Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Front Neurol ; 14: 1235312, 2023.
Article in English | MEDLINE | ID: mdl-37849835

ABSTRACT

Objective: This study aimed to analyze the relationship between the rate of morphological changes and intracranial aneurysm rupture during the cardiac cycle. Methods: Eighty-four patients with intracranial aneurysms were retrospectively analyzed and divided into the rupture (42 cases) and unruptured (42 cases) groups. Four-dimensional computed tomography angiography (4D-CTA) was performed to collect quantitative parameters of aneurysm morphology and calculate the morphological change rate. The potential factors associated with aneurysm rupture were determined by comparing the general clinical data and rate of change in the location and morphology of the aneurysm between the two groups. Results: Each morphological change rate in the rupture group was generally higher than that of the unruptured group. The rate of dome height change and aneurysm volume change were independent factors associated with aneurysm rupture. ROC curve analysis revealed that the diagnostic accuracy of the aneurysm volume change rate was higher. When the volume change rate was 12.33%, the sensitivity and specificity of rupture were 90.5 and 55.8%, respectively. Conclusion: The rate of change in dome height and volume of intracranial aneurysms during one cardiac cycle were independent factors associated with aneurysm rupture.

2.
Cell Biol Int ; 47(4): 776-786, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36576012

ABSTRACT

Gliomas account for about 80% of malignant brain tumors. The incidence of a new brain tumor is 6.4 per 100,000 persons per year with an overall 5-year survival rate of 33.4%. Regardless of the great advances that have been made in recent years, the causes and pathogenesis of glioma remain unclear. Here we study how phosphoglycerate mutase 4 (PGAM4) contributes to glioma. Using a variety of methods to examine glioma cell viability, proliferation, apoptosis, glycolysis, as well as ChIP coanalysis with modified histone H3, we showed that PGAM4 was significantly upregulated in patients with glioma and associated with poor survival. Silencing PGAM4 attenuated cell viability, proliferation, and glycolysis in T98G cells and suppressed tumor growth in vivo, while overexpressing PGAM4 promoted cell viability, proliferation, and glycolysis in U251 cells via regulating glycolysis pathway. Study also revealed that PGAM4 was regulated by EP300-mediated modifications of H3K27ac. PGAM4 silencing inhibited cell viability and proliferation, suppressed tumor growth, and decreased chemoresistance to temozolomide in glioma cells through suppressing glycolysis.


Subject(s)
Brain Neoplasms , Glioma , Humans , Temozolomide/pharmacology , Phosphoglycerate Mutase/metabolism , Drug Resistance, Neoplasm , Glioma/metabolism , Brain Neoplasms/metabolism , Apoptosis , Glycolysis , Cell Line, Tumor , Cell Proliferation
3.
Transl Stroke Res ; 14(6): 987-1001, 2023 12.
Article in English | MEDLINE | ID: mdl-36418735

ABSTRACT

NLRP3 inflammasomes have been reported to be an essential mediator in the inflammatory response during early brain injury (EBI) following subarachnoid hemorrhage (SAH). Recent studies have indicated that NLRP3 inflammasome-mediated pyroptosis and long non-coding RNA (lncRNA) H19 can participate in the inflammatory response. However, the roles and functions of lncRNA H19 in NLRP3 inflammasome-mediated pyroptosis during EBI after SAH are unknown and need to be further elucidated. NLRP3 inflammasome proteins were significantly elevated in CSF of human with SAH induced EBI and presented a positive correlation with severity. In ipsilateral hemisphere cortex of rats, these NLRP3 inflammasome proteins were also increased and accompanied with upregulation of H19, and both of NLRP3 and H19 were peaked at 24 h after SAH. However, knockdown of H19 markedly decreased the expression of NLRP3 inflammasome proteins at 24 h after SAH in rats and also ameliorated EBI, showing improved neurobehavioral deficits, cerebral edema, and neuronal injury. Moreover, knocking down of H19 downregulated the expression of Gasdermin D (GSDMD) in microglia in SAH rats. Similarly, knockdown of H19 also alleviated OxyHb-induced pyroptosis and NLRP3-mediated inflammasomes activation in primary microglia. Lastly, H19 competitively sponged with rno-miR-138-5p and then upregulated NLRP3 expression in the post-SAH inflammatory response. lncRNA H19 promotes NLRP3-mediated pyroptosis by functioning as rno-miR-138-5p sponge in rats during EBI after SAH, which might provide a potential therapeutic target for post-SAH inflammation regulation.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Subarachnoid Hemorrhage , Rats , Humans , Animals , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis , RNA, Long Noncoding/genetics , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/complications , MicroRNAs/genetics , MicroRNAs/therapeutic use
4.
Front Aging Neurosci ; 14: 857521, 2022.
Article in English | MEDLINE | ID: mdl-35783143

