness of glioma cells and chemotherapy resistance. METHODS: Plasmid transfection was used to overexpress RIP2. Chemical inhibitors were used to inhibit RIP2 or NF-κB activity. Cancer stemness of glioma cells was investigated by sphere formation assays, clone formation assays, and xenograft tumor formation assays. The expression of RIP2, p-NF-κB, IκBα, CD133, or SOX-2 was detected by Western blotting and immunofluorescence. Apoptosis was detected by flow cytometry. Immunohistochemical staining was used to detect the expression of RIP2, CD133, and SOX-2 in xenograft tumor tissue. The effect of the RIP2/NF-κB pathway on temozolomide (TMZ) resistance was evaluated by xenograft tumor assay. RESULTS: Transfection with RIP2 plasmid enhanced the sphere formation capability of U251 cells, clone formation capability, and xenograft tumor formation capability. RIP2 could mediate TMZ resistance by upregulating the expression of CD133 and SOX-2 by activating the NF-κB pathway. Both RIP2 inhibitor GSK583 and the NF-κB inhibitor SC75741 could reverse the resistance of U251 cells to TMZ. CONCLUSION: RIP2 mediates TMZ resistance by regulating the maintenance of stemness in glioma cells through NF-κB. Interventions targeting the RIP2/NF-κB pathway may be a new strategy for TMZ-resistant gliomas.


Subject(s)
Brain Neoplasms , Glioma , Neoplastic Stem Cells , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Humans , Brain Neoplasms/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Glioma/metabolism , NF-kappa B/metabolism , Temozolomide/therapeutic use , Animals , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics
2.
Bioeng Transl Med ; 6(3): e10203, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34589592

ABSTRACT

Treatments of glioblastoma (GBM) have not been very effective, largely due to the inefficiency of drugs in penetrating the blood brain barrier (BBB). In this study, we investigated the potential of exosome-coated doxorubicin (DOX)-loaded nanoparticles (ENPDOX) in BBB penetration, inducing immunogenic cell death (ICD) and promoting survival of GBM-bearing mice. DOX-loaded nanoparticles (NPDOX) were coated with exosomes prepared from mouse brain endothelial bEnd.3 cells. ENPDOX cellular uptake was examined. Penetration of ENPDOX through the BBB was tested in an in vitro transwell system and a GBM mouse model. The effects of ENPDOX in inducing apoptosis and ICD were assessed. Finally, the efficacy of ENPDOX in the treatment of GBM-bearing mice was assessed. ENPDOX was taken up by bEnd.3 cells and could penetrate the BBB both in vitro and in vivo. In vitro, ENDDOX induced apoptosis and ICD of glioma GL261 cells. Systemic administration of ENPDOX resulted in maturation of dendritic cells, activation of cytotoxic cells, altered production of cytokines, suppressed proliferation and increased apoptosis of GBM cells in vivo and prolonged survival of GBM-bearing mice. Our findings indicate that ENPDOX may be a potent therapeutic strategy for GBM which warrants further investigation in clinical application.

3.
Pharm Biol ; 59(1): 904-911, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34225581

ABSTRACT

CONTEXT: The phosphorylation of signal transducer and activator of transcription protein 3 (STAT3) is up-regulated in glioblastoma (GBM) cells and is regulated by protein tyrosine phosphatase receptor type M (PTPRM). Fibronectin-1 (FN1) is also reported to be up-regulated in GBM. OBJECTIVE: We explored the role of FN1-induced PTPRM methylation in GBM. MATERIALS AND METHODS: The lentivirus particles of oe-PTPRM, sh-PTPRM, oe-FN1, sh-FN1, or their negative controls (NSCs) were transfected into GBM cells with or without stattic (0.5 µM, 24 h) or 5-aza (1 µM, 0, 2, 4 h) treatments. Methylation-specific PCR was performed to detect PTPRM methylation levels. RESULTS: PTPRM was down-regulated (0.373 ± 0.124- and 0.455 ± 0.109-fold), FN1 and p-STAT3 were up-regulated (p < 0.001) in A172 and U87 MG cells as compared to NSCs. Overexpressing PTPRM inhibited STAT3 phosphorylation. Interfering with PTPRM increased colony numbers in A172 and U-87 MG cells (2.253 ± 0.111- and 2.043 ± 0.19-fold), and stattic reduced them. Cell viability was reduced after treatment with 5-aza in A172 and U-87 MG cells (p < 0.05). P-STAT3 was down-regulated after 5-aza treatment. Overexpressing FN1 decreased PTPRM levels (p < 0.001), knockdown of FN1 decreased PTPRM methylation and inhibited STAT3 phosphorylation. Overexpressing FN1 increased cell viability (1.497 ± 0.114- and 1.460 ± 0.151-fold), and stattic or 5-aza reversed such effects (p < 0.05). DISCUSSION AND CONCLUSIONS: The up-regulation of FN1 reduced PTPRM by increasing its methylation, resulting in an increase of STAT3 phosphorylation and promoting GBM cell proliferation. Interfering with FN1 may be a potential therapeutic target for GBM.


