Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Discov Oncol ; 15(1): 110, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38598023

ABSTRACT

OBJECTIVE: Glioma, a malignant primary brain tumor, is notorious for its high incidence rate. However, the clinical application of temozolomide (TMZ) as a treatment option for glioma is often limited due to resistance, which has been linked to hypoxic glioma cell-released exosomes. In light of this, the present study aimed to investigate the role of exosomal pyruvate kinase M2 (PKM2) in glioma cells that exhibit resistance to TMZ. METHODS: Sensitive and TMZ-resistant glioma cells were subjected to either a normoxic or hypoxic environment, and the growth patterns and enzymatic activity of glycolysis enzymes were subsequently measured. From these cells, exosomal PKM2 was isolated and the subsequent effect on TMZ resistance was examined and characterized, with a particular focus on understanding the relevant mechanisms. Furthermore, the intercellular communication between hypoxic resistant cells and tumor-associated macrophages (TAMs) via exosomal PKM2 was also assessed. RESULTS: The adverse impact of hypoxic microenvironments on TMZ resistance in glioma cells was identified and characterized. Among the three glycolysis enzymes that were examined, PKM2 was found to be a critical mediator in hypoxia-triggered TMZ resistance. Upregulation of PKM2 was found to exacerbate the hypoxia-mediated TMZ resistance. Exosomal PKM2 were identified and isolated from hypoxic TMZ-resistant glioma cells, and were found to be responsible for transmitting TMZ resistance to sensitive glioma cells. The exosomal PKM2 also contributed towards mitigating TMZ-induced apoptosis in sensitive glioma cells, while also causing intracellular ROS accumulation. Additionally, hypoxic resistant cells also released exosomal PKM2, which facilitated TMZ resistance in tumor-associated macrophages. CONCLUSION: In the hypoxic microenvironment, glioma cells become resistant to TMZ due to the delivery of PKM2 by exosomes. Targeted modulation of exosomal PKM2 may be a promising strategy for overcoming TMZ resistance in glioma.

2.
Pak J Med Sci ; 40(3Part-II): 467-472, 2024.
Article in English | MEDLINE | ID: mdl-38356806

ABSTRACT

Objective: To explore the changes of serum-related indexes at different time points, so as to identify the critical time of converting from simple premature thelarche (PT) to idiopathic central precocious puberty (ICPP). Methods: This is a retrospective study. The subjects of the study were 50 girls with PT who were admitted to the Children's Hospital of Hebei Province from January 2019 to September 2020. The enrolled 50 children were divided into the conversion group(n=12) and the non-conversion group(n=38) according to whether PT was converted into ICPP during follow-up. Furthermore, the levels of serum-related indexes and uterine and ovarian volumes were compared after the diagnosis of PT. Results: The IGF-1 and IGFBP-3 levels of children in the conversion group began to change significantly from six months after the diagnosis, with statistically significant differences when compared with the levels of children at the initial diagnosis, three months and those of the non-conversion group at the same time points (p<0.05). The levels of vitamin-D, DHEA and leptin began to change significantly at nine months after the diagnosis (p<0.05). Besides, uterine and ovarian volumes in the conversion group began to increase significantly six months after the diagnosis, with statistically significant differences when compared with those in the non-conversion group (p<0.05). Conclusion: Findings in our study suggest that regular monitoring of vitamin-D, IGF-1, IGFBP-3, DHEA and leptin levels, and uterine and ovarian volumes can predict the conversion from PT to ICPP at an early stage.

3.
iScience ; 25(12): 105642, 2022 Dec 22.
Article in English | MEDLINE | ID: mdl-36465134

ABSTRACT

Cerebral cavernous malformation (CCM) is caused by loss-of-function mutations in CCM1, CCM2, or CCM3 genes of endothelial cells. It is characterized by pericyte deficiency. However, the role of pericytes in CCMs is not yet clarified. We found pericytes in Cdh5Cre ERT2 ;Ccm1 fl/fl (Ccm1 ECKO ) mice had a high expression of PDGFRß. The inhibition of pericyte function by CP-673451 aggravated the CCM lesion development. RNA-sequencing analysis revealed the molecular traits of pericytes, such as highly expressed ECM-related genes, especially Fn1. Furthermore, KLF4 coupled with phosphorylated SMAD3 (pSMAD3) promoted the transcription of fibronectin in the pericytes of CCM lesions. RGDS peptide, an inhibitor of fibronectin, decreased the lesion area in the cerebella and retinas of Ccm1 ECKO mice. Also, human CCM lesions had abundant fibronectin deposition, and pSMAD3- and KLF4-positive pericytes. These findings indicate that pericytes are essential for CCM lesion development, and fibronectin intervention may provide a novel target for therapeutic intervention in such patients.