ABSTRACT

Background: Timely and accurate prediction of delayed cerebral ischemia is critical for improving the prognosis of patients with aneurysmal subarachnoid hemorrhage. Machine learning (ML) algorithms are increasingly regarded as having a higher prediction power than conventional logistic regression (LR). This study aims to construct LR and ML models and compare their prediction power on delayed cerebral ischemia (DCI) after aneurysmal subarachnoid hemorrhage (aSAH). Methods: This was a multicenter, retrospective, observational cohort study that enrolled patients with aneurysmal subarachnoid hemorrhage from five hospitals in China. A total of 404 aSAH patients were prospectively enrolled. We randomly divided the patients into training (N = 303) and validation cohorts (N = 101) according to a ratio of 75-25%. One LR and six popular ML algorithms were used to construct models. The area under the receiver operating characteristic curve (AUC), accuracy, balanced accuracy, confusion matrix, sensitivity, specificity, calibration curve, and Hosmer-Lemeshow test were used to assess and compare the model performance. Finally, we calculated each feature of importance. Results: A total of 112 (27.7%) patients developed DCI. Our results showed that conventional LR with an AUC value of 0.824 (95%CI: 0.73-0.91) in the validation cohort outperformed k-nearest neighbor, decision tree, support vector machine, and extreme gradient boosting model with the AUCs of 0.792 (95%CI: 0.68-0.9, P = 0.46), 0.675 (95%CI: 0.56-0.79, P < 0.01), 0.677 (95%CI: 0.57-0.77, P < 0.01), and 0.78 (95%CI: 0.68-0.87, P = 0.50). However, random forest (RF) and artificial neural network model with the same AUC (0.858, 95%CI: 0.78-0.93, P = 0.26) were better than the LR. The accuracy and the balanced accuracy of the RF were 20.8% and 11% higher than the latter, and the RF also showed good calibration in the validation cohort (Hosmer-Lemeshow: P = 0.203). We found that the CT value of subarachnoid hemorrhage, WBC count, neutrophil count, CT value of cerebral edema, and monocyte count were the five most important features for DCI prediction in the RF model. We then developed an online prediction tool (https://dynamic-nomogram.shinyapps.io/DynNomapp-DCI/) based on important features to calculate DCI risk precisely. Conclusions: In this multicenter study, we found that several ML methods, particularly RF, outperformed conventional LR. Furthermore, an online prediction tool based on the RF model was developed to identify patients at high risk for DCI after SAH and facilitate timely interventions. Clinical Trial Registration: http://www.chictr.org.cn, Unique identifier: ChiCTR2100044448.

5.
Cell Biol Int ; 46(5): 819-828, 2022 May.
Article in English | MEDLINE | ID: mdl-35178826

ABSTRACT

The incidence of gliomas is increasing. Although great progress in glioma treatment has been made, the clinical outcome remains unsatisfactory. Chemokine (C-C motif) ligand 2 (CCL2) plays a key role in different types of cancers, including glioma. However, the function of CCL2 in glioma chemoresistance is not fully understood. In the current study, CCL2 was significantly upregulated in glioma. More importantly, CCL2 and CCR2 were significantly upregulated in temozolomide (TMZ)-resistant glioma. TMZ-resistant malignant glioblastoma cells (U251/TMZ) had higher expressions of CCL2 and CCR2 and a higher level of glycolysis as compared to its parental cell line U251. Silencing of CCL2 in U251/TMZ cells inhibited glycolysis. Overexpression of CCL2 reduced TMZ-induced apoptosis through activation of the AKT pathway and promotion of glycolysis. Moreover, overexpression of CCL2 significantly reduced the antitumor effect of TMZ in vivo. In conclusion, CCL2 overexpression reduced the antitumor effect of TMZ by enhancing glycolysis through activation of AKT signaling. The findings highlighted the importance of CCL2/CCR2/glycolysis and its potential value in developing new treatment for glioma.