Subject(s)
Brain Neoplasms/pathology , Fibronectins/genetics , Glioblastoma/pathology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/genetics , DNA Methylation , Glioblastoma/genetics , Humans , Phosphorylation/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , Up-Regulation
4.
CNS Neurosci Ther ; 27(5): 552-563, 2021 05.
Article in English | MEDLINE | ID: mdl-33460245

ABSTRACT

BACKGROUND: Temozolomide (TMZ) is a first-line chemotherapy drug for the treatment of malignant glioma and resistance to it poses a major challenge. Receptor-interacting protein 2 (RIP2) is associated with the malignant character of cancer cells. However, it remains unclear whether RIP2 is involved in TMZ resistance in glioma. METHODS: RIP2 expression was inhibited in TMZ-resistant glioma cells and normal glioma cells by using small interfering RNA (siRNA) against RIP2. Plasmid transfection method was used to overexpress RIP2. Cell counting kit-8 assays were performed to evaluate cell viability. Western blotting or immunofluorescence was performed to determine RIP2, NF-κB, and MGMT expression in cells. Flow cytometry was used to investigate cell apoptosis. TMZ-resistant glioma xenograft models were established to evaluate the role of the RIP2/NF-κB/MGMT signaling pathway in drug resistance. RESULTS: We observed that RIP2 expression was upregulated in TMZ-resistant glioma cells, whereas silencing of RIP2 expression enhanced cellular sensitivity to TMZ. Similarly, upon the induction of RIP2 overexpression, glioma cells developed resistance to TMZ. The molecular mechanism underlying the process indicated that RIP2 can activate the NF-κB signaling pathway and upregulate the expression of O6-methylguanine-DNA methyltransferase (MGMT), following which the glioma cells develop drug resistance. In the TMZ-resistant glioma xenograft model, treatment with JSH-23 (an NF-κB inhibitor) and lomeguatrib (an MGMT inhibitor) could enhance the sensitivity of the transplanted tumor to TMZ. CONCLUSION: We report that the RIP2/NF-κB/MGMT signaling pathway is involved in the regulation of TMZ resistance. Interference with NF-κB or MGMT activity could constitute a novel strategy for the treatment of RIP2-positive TMZ-resistant glioma.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , DNA Modification Methylases/drug effects , DNA Modification Methylases/genetics , DNA Repair Enzymes/drug effects , DNA Repair Enzymes/genetics , Drug Resistance, Neoplasm/genetics , Glioma/drug therapy , NF-kappa B/drug effects , NF-kappa B/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/drug effects , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Signal Transduction/drug effects , Temozolomide/pharmacology , Tumor Suppressor Proteins/drug effects , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/drug effects , Brain Neoplasms/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Humans , Male , Mice , Mice, Inbred BALB C , NF-kappa B/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Xenograft Model Antitumor Assays
5.
PLoS One ; 14(7): e0219904, 2019.
Article in English | MEDLINE | ID: mdl-31314786

ABSTRACT

Generalized seizures engage bilateral networks from their onset at a low temporal scale. Previous studies findings have demonstrated focal/local brain activity abnormalities in the patients with generalized tonic-clonic seizures (GTCS). Resting state functional magnetic resonance imaging (fMRI) allows the detection of aberrant spontaneous brain activity in GTCS. Little is known, however, about alterations of dynamics (temporal variability) of spontaneous brain activity. It also remains unclear whether temporal variability of spontaneous brain activity is associated with disease severity. To address these questions, the current study assessed patients with GTCS (n = 35), and age- and sex-matched healthy controls (HCs, n = 33) who underwent resting state fMRI. We first assessed the dynamics of spontaneous brain activity using dynamic amplitude of low-frequency fluctuation (dALFF). Furthermore, the temporal variability of brain activity was quantified as the variance of dALFF across sliding window. Compared to HCs, patients with GTCS showed hyper-temporal variability of dALFF in parts of the default mode network, whereas they showed hypo-temporal variability in the somatomotor cortex. Furthermore, dynamic ALFF in the subgenual anterior cingulate cortex was positively correlated with duration of disease, indicating that disease severity is associated with excessive variability. These results suggest both an excessive variability and excessive stability in patients with GTCS. Overall, the current findings from brain activity dynamics contribute to our understanding of the pathophysiological mechanisms of generalized seizure.