4.
Cancer Sci ; 110(1): 107-117, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30387548

ABSTRACT

Gliomas are the most common central nervous system tumors. They show malignant characteristics indicating rapid proliferation and a high invasive capacity and are associated with a poor prognosis. In our previous study, p68 was overexpressed in glioma cells and correlated with both the degree of glioma differentiation and poor overall survival. Downregulating p68 significantly suppressed proliferation in glioma cells. Moreover, we found that the p68 gene promoted glioma cell growth by activating the nuclear factor-κB signaling pathway by a downstream molecular mechanism that remains incompletely understood. In this study, we found that dual specificity phosphatase 5 (DUSP5) is a downstream target of p68, using microarray analysis, and that p68 negatively regulates DUSP5. Upregulating DUSP5 in stably expressing cell lines (U87 and LN-229) suppressed proliferation, invasion, and migration in glioma cells in vitro, consistent with the downregulation of p68. Furthermore, upregulating DUSP5 inhibited ERK phosphorylation, whereas downregulating DUSP5 rescued the level of ERK phosphorylation, indicating that DUSP5 might negatively regulate ERK signaling. Additionally, we show that DUSP5 levels were lower in high-grade glioma than in low-grade glioma. These results suggest that the p68-induced negative regulation of DUSP5 promoted invasion by glioma cells and mediated the activation of the ERK signaling pathway.


Subject(s)
Brain Neoplasms/genetics , DEAD-box RNA Helicases/genetics , Dual-Specificity Phosphatases/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioma/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , DEAD-box RNA Helicases/metabolism , Dual-Specificity Phosphatases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioma/metabolism , Glioma/pathology , Humans , MAP Kinase Signaling System/genetics , Neoplasm Invasiveness , Phosphorylation , RNA Interference
5.
BMC Neurol ; 18(1): 133, 2018 Aug 31.
Article in English | MEDLINE | ID: mdl-30170559

ABSTRACT

BACKGROUND: Glioma metastasis, invasion, epithelial-mesenchymal transition (EMT) and chemoresistance indicate poor prognosis. Accumulating evidence reveals that Notch1 is an important factor in tumour progression. However, the role of Notch1 in glioma EMT and associated microRNAs (miRNAs) with the Notch pathway remain controversial. METHODS: Utilizing cBioPortal database to examine the gene signature of NOTCH1 (encoding Notch1), CDH2 (encoding N-cadherin) and SNAI1 (encoding Snail-1) in disease-free survival (DFS) and overall survival (OS). We analyzed the Notch1 expression from Oncomine. We used Western blot (WB), immunohistochemistry (IHC) and immunofluorescence to determine protein levels. Transcription was evaluated by quantitative real-time (qRT)-PCR. siRNA and lentivirus were used to knock down Notch1 and overexpress miR-139-5p, respectively. The migration and invasion of glioma cells were assessed by wound healing and transwell assays. Luciferase reporter assays were utilized to verify the relationship between Notch1 and miR-139-5p. A U87-implanted intracranial model was used to study the effect of miR-139-5p on tumour growth and Notch1 suppression efficacy or EMT reversion. RESULTS: It revealed the association of NOTCH1, CDH2, SNAI1 genomic alterations with decreases in DFS and OS. Notch1 was upregulated in classical and proneural subtypes of GBM, and associated with tumour grade. Notch1 inhibition suppressed the biological behaviours of metastasis, invasion and EMT. Notch1 was identified as a novel direct target of miR-139-5p. MiR-139-5p overexpression partially phenocopied Notch1 siRNA, whereas the forced expression of Notch1 reversed the effects of miR-139-5p on the invasion of glioma. Moreover, intracranial tumourigenicity and EMT behaviours were reduced by the introduction of miR-139-5p and partially mediated by the decreased Notch1 expression. CONCLUSIONS: miR-139-5p was identified as a tumour suppressor by negatively targeting Notch1, and this work suggests a possible molecular mechanism of the miR-139/Notch1/EMT axis for glioma treatment.