Subject(s)
Brain Neoplasms , Glioma , Apoptosis , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Chemokine CCL2/metabolism , Drug Resistance, Neoplasm , Glioma/metabolism , Glycolysis , Humans , Proto-Oncogene Proteins c-akt/metabolism , Temozolomide/pharmacology , Temozolomide/therapeutic use
6.
J Oncol ; 2022: 6093216, 2022.
Article in English | MEDLINE | ID: mdl-36618071

ABSTRACT

Background: Paeoniflorin is an active component of a widely used traditional Chinese medicine with antitumor activity through ferroptosis induction. It has been reported recently that ferroptosis is emerging in certain types of cancer; however, its relevance in glioma is still not well studied. Methods: CCK8 assay was performed for cell proliferation. Expression of mRNA and protein was tested by qPCR and western blot, respectively. Clinical section samples were detected by IHC. The relationship between NEDD4L and STAT3 was validated by a coimmunoprecipitation assay. Apoptosis was identified by TUNEL assay. A xenograft mouse model was utilized to validate the potential of paeoniflorin toward glioma cancer cells. Results: The data suggested that paeoniflorin could increase NEDD4L expression in glioma cells. The NEDD4L expression level was lower in glioma cancer tissues compared to adjacent normal tissues, and it correlates with poor prognosis. Meanwhile, NEDD4L mediates the ubiquitination of STAT3. Furthermore, increased NEDD4L significantly inhibited cell viability and induced accumulation of intracellular ROS levels, accompanied by decreased expression of key ferroptosis factors Nrl2 and GPX4, while NEDD4L knockdown had a reverse effect, suggesting that ferroptosis could be involved. NEDD4L-induced ferroptosis could be rescued by forced expression of STAT3. A xenograft nude mouse model showed that paeoniflorin inhibits tumor growth and further sensitizes glioma cells to RSL3, another well-known ferroptosis inducer. Conclusions: In summary, this study demonstrated that paeoniflorin might function as an effective drug for glioma by inducing ferroptosis via upregulation of NEDD4L and repression of Nrl2, GPX4, and STAT3.

7.
Exp Ther Med ; 22(6): 1391, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34650639

ABSTRACT

Subarachnoid hemorrhage (SAH) is a condition with a high associated mortality rate that is caused by hemorrhagic stroke. Activated protein C (APC) serves a neuroprotective role in central nervous system diseases. However, its role in SAH remains unclear. The present study aimed to investigate the role of APC and its regulatory mechanism in SAH. The SAH rat model was constructed through internal carotid artery puncture, while the SAH cell model was established via the application of oxygenated hemoglobin. ELISA was performed to detect the level of cytokines, and flow cytometry was used to determine the population of pyroptotic cells. Reverse transcription-quantitative PCR and western blotting were used to examine the relative mRNA and protein levels of APC. APC was silenced using specific APC short hairpin RNA. Neurological functions of rats were estimated using modified Garcia scoring and the balance beam test, while SAH was estimated using modified Sugawara's scoring. The results demonstrated that the expression of APC was significantly decreased, whereas the expression of NLR family pyrin domain-containing 3 (NLRP3) was increased in the SAH rat model in a time-dependent manner. The application of APC recombinant protein 3K3A-APC could significantly ameliorate SAH and improve neurological functions. In addition, 3K3A-APC could inhibit pyroptosis in a dose-dependent manner in the SAH cell model. Moreover, the NLRP3 inhibitor BAY11-7082 could reverse the upregulation of pyroptosis induced by APC-knockdown. Overall, the present study revealed that APC could ameliorate SAH-induced early brain injury by suppressing pyroptosis via inhibition of the NLRP3 inflammasome, which could provide a novel strategy for the treatment of SAH.