Subject(s)
Brain/physiopathology , Seizures/physiopathology , Adult , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Brain/diagnostic imaging , Brain Mapping , Electroencephalography , Female , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Seizures/diagnostic imaging , Seizures/drug therapy , Treatment Outcome , Young Adult
6.
Oncol Lett ; 17(6): 5754-5760, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31186801

ABSTRACT

Growth-arrest-associated long non-coding RNA (lncRNA) 1 (GASL1) is an lncRNA with a tumor suppression role in osteosarcoma, whereas its involvement in other malignancies is unknown. In the present study, tumor tissues and adjacent healthy tissues were collected from patients with glioma, and blood samples were collected from patients and healthy controls to detect the expression of GASL1. All patients were followed up for 5 years, and the diagnostic and prognostic values for glioma were evaluated by receiver operating characteristic curve analysis and survival curve analysis, respectively. Potential associations between serum GASL1 and clinicopathological data of patients with glioma were investigated using χ2 testing. A GASL1 expression vector and short hairpin RNA targeting GASL1 were transfected into glioma cells and the effects on TGF-ß1 expression and cell proliferation were investigated by western blotting and Cell Counting Kit-8 assay. Glioma tumor tissue exhibited significantly lower GASL1 expression compared with in adjacent healthy tissue. Serum levels of GASL1 were lower in patients compared with in healthy controls. Serum GASL1 was identified to be a sensitive biomarker for glioma cancer, and a low expression level of GASL1 was associated with a decreased postoperative survival rate. In glioma cell lines with GASL1 overexpression, TGF-ß1 expression was decreased and proliferation was inhibited. GASL1 knockdown in glioma cell lines led to increased TGF-ß1 expression and proliferation. TGF-ß1 treatment had no effect on GASL1 expression, but TGF-ß1 treatment partially rescued the inhibition of proliferation in cells overexpressing GASL1. Therefore, GASL1 may inhibit tumor growth of glioma by inactivating the TGF-ß signaling pathway.

7.
J Cell Mol Med ; 23(6): 4386-4394, 2019 06.
Article in English | MEDLINE | ID: mdl-30973678

ABSTRACT

Mounting evidence suggests the vital roles of long noncoding RNA (lncRNAs) in the glioma. However, the role of LINC00511 in gliomagenesis is still uncovered. Here, in this study, we aim to investigate the effects of LINC00511 on the glioma cancer phenotype and its deepgoing mechanism. Results indicated that LINC00511 was up-regulated in glioma tissues and cell lines, moreover its overexpression positively correlated with the poor prognosis and advanced pathological stages. For the upstream regulation, LINC00511 was epigenetically up-regulated by transcription factor specificity protein 1 (SP1). Gain and loss of functional experiments demonstrated that LINC00511 promoted the proliferation and invasion of glioma cells in vitro. The knockdown of LINC00511 repressed the tumour growth in vivo. Mechanistically, LINC00511 positively regulated the CCND2 expression via competitively sponging with miR-124-3p. Overall, our finding illuminates that LINC00511 is induced by SP1 and accelerates the glioma progression through targeting miR-124-3p/CCND2 axis, constructing the SP1/LINC00511/miR-124-3p/CCND2 axis.


Subject(s)
Biomarkers, Tumor/metabolism , Cyclin D2/metabolism , Gene Expression Regulation, Neoplastic , Glioma/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Sp1 Transcription Factor/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Movement , Cell Proliferation , Cyclin D2/genetics , Disease Progression , Follow-Up Studies , Glioma/genetics , Glioma/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prognosis , Sp1 Transcription Factor/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Cell Struct Funct ; 44(1): 29-39, 2019.
Article in English | MEDLINE | ID: mdl-30787206

ABSTRACT

PUM2, an RNA binding protein, is known to promote stem cell proliferation via repressing expressions of cell cycle genes. Similar with stem cells, malignant cells are characterized by unlimited proliferation and remote migration. However, roles of PUM2 in cancer development are controversial. Here, we investigated PUM2's role in glioblastoma development and its relationship with the cell cycle regulator BTG1. Immunoblotting and RT-qPCR were used to evaluate protein expression level and transcript level, respectively. ShRNAs were designed to knock down PUM2 and BTG1 expression. CCK-8 assay was used to evaluate cell viability. Cell migration assay and evasion assay were used to evaluate metastatic capability of glioblastoma cell. RNA pull-down assay and RNA immunoprecipitation assay were used to test the interaction between PUM2 and BTG1 3'UTR. PUM2 expression is elevated in glioblastoma tumor tissues as well as glioblastoma cell lines. PUM2 knockdown remarkably suppresses glioblastoma cell proliferation and migration. In addition, PUM2 knockdown increases BTG1 expression. RNA pull-down assay and RNA immunoprecipitation assay show PUM2 binds to BTG1 3'UTR directly. Furthermore, knockdown of BTG1 reverses the effect of PUM2 knockdown on glioblastoma cell proliferation and migration. Our results suggest that PUM2 promote glioblastoma development via repressing BTG1 expression.Key words: PUM2, BTG1, glioblastoma, cell proliferation, metastasis.