Subject(s)
Brain Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioma/pathology , MicroRNAs/genetics , Receptor, Notch1/biosynthesis , Adult , Aged , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Female , Genes, Tumor Suppressor/physiology , Glioma/genetics , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Receptor, Notch1/genetics
6.
Cancer Biol Ther ; 19(4): 260-270, 2018 04 03.
Article in English | MEDLINE | ID: mdl-27819521

ABSTRACT

Glioblastoma multiforme is the most malignant and common brain tumor in adults and is characterized by poor survival and high resistance to chemotherapy and radiotherapy. Among the new chemotherapy drugs, curcumin, a popular dietary supplement, has proven to have a potent anticancer effect on a variety of cancer cell types; however, it remains difficult to achieve a satisfactory therapeutic effect with curcumin using the traditional single-drug treatment. In this study, we found that expression of miR-326, a tumor suppressor microRNA in various tumor types, resulted in a marked increase of curcumin-induced cytotoxicity and apoptosis and a decrease of proliferation and migration in glioma cells. Moreover, we found that combination treatment of miR-326 and curcumin caused significant inhibition of the SHH/GLI1 pathway in glioma cells compared with either treatment alone, independent of p53 status. Furthermore, in vivo, the curcumin-induced increase in miR-326 expression altered the anti-glioma mechanism of this combination treatment, which further reduced tumor volume and prolonged the survival period compared to either treatment alone. Taken together, our data strongly support an important role for miR-326 in enhancing the chemosensitivity of glioma cells to curcumin.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Curcumin/pharmacology , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , MicroRNAs/metabolism , Adult , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Curcumin/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Hedgehog Proteins/metabolism , Humans , MicroRNAs/antagonists & inhibitors , Signal Transduction/genetics , Zinc Finger Protein GLI1/metabolism
7.
Tumour Biol ; 37(3): 3979-86, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26482617

ABSTRACT

Inactivation of hedgehog-interacting protein (HHIP) and overexpression of Gli1 play vital roles in the development of diverse human cancers. The aim of this study is to examine the association of HHIP and Gli1 with the clinicopathologic features and prognosis of patients with glioblastoma (GBM). The expression of HHIP and Gli1 in 103 patients with GBM and 32 control patients was investigated by immunohistochemistry. Statistical analysis was utilized to evaluate the association of HHIP as well as Gli1 with clinicopathological characteristics and prognosis of patients. HHIP and Gli1 were dysregulated in GBM. Spearman's rank analysis showed that HHIP and Gli1 had an inverse correlation (r = -0.386, P = 0.000). Expression of HHIP was significantly correlated with age (P = 0.000), gender (P = 0.003), seizure (P = 0.013), resection degree (P = 0.033), adjuvant treatment (P = 0.030), and O(6)-methylguanine-DNA methyltransferase (MGMT) methylation (P = 0.021), while Gli1 expression was significantly correlated with age (P = 0.002), gender (P = 0.033), Karnofsky performance status (KPS) score (P = 0.028), resection degree (P = 0.000), adjuvant treatment (P = 0.014), and MGMT methylation (P = 0.030). Kaplan-Meier method showed that patients with low Gli1 expression had longer overall survival (OS) than those with high Gli1 expression (P = 0.000) and the OS of the patients with HHIP-positive GBM was significantly longer than that of the patients with HHIP-negative GBM (P = 0.000). Univariate and multivariate analyses confirmed that HHIP expression and Gli1 expression were independent prognostic factors. Our data suggested that expression of HHIP could be considered as significant prognostic marker for patients with GBM.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Carrier Proteins/metabolism , Glioblastoma/metabolism , Membrane Glycoproteins/metabolism , Adult , Aged , Brain Neoplasms/diagnosis , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Glioblastoma/diagnosis , Glioblastoma/mortality , Glioblastoma/therapy , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Prognosis , Proportional Hazards Models , Young Adult , Zinc Finger Protein GLI1/metabolism
8.
Mol Med Rep ; 12(5): 7815, 2015 11.
Article in English | MEDLINE | ID: mdl-26460078

ABSTRACT

Mol Med Rep 12: [Related article:] 6702­6710, 2015; DOI: 10.3892/mmr.2015.4229 After the publication of the article, it has been brought to the authors' attention by an interested reader that we had made an error regarding the presentation of certain data in the manuscript. The error relates to the presentation of Figs. 1 and 2 in the paper: The control panels for Fig. 1C [labelled 'cyclopamine (µM)'] and Figs. 2B and C [labelled 'rhSSH (µg/ml)'] were derived from the same image. The control U251 cells, featured in Fig. 1 and Figs. 2B and C, were treated without cyclopamine and rhSHH. Therefore, the U251 cells treated without cyclopamine and rhSHH were considered as a control group compared with U251 cells that were separately treated with cyclopamine or rhSHH, and these were photographed randomly. A new Fig. 2 is provided, which contains the correct data for the control panels for Figs. 2B and C.