8.
Gastroenterol Res Pract ; 2021: 6655900, 2021.
Article in English | MEDLINE | ID: mdl-34221007

ABSTRACT

AIM: Irritable bowel syndrome (IBS) is a multifactorial functional bowel disorder characterized by disruption of the intestinal barrier. Circulating exosomal microRNAs (miRNAs) are involved in regulating epithelial barrier function, and upregulation of miR-148b-5p has been detected in IBS. However, whether exosomal miR-148-5p is involved in the IBS pathogenesis remains unclear. This study was aimed at investigating the relationship of exosomal miR-148-5p with colonic epithelial permeability. METHODS: Exosomes were isolated from the serum of IBS patients and healthy controls. HT-29 cells were cultured with the IBS-derived serum exosomes (IBS-exo). Exosome uptake assay was used to evaluate whether the IBS-exo could be absorbed by HT-29 cells. FITC-Dextran flux and transepithelial/endothelial electrical resistance were measured to evaluate epithelial permeability. A luciferase reporter assay was used to determine whether the regulator of G protein signaling- (RGS-) 2 is a target gene of miR-148b-5p. RESULTS: miR-148b-5p was obviously elevated in the IBS-exo compared to the control-exo. Upregulation of miR-148b-5p was observed in the HT-29 cells cultured with IBS-exo. Exposure to IBS-exo increased cell permeability and decreased RGS2 expression. The IBS-exo-induced alterations were obviously reversed by interfering with the miR-148b-5p expression. Mimicking the IBS-exo treatment, miR-148b-5p overexpression increased cell permeability and downregulated RGS2 expression, which were abrogated by overexpressing RGS2. The luciferase reporter assay revealed that RGS2 was a direct target of miR-148b-5p. CONCLUSIONS: Serum-derived exosomes from IBS patients increase colonic epithelial permeability via miR-148b-5p/RGS2 signaling.

9.
Cell Biol Int ; 45(10): 2150-2158, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34288252

ABSTRACT

Glioblastoma is an aggressive type of brain cancer with an extremely poor prognosis. Additionally, the F-box WD repeat-containing protein 7 (FBXW7) is a component of the ubiquitin-proteasome system that has been widely implicated in human cancers. In this study, we investigated the role and mechanism of FBXW7 in glioblastoma. FBXW7 expression was analyzed in normal and glioblastoma tissue samples using The Cancer Genome Atlas Glioblastoma Multiforme (TCGA-GBM) database. Then, quantitative reverse transcription-polymerase chain reaction (RT-PCR) was used to examine mRNA expression, whereas, western blot analysis was conducted to determine protein levels of the samples. Furthermore, cell apoptosis was assessed using the Annexin V staining method, followed by flow cytometry analysis. Immunoprecipitation (IP) assay was conducted as well to test protein-protein interactions. Lastly, protein expression in tissues was examined by conducting immunohistochemistry (IHC). Results showed that the glioblastoma tissue samples displayed an FBXW7 downregulation compared with normal tissues. In vitro, the overexpression of FBXW7 in glioblastoma cells induced apoptosis, whereas, its knockdown displayed the opposite effect. Mechanistically, FBXW7 interacted with HDAC7 to promote HDAC7 ubiquitination, however, the overexpression of HDAC7 in glioblastoma cells blocked FBXW7-induced apoptosis. Finally, FBXW7 and HDAC7 displayed an inverse correlation in glioblastoma tissues in vivo. Therefore, our data demonstrated an important function of FBXW7 in promoting glioblastoma apoptosis by interacting with HDAC7 and promoting HDAC7 ubiquitination.


Subject(s)
Apoptosis , Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , F-Box-WD Repeat-Containing Protein 7/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Histone Deacetylases/metabolism , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Case-Control Studies , Cell Proliferation , F-Box-WD Repeat-Containing Protein 7/genetics , Glioblastoma/genetics , Glioblastoma/metabolism , Histone Deacetylases/genetics , Humans , Prognosis , Tumor Cells, Cultured , Ubiquitination
10.
J Biosci ; 44(4)2019 Sep.
Article in English | MEDLINE | ID: mdl-31502574

ABSTRACT

The antitumor effect of calycosin has been widely studied, but the targets of calycosin against glioblastomas are still unclear. In this study we focused on revealing c-Met as a potential target of calycosin suppressing glioblastomas. In this study, suppressed-cell proliferation and cell invasion together with induced-cell apoptosis appeared in calycosin-treated U251 and U87 cells. Under treatment of calycosin, the mRNA expression levels of Dtk, c-Met, Lyn and PYK2 were observed in U87 cells. Meanwhile a western blot assay showed that c-Met together with matrix metalloproteinases-9 (MMP9) and phosphorylation of the serine/threonine kinase AKT (p-AKT) was significantly down-regulated by calycosin. Furthermore, overexpressed c-Met in U87 enhanced the expression level of MMP9 and p-AKT and also improved cell invasion. Additionally, the expression levels of c-Met, MMP9 and p-AKT were inhibited by calycosin in c-Met overexpressed cells. However, an AKT inhibitor (LY294002) only effected on MMP9 and p-AKT, not on c-Met. These data collectively indicated that calycosin possibility targeting on c-Met and exert an anti-tumor role via MMP9 and AKT.