Subject(s)
Cell Movement/genetics , Glioblastoma/pathology , Neoplasm Proteins/genetics , RNA-Binding Proteins/metabolism , 3' Untranslated Regions/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Knockdown Techniques , Humans , Neoplasm Invasiveness
9.
Front Immunol ; 10: 3161, 2019.
Article in English | MEDLINE | ID: mdl-32117213

ABSTRACT

Glioma stem cell (GSC)-derived extracellular vesicles (EVs) can mediate the communication between GSCs and microglia. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) expression in GSCs, EVs, and supernatant was detected by real-time PCR. The direct targeting between MALAT1 and miR-129-5p, miR-129-5p, and HMGB1 were tested with luciferase reporter analysis. The expression and secretion of interleukin (IL)-6, IL-8, and tumor necrosis factor (TNF)-α were determined in lipopolysaccharide-stimulated microglia or miR-129-5p inhibitor transferred to microglia exposed to GSC EVs or EVs derived from siMALAT1 pre-transferred GSCs. MALAT1 was enriched in GSC EVs compared with GSCs, and up-regulated MALAT1 was also observed in microglia upon GSC EVs incubation. The relative expression and secretion of IL-6, IL-8, and TNF-α in lipopolysaccharide-stimulated microglia were up-regulated in the GSC supernatant group, which could be reversed by dimethyl amiloride (DMA) (EV secretion inhibitor) co-administration or si-MALAT1 pre-transfection of GSCs. Luciferase reporter assay testified the direct binding of MALAT1 and miR-129-5p, miR-129-5p, and HMGB1, and si-MALAT1 could up-regulate miR-129-5p expression and down-regulate HMGB1 expression in microglia cells. The concentration of IL-6, IL-8, and TNF-α in lipopolysaccharide-stimulated microglia exposed to EVs from siMALAT1 transfected GSCs could be up-regulated by miR-129-5p inhibition. EVs lncRNA MALAT1 released from GSCs could modulate the inflammatory response of microglia after lipopolysaccharide stimulation through regulating the miR-129-5p/HMGB1 axis.


Subject(s)
Brain Neoplasms/immunology , Glioma/immunology , MicroRNAs/immunology , Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/immunology , Tumor Escape/immunology , Brain Neoplasms/metabolism , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Gene Expression Regulation, Neoplastic/physiology , Glioma/metabolism , HMGB1 Protein/immunology , HMGB1 Protein/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Lipopolysaccharides/pharmacology , MicroRNAs/metabolism , Microglia/immunology , Microglia/metabolism , Neoplastic Stem Cells/immunology , RNA, Long Noncoding/metabolism , Signal Transduction/immunology
10.
Biosci Rep ; 39(1)2019 01 31.
Article in English | MEDLINE | ID: mdl-30498093

ABSTRACT

The present study was undertaken to investigate the underlying mechanisms of long noncoding RNA OIP5-AS1 via regulating miR-410 to modulate Wnt-7b in the progression of glioma. To address this problem, we measured the expression of OIP5-AS1 and miR-410 in glioma tissues by qRT-PCR. Glioma U87 cells were transfected with OIP5-AS1 siRNA or miR-410 inhibitors. The targeting relationships among miR-410, OIP5-AS1 and Wnt-7b were verified by luciferase reporter assays. Western blotting was employed to determine the expression of Wnt-7b/ß-catenin pathway-related proteins, while MTT, flow cytometry, Transwell assays and wound-healing assays were used to measure the biological characteristics of glioma cells. The results showed that OIP5-AS1 expression was higher and miR-410 was lower in glioma tissues. Luciferase reporter assays confirmed a targeting relationship between OIP5-AS1 and miR-410, as well as between miR-410 and Wnt-7b. Silencing OIP5-AS1 reduced cell proliferation, invasion and migration of glioma U87 cells and led to depressed expression levels of miR-410, Wnt-7b, p-ß-catenin, GSK-3ß-pS9, c-Myc and cyclin D1. Furthermore, down-regulation of OIP5-AS1 induced G0/G1 phase cell cycle arrest and apoptosis of glioma cells. Inhibitors of miR-410 abolished the biological effects of OIP5-AS1 siRNA in glioma cells. In vivo, OIP5-AS1 knockdown also inhibited tumor growth. Taken together, this research suggested that silencing OIP5-AS1 may specifically block the Wnt-7b/ß-catenin pathway via targeted up-regulating miR-410, thereby inhibiting growth, invasion and migration while promoting apoptosis in glioma cells.


Subject(s)
Brain Neoplasms/therapy , Gene Expression Regulation, Neoplastic , Glioma/therapy , RNA, Long Noncoding/genetics , Wnt Proteins/genetics , Adult , Animals , Apoptosis/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle/genetics , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Disease Progression , Female , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Neuroglia/metabolism , Neuroglia/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/genetics , beta Catenin/metabolism
11.
CNS Neurol Disord Drug Targets ; 18(1): 78-87, 2019.
Article in English | MEDLINE | ID: mdl-30394221

ABSTRACT

BACKGROUND AND OBJECTIVE: Exosomes communicate inter-cellularly and miRNAs play critical roles in this scenario. MiR-214-5p was implicated in multiple tumors with diverse functions uncovered. However, whether miR-214-5p is mechanistically involved in glioblastoma, especially via exosomal pathway, is still elusive. Here we sought to comprehensively address the critical role of exosomal miR-214-5p in glioblastoma (GBM) microenvironment. METHODS: The relative expression of miR-214-5p was determined by real-time PCR. Cell viability and migration were measured by MTT and transwell chamber assays, respectively. The secretory cytokines were measured with ELISA kits. The regulatory effect of miR-214-5p on CXCR5 expression was interrogated by luciferase reporter assay. Protein level was analyzed by Western blot. RESULTS: We demonstrated that miR-214-5p was aberrantly overexpressed in GBM and associated with poorer clinical prognosis. High level of miR-214-5p significantly contributed to cell proliferation and migration. GBM-derived exosomal miR-214-5p promoted inflammatory response in primary microglia upon lipopolysaccharide challenge. We further identified CXCR5 as the direct target of miR-214- 5p in this setting. CONCLUSION: Overexpression of miR-214-5p in GBM modulated the inflammatory response in microglia via exosomal transfer.


Subject(s)
Glioblastoma/metabolism , Inflammation/metabolism , MicroRNAs/metabolism , Microglia/metabolism , Receptors, CXCR5/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Survival/physiology , Cells, Cultured , Exosomes/metabolism , Glioblastoma/physiopathology , Humans , Inflammation/chemically induced , Interleukin-6/metabolism , Interleukin-8/metabolism , Lipopolysaccharides , Primary Cell Culture , Tumor Necrosis Factor-alpha/metabolism
12.
J Cell Mol Med ; 22(12): 6338-6344, 2018 12.
Article in English | MEDLINE | ID: mdl-30270508

ABSTRACT

Emerging evidence have illustrated the vital roles of long noncoding RNAs (lncRNAs) in glioma. Nevertheless, the majority of their roles and mechanisms in gliomagenesis are still largely unclear. In this study, we investigate the roles of lncRNA CASC9 on glioma tumourigenesis and authenticate its potential mechanisms. Results manifested that CASC9 was highly expressed in glioma specimens and cells, moreover, the ectopic overexpression was correlated with glioma patients' clinic. Functional studies found that siRNA-mediated CASC9 silencing inhibited the proliferative ability, invasion in vitro, and impaired the tumour growth in vivo. Mechanical studies revealed that miR-519d both targeted the 3'-UTR of CASC9 and STAT3 mRNA, which was identified by luciferase reporter assay and RNA immunoprecipitation (RIP). Moreover, chromatin immunoprecipitation (ChIP) and luciferase reporter assay revealed that STAT3, an oncogenic transcription factor, could bind with the promoter of CASC9 and activate its transcriptional level. In conclusion, our results concluded that CASC9 promotes STAT3 expression via sponging miR-519d, in return, STAT3 activate CASC9 transcription, forming a positive feedback loop of CASC9/miR-519d/STAT3. The novel finding provides a potential therapeutic target for glioma.


Subject(s)
Glioma/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , STAT3 Transcription Factor/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Glioma/pathology , Humans
13.
JAMA Neurol ; 75(11): 1338-1346, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30073290