Subject(s)
Glioblastoma/drug therapy , Isoflavones/pharmacology , Matrix Metalloproteinase 9/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-met/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Chromones/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Morpholines/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects
11.
Oncol Res ; 27(7): 819-826, 2019 Jul 12.
Article in English | MEDLINE | ID: mdl-30940290

ABSTRACT

Human glioblastoma multiforme (GBM) accounts for the majority of human brain gliomas. Several TMEM proteins, such as TMEM 45A, TMEM 97, and TMEM 140, are implicated in human brain gliomas. However, the roles of TMEM168 in human GBM remain poorly understood. Herein we found that mRNA levels of TMEM168 were overexpressed in GBM patients (n = 85) when compared with healthy people (n = 10), which was also supported by data from The Cancer Genome Atlas (TCGA). Kaplan-Meier analysis of Gene Expression Omnibus dataset GSE16011 suggested that enhanced TMEM168 expression was associated with shorter survival time. To investigate whether and how TMEM168 functioned in the tumorigenesis of human GBM cells, two human GBM cell lines (U87 and U373) were used for study. Lithium chloride (LiCl), an activator for Wnt/ß-catenin pathway, was used for the treatment. Our data suggested that siRNA-TMEM168 (siTMEM168) prevented viability of U87 and U373 cells, induced cell cycle arrest (G0/G1 phase) and promoted apoptosis, and the mechanisms involved in blocking Wnt/ß-catenin pathway, as evidenced by reducing expression of ß-catenin, C-myc, cyclin D1, and survivin. Furthermore, the inhibited effect of siTMEM168 on human GBM cell growth was significantly alleviated with additional LiCl treatment, substantiating the involvement of the Wnt/ß-catenin pathway in this process. In summary, our data demonstrated that TMEM168 may represent a therapeutic target for the treatment of human GBM.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Carcinogenesis/genetics , Genes, Tumor Suppressor/physiology , Glioblastoma/genetics , Glioblastoma/pathology , Membrane Proteins/genetics , Case-Control Studies , Cell Line, Tumor , Cell Proliferation/physiology , Female , Gene Knockdown Techniques , Humans , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Suppression, Genetic , Wnt Signaling Pathway/genetics , beta Catenin/genetics
12.
Onco Targets Ther ; 12: 41-49, 2019.
Article in English | MEDLINE | ID: mdl-30613151

ABSTRACT

BACKGROUND: MicroRNA-562 (miR-562) has been found to possess anti-cancer function in certain tumors. However, the function of miR-562 in glioblastoma (GBM) is still not fully understood. PURPOSE: The aim at present study is to analyze the function of miR-562 and its possible target in GBM cells. PATIENTS AND METHODS: In the present study, a total of 80 GBM samples and 16 adjacent noncancerous tissues were used to examine the expression of miR-562 and c-MET. In order to gain a deep insight into the molecular network of miR-562 and c-MET in GBM, the miR-562 mimic and inhibitor were transfected into two GBM cell lines (U251 and U87), respectively. Meanwhile, lentiviral vector was used to mediate overexpression of c-MET. Cell proliferation was examined via Cell Counting Kit-8 (CCK-8) assays. Meanwhile, cell apoptosis was analyzed by Annexin V-FTTC/PI staining assay. RESULTS: Our results indicated that the level of miR-562 was downregulated in GBM tissues and the expression of c-MET was upregulated in tumors. Cell proliferation analysis indicated that miR-562 was an anti-proliferation effector in GBM cells. Moreover, cell apoptosis analysis suggested the pro-apoptosis function of miR-562 in GBM cells. CONCLUSION: Our results demonstrated that miR-562 negatively regulated the c-MET/AKT signal pathway. In addition, caspase-3 might also serve as another target for miR-562 in GBM cells. This research not only obtained a deep understanding of miR-562 but also provided evidence in terms of developing new prognostic biomarker for GBM.