ABSTRACT

Importance: Chronic subdural hematoma (CSDH) is a trauma-associated condition commonly found in elderly patients. Surgery is currently the treatment of choice, but it carries a significant risk of recurrence and death. Nonsurgical treatments remain limited and ineffective. Our recent studies suggest that atorvastatin reduces hematomas and improves the clinical outcomes of patients with CSDH. Objective: To investigate the safety and therapeutic efficacy of atorvastatin to nonsurgically treat patients with CSDH. Design, Setting, and Participants: The Effect of Atorvastatin on Chronic Subdural Hematoma (ATOCH) randomized, placebo-controlled, double-blind phase II clinical trial was conducted in multiple centers in China from February 2014 to November 2015. For this trial, we approached 254 patients with CSDH who received a diagnosis via a computed tomography scan; of these, 200 (78.7%) were enrolled because 23 patients (9.1%) refused to participate and 31 (12.2%) were disqualified. Interventions: Patients were randomly assigned to receive either 20 mg of atorvastatin or placebo daily for 8 weeks and were followed up for an additional 16 weeks. Main Outcomes and Measures: The primary outcome was change in hematoma volume (HV) by computed tomography after 8 weeks of treatment. The secondary outcomes included HV measured at the 4th, 12th, and 24th weeks and neurological function that was evaluated using the Markwalder grading scale/Glasgow Coma Scale and the Barthel Index at the 8th week. Results: One hundred ninety-six patients received treatment (169 men [86.2%]; median [SD] age, 63.6 [14.2] years). The baseline HV and clinical presentations were similar between patients who were taking atorvastatin (98 [50%]) and the placebo (98 [50%]). After 8 weeks, the HV reduction in patients who were taking atorvastatin was 12.55 mL more than those taking the placebo (95% CI, 0.9-23.9 mL; P = .003). Forty-five patients (45.9%) who were taking atorvastatin significantly improved their neurological function, but only 28 (28.6%) who were taking the placebo did, resulting in an adjusted odds ratio of 1.957 for clinical improvements (95% CI, 1.07-3.58; P = .03). Eleven patients (11.2%) who were taking atorvastatin and 23 (23.5%) who were taking the placebo underwent surgery during the trial for an enlarging hematoma and/or a deteriorating clinical condition (hazard ratio, 0.47; 95% CI, 0.24-0.92; P = .03). No significant adverse events were reported. Conclusions and Relevance: Atorvastatin may be a safe and efficacious nonsurgical alternative for treating patients with CSDH. Trial Registration: ClinicalTrials.gov Identifier: NCT02024373.


Subject(s)
Atorvastatin/pharmacology , Hematoma, Subdural, Chronic/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Outcome Assessment, Health Care , Aged , Atorvastatin/administration & dosage , Atorvastatin/adverse effects , China , Double-Blind Method , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Male , Middle Aged
14.
Oncol Lett ; 16(2): 2525-2532, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30013647

ABSTRACT

Radiation therapy is important for the comprehensive treatment of intracranial tumors. However, the molecular mechanisms underlying the pathogenesis of delayed cognitive dysfunction are not well-defined and effective treatments or prevention measures remain insufficient. In the present study, 60 adult male Wistar rats were randomly divided into three groups, which included a control, whole brain radiotherapy (WBRT) (single dose of 30 Gy of WBRT) and nimodipine (single dose of 30 Gy of WBRT followed by nimodipine injection intraperitoneally) groups. The rats were sacrificed 7 days or 3 months following irradiation. At 3 months, the Morris water maze test was used to assess spatial learning and memory function in rats. The results demonstrated that the WBRT group demonstrated a significantly impaired cognitive performance, decreased numbers of hippocampal Cornu Ammonis (CA)1 neurons and upregulated expression of caspase-3 in the dentate gyrus compared with those in the control and nimodipine groups. Reverse transcription-quantitative polymerase chain reaction analysis demonstrated that the WBRT group exhibited increased ratio of B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax)/Bcl-2 compared with that in control and nimodipine groups on day 7 following irradiation. However, the WBRT group exhibited decreased levels of brain-derived neurotrophic factor (BDNF) compared with that in control and nimodipine groups at 3 months following brain irradiation. The levels of growth-associated protein 43 and amyloid precursor protein between the nimodipine group and WBRT group were not statistically significant. The present study demonstrated that neuron apoptosis may lead to delayed cognitive deficits in the hippocampus, in response to radiotherapy. The cognitive impairment may be alleviated in response to a calcium antagonist nimodipine. The molecular mechanisms involved in nimodipine-mediated protection against cognitive decline may involve the regulation of Bax/Bcl-2 and BDNF in the hippocampus.

15.
Neurol Res ; 40(5): 340-363, 2018 May.
Article in English | MEDLINE | ID: mdl-29528274

ABSTRACT

Purpose Tethered cord syndrome (TCS) is the clinical manifestation of an abnormal stretch on the spinal cord, caused by several pathological conditions. Tethered cord release is the gold standard treatment for TCS. However, direct untethering carries potential risks of spinal cord injury, post-operative retethering, and CSF-related complications. Spine-shortening osteotomy (SSO) has recently been performed as an alternative technique to avoid these risks. We aimed to systematically review the literature on indications and outcome of SSO in TCS patients. Methods We searched PubMed, Embase, Google Scholar, and the Cochrane Library to identify all studies on SSO in TCS patients. We used random or fixed-effects models to calculate rates and 95% confidence intervals to establish the rates of clinical improvement in TCS patients performed with SSO. Sensitive analysis and metaregression were made to explore potential sources of heterogeneit. Results We identified six eligible surveys with a total population of 57. Rates ranged from 62 to 88% for neurological deficits improvement, 80-100% for motor function improvement, 60-96% for pain or numbness scores improvement, 13-67% for sensory function improvement, and 79-100% for urinary and bowel dysfunction improvement. We noted substantial heterogeneity in rate estimates for motor function and urinary and bowel dysfunction improvement (all Cochran's χ² significant at P < 0.001; I² = 78.11%, 95%CI 61-94%; 84.28%, 18-100%; respectively). Conclusion SSO is a safe and effective technique for TCS patients, especially in more challenging cases, such as complex malformations or revision surgery. However, future cohort studies and randomized studies with large numbers and the power to provide illumination for the surgical decision-making of TCS are warranted.


Subject(s)
Neural Tube Defects/surgery , Osteotomy , Spine/surgery , Humans
16.
World Neurosurg ; 110: 460-474.e5, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29138073

ABSTRACT

BACKGROUND: Posterior fossa decompression without (PFD) or with duraplasty (PFDD) for the treatment of type 1 Chiari malformation (CM-1) is controversial. We thus performed a systematic review and meta-analysis of studies to assess the effect on clinical and imaging improvement, operative time, complications, and recurrence rate between PFD and PFDD in patients with CM-1. METHODS: We systematically searched PubMed, Embase, Cochrane, Web of Knowledge, and ClinicalTrials.gov for retrospective or prospective studies comparing PFD with PFDD. Our main end points were clinical and imaging improvement, operative time, complications, and recurrence rate. We assessed pooled data by use of a fixed-effects or random-effects model according to the between-study heterogeneity. RESULTS: Of 214 identified studies, 13 were eligible and were included in our analysis (N = 3481 patients). Compared with PFD, PFDD led to a mean greater increase in operative time than did PFD [standardized mean difference, -2.35; 95% confidence interval [CI], (-2.70 to -1.99)], a higher likelihood of clinical improvement in patients with syringomyelia (relative risk [RR], 0.70; 95% CI, 0.49-0.98), no increased RR of clinical improvement in patients without syringomyelia, no increased RR of imaging improvement, but an increased RR of cerebrospinal fluid-related complications (RR, 0.29; 95% CI, 0.15-0.58), cerebrospinal fluid leak, aseptic meningitis, pseudomeningocele, and a decreased likelihood of recurrence rate. CONCLUSIONS: PFDD can be an optimal surgical strategy because of its higher clinical improvement and lower recurrence rate in the patients with syringomyelia. In patients without syringomyelia, PFD can be a preferred choice because of its similar clinical improvement and lower costs. Future randomized studies with large numbers and the power to provide illumination for surgical decision making in CM-1 are warranted.


Subject(s)
Arnold-Chiari Malformation/surgery , Cranial Fossa, Posterior/surgery , Decompression, Surgical/methods , Dura Mater/surgery , Arnold-Chiari Malformation/complications , Humans , Recurrence , Syringomyelia/complications , Syringomyelia/surgery
17.
Biochem Biophys Res Commun ; 494(3-4): 674-680, 2017 12 16.
Article in English | MEDLINE | ID: mdl-29066350

ABSTRACT

ZMYND11 (zinc finger MYND-type containing 11) has been widely regarded to be involved in a variety of cancers as a potential suppressor. However, the biological role and mechanism of ZMYND11 in glioblastoma multiform (GBM) remain unknown. In this study, we found that ZMYND11 expression was remarkably decreased in GBM tissues from 20 cases and cell line (U87) compared to normal brain tissue from 10 cases (P < 0.001). Furthermore, we explored that ZMYND11 upregulation significantly suppressed U87 cells proliferation and invasion, induced cell cycle arrest and apoptosis in vitro. Subsequently, we identified increased ZMYND11 inhibited the tumor growth using tumor cells xenograft experiment on rude mice. Moreover, we explored that ZMYND11 was a new direct and functional target of miR-196a-5p in U87 via luciferase reporter assay. In addition, we confirmed the negative correlation between miR-196a-5p and ZMYND11 in GBM tissue and U87 cells by changing the expression level of miR-196a-5p with lentivirus and plasmid vector. Furthermore, we demonstrated that decreased ZMYND11 could reverse suppressive effect of downregulated miR-196a-5p on U87 by rescue experiment. Taken together, ZMYND11 was demonstrated to be a potential and extremely promising suppressor of GBM, while miRNA-196a-5p was quite an important target of treatment of GBM.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Carrier Proteins/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , MicroRNAs/metabolism , Adult , Aged , Animals , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Co-Repressor Proteins , DNA-Binding Proteins , Down-Regulation , Female , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness
18.
Mol Ther Nucleic Acids ; 8: 482-492, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28918048