13.
Pathol Res Pract ; 214(9): 1330-1339, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30033329

ABSTRACT

Glioblastoma(GBM) is most common brain tumor in adults. Currently standard treatments have limited effect to increase the survival, because there are still largely unclear mechanisms in glioblastoma development. miR-223 was involved in various types of cancer, however, the function of miR-223-3p in GBM was still unclear. In our study, real-time PCR was performed to exam the expression level of miR-223-3p and NLRP3 (Nucleotide-binding oligomerization domain(NOD)-like receptor family PYRIN domain containing-3) in GBM tissues. Following that, mimic or inhibitor of miR-223-3p were used to modulate miR-223-3p expression in GBM cell lines respectively. Then, we analyzed cell proliferation and migration by cell counting kit and transwell assay. Further, western blot was performed to detect several inflammation-associated cytokines level in GBM cell lines. We found that miR-223-3p was decreased but NLRP3 was increased in GBM tissues. Treatment with miR-223-3p mimic inhibits cell proliferation and migration via decreasing several inflammation-associated cytokines, including interleukin-1ß (IL-1ß), monocyte chemoattractant protein-1 (MCP-1), IL-8 and IL-18. Importantly, these effects induced by miR-223-3p could be attenuated by NLRP3 overexpression, which was considered as one of target genes of miR-223-3p. In conclusion, these results indicated that miR-223-3p might act as a suppressor and a potential therapy target of GBM.


Subject(s)
Brain Neoplasms/pathology , Cytokines/biosynthesis , Glioblastoma/pathology , MicroRNAs/metabolism , Adult , Aged , Cell Movement/genetics , Cell Proliferation/genetics , Cytokines/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Inflammation/metabolism , Male , Middle Aged , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis
14.
Neurochem Res ; 43(3): 760-774, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29423667

ABSTRACT

Paeoniflorin (PF) is a polyphenolic compound derived from Radix Paeoniae Alba thathas anti-cancer activities in a variety of human malignancies including glioblastoma. However, the underlying mechanisms have not been fully elucidated. Epithelial to mesenchymal transition (EMT), characterized as losing cell polarity, plays an essential role in tumor invasion and metastasis. TGFß, a key member of transforming growth factors, has been demonstrated to contribute to glioblastoma aggressiveness through inducing EMT. Therefore, the present studies aim to investigate whether PF suppresses the expression of TGFß and inhibits EMT that plays an important role in anti-glioblastoma. We found that PF dose-dependently downregulates the expression of TGFß, enhances apoptosis, reduces cell proliferation, migration and invasion in three human glioblastoma cell lines (U87, U251, T98G). These effects are enhanced in TGFß siRNA treated cells and abolished in cells transfected with TGFß lentiviruses. In addition, other EMT markers such as snail, vimentin and N-cadherin were suppressed by PF in these cell lines and in BALB/c nude mice injected with U87 cells. The expression of MMP2/9, EMT markers, are also dose-dependently reduced in PF treated cells and in U87 xenograft mouse model. Moreover, the tumor sizes are reduced by PF treatment while there is no change in body weight. These results indicate that PF is a potential novel drug target for the treatment of glioblastoma by suppression of TGFß signaling pathway and inhibition of EMT.


Subject(s)
Cell Movement/drug effects , Glioblastoma/drug therapy , Glucosides/pharmacology , Monoterpenes/pharmacology , Neoplasm Invasiveness/pathology , Transforming Growth Factor beta/drug effects , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/metabolism , Humans , Mice, Nude
15.
PLoS One ; 11(12): e0168901, 2016.
Article in English | MEDLINE | ID: mdl-28030638

ABSTRACT

BACKGROUND: The Brain Trauma Foundation (BTF) guidelines published in 2007 suggest some indications for intracranial pressure (ICP) monitoring in severe traumatic brain injury (TBI). However, some studies had not shown clinical benefit in patients with severe TBI; several studies had even reported that ICP monitoring was associated with an increased mortality rate. The effect of ICP monitoring has remained controversial, regardless of the ICP monitoring guidelines. Here we performed a meta-analysis of published studies to assess the effects of ICP monitoring in patients with severe TBI. METHODS: We searched three comprehensive databases, the Cochrane Library, PUBMED, and EMBASE, for studies without limitations published up to September 2015. Mortality, ICU LOS, and hospital LOS were analyzed with Review Manager software according to data from the included studies. RESULTS: Eighteen eligible studies involving 25229 patients with severe TBI were included in our meta-analysis. The results indicated no significant reduction in the ICP monitored group in mortality (hospitalized before 2007), hospital mortality (hospitalized before 2007), mortality in randomized controlled trials. However, overall mortality, mortality (hospitalized after 2007), hospital mortality (hospitalized after 2007), mortality in observational studies (hospitalized after 2007), 2-week mortality, 6-month mortality, were reduced in ICP monitored group. Patients with an increased ICP were more likely to require ICP monitoring. CONCLUSION: Superior survival was observed in severe TBI patients with ICP monitoring since the third edition of "Guidelines for the Management of Severe Traumatic Brain Injury," which included "Indications for intracranial pressure monitoring," was published in 2007.


Subject(s)
Brain Injuries, Traumatic/mortality , Intracranial Pressure/physiology , Length of Stay/statistics & numerical data , Monitoring, Physiologic/methods , Brain Injuries, Traumatic/physiopathology , Critical Care , Hospital Mortality , Humans , Survival Rate , Treatment Outcome
16.
Drug Des Devel Ther ; 10: 767-79, 2016.
Article in English | MEDLINE | ID: mdl-26955262

ABSTRACT

In this study, we investigated the potential anticancer effects of calycosin against human glioblastoma cells, including the impacts on cell proliferation, apoptosis, and cell cycle distribution. We further studied its inhibitory activity on migration and invasion in U87 and U251 cells. Furthermore, transforming growth factor beta-mediated reductions of mesenchymal-associated genes/activators, matrix metalloproteinases-2, and -9 were detected in this process. Administration of calycosin in a glioblastoma xenograft model showed that calycosin could not only reduce tumor volume but also suppress transforming growth factor beta as well as its downstream molecules. These results revealed calycosin as a potential antitumor agent in human glioblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Glioblastoma/drug therapy , Glioblastoma/pathology , Isoflavones/pharmacology , Mesenchymal Stem Cells/drug effects , Transforming Growth Factor beta/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Glioblastoma/metabolism , Humans , Infant , Isoflavones/administration & dosage , Isoflavones/chemistry , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Structure-Activity Relationship , Tumor Cells, Cultured
17.
Drug Des Devel Ther ; 9: 5611-22, 2015.
Article in English | MEDLINE | ID: mdl-26508835

ABSTRACT

We investigated the underlying mechanism for the potent proapoptotic effect of paeoniflorin (PF) on human glioma cells in vitro, focusing on signal transducer and activator of transcription 3 (STAT3) signaling. Significant time- and dose-dependent apoptosis and inhibition of proliferation were observed in PF-treated U87 and U251 glioma cells. Expression of STAT3, its active form phosphorylated STAT3 (p-STAT3), and several downstream molecules, including HIAP, Bcl-2, cyclin D1, and Survivin, were significantly downregulated upon PF treatment. Overexpression of STAT3 induced resistance to PF, suggesting that STAT3 was a critical target of PF. Interestingly, rapid downregulation of STAT3 was consistent with its accelerated degradation, but not with its dephosphorylation or transcriptional modulation. Using specific inhibitors, we demonstrated that the prodegradation effect of PF on STAT3 was mainly through the ubiquitin-proteasome pathway rather than via lysosomal degradation. These findings indicated that PF-induced growth suppression and apoptosis in human glioma cells through the proteasome-dependent degradation of STAT3.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Glucosides/pharmacology , Monoterpenes/pharmacology , Proteasome Endopeptidase Complex/metabolism , STAT3 Transcription Factor/metabolism , Ubiquitin/metabolism , Apoptosis/drug effects , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Glioma/enzymology , Glioma/genetics , Glioma/pathology , Humans , Phosphorylation , Proteolysis , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Time Factors , Transfection , Ubiquitination
18.
PLoS One ; 10(9): e0137211, 2015.
Article in English | MEDLINE | ID: mdl-26352672

ABSTRACT

Induced neural stem cells (iNSCs) can be directly transdifferentiated from somatic cells. One potential clinical application of the iNSCs is for nerve regeneration. However, it is unknown whether iNSCs function in disease models. We produced transdifferentiated iNSCs by conditional overexpressing Oct4, Sox2, Klf4, c-Mycin mouse embryonic fibroblasts. They expanded readily in vitro and expressed NSC mRNA profile and protein markers. These iNSCs differentiated into mature astrocytes, neurons and oligodendrocytes in vitro. Importantly, they reduced lesion size, promoted the recovery of motor and sensory function as well as metabolism status in middle cerebral artery stroke rats. These iNSCs secreted nerve growth factors, which was associated with observed protection of neurons from apoptosis. Furthermore, iNSCs migrated to and passed through the lesion in the cerebral cortex, where Tuj1+ neurons were detected. These findings have revealed the function of transdifferentiated iNSCs in vivo, and thus provide experimental evidence to support the development of personalized regenerative therapy for CNS diseases by using genetically engineered autologous somatic cells.