ABSTRACT

Recombinant tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), as a novel cancer therapeutic, is being tested in phase II and III clinical trials; however, TRAIL resistance remains a big obstacle preventing its clinical application. Considering that TRAIL-induced apoptosis through death receptors DR4 and DR5, their activation may be an alternative pathway to suppress TRAIL resistance. In this study, a negative correlation between DR5 expression and TRAIL resistance was observed, and miR-133a was predicted to be the most promising candidate to suppress DR5 expression. Further investigation demonstrated that miR-133a knockdown dramatically suppressed TRAIL resistance in glioblastoma in vitro and in vivo. An NF-κB family member, phosphorylated IκBα (P-IκBα), was shown to be stimulated by miR-133a, leading to the activation of this signaling. Finally, miR-133a was found to be inversely correlated with DR5 expression in human clinical specimens. In conclusion, our data demonstrate that miR-133a promotes TRAIL resistance in glioblastoma by suppressing DR5 expression and activating NF-κB signaling.

19.
Ther Adv Med Oncol ; 9(12): 741-754, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29449895

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM) is the most aggressive and deadly primary brain cancer that arises from astrocytes and classified as grade IV. Recently, exosomes have been reported as an essential mediator in diverse cancer carcinogenesis and metastasis. However, their role in GBM is still unclear. In this study, we aimed to investigate whether blood exosomes can be potential clinical diagnostic markers for GBM. METHODS: We used a xenograft orthotopic mouse model to detect the differentially expressed genes in the brain and blood exosomes of original/recurrent GBM. RESULTS: We found that recurrent GBM had stronger growth capacity and lethality than original GBM in the mouse model. A gene microarray of original tumors and blood exosomes from GBM orthotopic xenografts results showed that DNM3, p65 and CD117 expressions increased, whereas PTEN and p53 expressions decreased in both original tumors and blood exosomes. In the recurrent GBM tumor model, DNM3 and p65 showed increased expressions, whereas ST14 and p53 showed decreased expressions in tumor and blood exosomes of the recurrent GBM mouse model. CONCLUSION: In summary, we found that DNM3, p65 and p53 had a similar trend in brain and blood exosomes both for original and recurrent GBM, and could serve as potential clinical diagnostic markers for GBM.

20.
J Neurooncol ; 131(2): 255-265, 2017 01.
Article in English | MEDLINE | ID: mdl-27837435

ABSTRACT

MicroRNA is an important regulator of glioblastoma. This study aims at validating microRNA-221 (miR-221) as a biomarker for glioblastoma, and understanding how miR-221 regulates glioblastoma progression. Using clinical samples, miR-221 expression was analyzed by quantitative reverse-transcriptase PCR (qPCR). SHG-44 cells were treated with anti-miR-221 or U87MG-derived exosomes followed by monitoring changes in cell viability, migration and temozolomide (TMZ) resistance. Bioinformatics approach was used to identify targets of miR-221. The interaction between miR-221 and its target, DNM3 gene, was studied with dual-luciferase reporter assay, Spearman's correlation analysis, and western blotting. To verify that RELA regulates miR-221 expression, RELA-expressing vector or shRNA was introduced into SHG-44 cells and its effect on miR-221 expression was monitored. Both tissue-level and exosomal miR-221 expression increased with glioma grades. In SHG-44 cells, downregulating miR-221 expression inhibited cell proliferation, migration, and TMZ resistance, whereas incubation with U87MG-derived exosomes exerted tumor-promoting effects. DNM3 gene is a target of miR-221. RELA induced miR-221 expression. In glioma, elevated miR-221 expression is a biomarker for glioma. DNM3 is a target of miR-221 and RELA regulates miR-221 expression. The RELA/miR-221 axis is a target for glioma diagnosis and therapy.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/metabolism , Dacarbazine/analogs & derivatives , Dynamin III/metabolism , Glioma/metabolism , MicroRNAs/metabolism , Transcription Factor RelA/metabolism , Apoptosis , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation , Cell Survival , Dacarbazine/therapeutic use , Disease Progression , Drug Resistance, Neoplasm , Exosomes/metabolism , Gene Expression Regulation, Neoplastic , Glioma/drug therapy , Humans , Temozolomide
SELECTION OF CITATIONS
SEARCH DETAIL
...