Subject(s)
Cell Transdifferentiation , Cerebral Cortex/growth & development , Infarction, Middle Cerebral Artery/therapy , Nerve Regeneration , Neural Stem Cells/transplantation , Animals , Astrocytes/transplantation , Cell Differentiation/genetics , Cerebral Cortex/pathology , Humans , Induced Pluripotent Stem Cells/transplantation , Kruppel-Like Factor 4 , Mice , Neural Stem Cells/cytology , Neurons/transplantation , Oligodendroglia/transplantation , Rats
19.
Exp Mol Pathol ; 98(2): 192-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25617528

ABSTRACT

Glioma is an aggressive tumor with poor prognosis. Identification of precise prognostic marker and effective therapeutic target is important in the treatment of glioma. HTATIP2 is a novel tumor suppressor gene, which is frequently silenced by epigenetic mechanisms in many caners. However, the expression of HTATIP2 and how it is regulated in glioma are unknown. Hence, we assessed whether loss of HTATIP2 expression occurs in glioma, and, if so, what is the mechanism of such loss. We found that HTATIP2 expression was absent or diminished in primary gliomas compared with normal brain tissue. In vitro experiments showed that HTATIP2 expression could be restored via 5-aza-2'deoxycytidine treatment in U87 and U251 cell lines. Methyl-specific PCR indicated that the two cell lines and 60% primary gliomas carried aberrant methylated HTATIP2 alleles while normal brain tissue did not. Pyrosequencing confirmed these results and showed a higher density of methylation in the minimal promoter element, which contains four Sp1 binding sites in primary gliomas, than in normal brain tissue. Finally, we found that the overall survival was significantly higher in patients with positive HTATIP2 expression than those with loss of HTATIP2 expression. Overexpression of HTATIP2 inhibited glioma proliferation and growth in vitro. Taken together, the present study showed that loss of HTATIP2 expression was a frequent event in glioma and is associated with poor prognosis. Promoter methylation may be an underlying mechanism.


Subject(s)
Acetyltransferases/biosynthesis , Brain Neoplasms/genetics , DNA Methylation/genetics , Glioma/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/biosynthesis , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Binding Sites , Brain Neoplasms/mortality , Cell Line, Tumor , Decitabine , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Glioma/mortality , HEK293 Cells , Humans , Male , Middle Aged , Prognosis , Sp1 Transcription Factor/metabolism
20.
Stem Cells Dev ; 22(24): 3236-51, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23885779

ABSTRACT

Mesenchymal stem cells (MSCs) have been optimal targets in the development of cell based therapies, but their limited availability and high death rate after transplantation remains a concern in clinical applications. This study describes novel effects of platelet rich clot releasate (PRCR) on rat bone marrow-derived MSCs (BM-MSCs), with the former driving a gene program, which can reduce apoptosis and promote the regenerative function of the latter in hostile microenvironments through enhancement of paracrine/autocrine factors. By using reverse transcription-polymerase chain reaction, immunofluorescence and western blot analyses, we showed that PRCR preconditioning could alleviate the apoptosis of BM-MSCs under stress conditions induced by hydrogen peroxide (H2O2) and serum deprivation by enhancing expression of vascular endothelial growth factor and platelet-derived growth factor (PDGF) via stimulation of the platelet-derived growth factor receptor (PDGFR)/PI3K/AKT/NF-κB signaling pathways. Furthermore, the effects of PRCR preconditioned GFP-BM-MSCs subcutaneously transplanted into rats 6 h after wound surgery were examined by histological and other tests from days 0-22 after transplantation. Engraftment of the PRCR preconditioned BM-MSCs not only significantly attenuated apoptosis and wound size but also improved epithelization and blood vessel regeneration of skin via regulation of the wound microenvironment. Thus, preconditioning with PRCR, which reprograms BM-MSCs to tolerate hostile microenvironments and enhance regenerative function by increasing levels of paracrine factors through PDGFR-α/PI3K/AKT/NF-κB signaling pathways would be a safe method for boosting the effectiveness of transplantation therapy in the clinic.


Subject(s)
Bone Marrow Cells/cytology , Myocardial Infarction/therapy , Platelet-Rich Plasma/chemistry , Stem Cell Niche/drug effects , Animals , Apoptosis/drug effects , Culture Media, Conditioned/pharmacology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Myocardial Infarction/blood , Myocardial Infarction/genetics , NF-kappa B/metabolism , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/biosynthesis , Rats , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...