9.
Mol Med Rep ; 12(5): 6702-10, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26299938

ABSTRACT

Aberrant hedgehog signaling contributes to the development of various malignancies, including glioblastoma (GBM). However, the potential mechanism of hedgehog signaling in GBM migration and invasion has remained to be elucidated. The present study showed that enhanced hedgehog signaling by recombinant human sonic hedgehog N­terminal peptide (rhSHH) promoted the adhesion, invasion and migration of GBM cells, accompanied by increases in mRNA and protein levels of matrix metalloproteinase­2 (MMP­2) and MMP­9. However, inhibition of hedgehog signaling with cyclopamine suppressed the adhesion, invasion and migration of GBM cells, accompanied by decreases in mRNA and protein levels of MMP­2 and ­9. Furthermore, it was found that MMP­2- and MMP­9-neutralizing antibodies or GAM6001 reversed the inductive effects of rhSHH on cell migration and invasion. In addition, enhanced hedgehog signaling by rhSHH increased AKT phosphorylation, whereas blockade of hedgehog signaling decreased AKT phosphorylations. Further experiments showed that LY294002, an inhibitor of phosphoinositide-3 kinase (PI3K), decreased rhSHH­induced upregulation of MMP­2 and ­9. Finally, the protein expression of glioblastoma-associated oncogene 1 was positively correlated with levels of phosphorylated AKT as well as protein expressions of MMP­2 and ­9 in GBM tissue samples. In conclusion, the present study indicated that the hedgehog pathway regulates GBM-cell migration and invasion by increasing MMP-2 and MMP-9 production via the PI3K/AKT pathway.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Glioblastoma/pathology , Hedgehog Proteins/metabolism , Signal Transduction , Brain/metabolism , Brain Neoplasms/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Glioblastoma/metabolism , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
10.
Neuro Oncol ; 17(2): 243-53, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25173582

ABSTRACT

BACKGROUND: Few studies have associated microRNAs (miRNAs) with the hedgehog (Hh) pathway. Here, we investigated whether targeting smoothened (SMO) with miR-326 would affect glioma biological behavior and stemness. METHODS: To investigate the expression of SMO and miR-326 in glioma specimens and cell lines, we utilized quantitative real-time (qRT)-PCR, Western blot, immunohistochemistry, and fluorescence in situ hybridization. The luciferase reporter assay was used to verify the relationship between SMO and miR-326. We performed cell counting kit-8, transwell, and flow cytometric assays using annexin-V labeling to detect changes after transfection with siRNA against SMO or miR-326. qRT-PCR assays, neurosphere formation, and immunofluorescence were utilized to detect the modification of self-renewal and stemness in U251 tumor stem cells. A U251-implanted intracranial model was used to study the effect of miR-326 on tumor volume and SMO suppression efficacy. RESULTS: SMO was upregulated in gliomas and was associated with tumor grade and survival period. SMO inhibition suppressed the biological behaviors of glioma cells. SMO expression was inversely correlated with miR-326 and was identified as a novel direct target of miR-326. miR-326 overexpression not only repressed SMO and downstream genes but also decreased the activity of the Hh pathway. Moreover, miR-326 overexpression decreased self-renewal and stemness and partially prompted differentiation in U251 tumor stem cells. In turn, the inhibition of Hh partially elevated miR-326 expression. Intracranial tumorigenicity induced by the transfection of miR-326 was reduced and was partially mediated by the decreased SMO expression. CONCLUSIONS: This work suggests a possible molecular mechanism of the miR- 326/SMO axis, which can be a potential alternative therapeutic pathway for gliomas.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line, Tumor , Hedgehog Proteins/metabolism , Humans , Kaplan-Meier Estimate , Mice, Inbred BALB C , Mice, Nude , Signal Transduction , Smoothened Receptor , Survival Rate , Up-Regulation
11.
Oncotarget ; 5(22): 11681-94, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25373737

ABSTRACT

Glioblastoma are highly aggressive brain tumors with poor prognosis. While various dysregulation of signaling pathways in gliomas have been described, the identification of biomarkers and therapy targets remains an important task for novel diagnostic and therapeutic approaches. Here we described that the Suppressor of fused (also known as Sufu) is significantly down-regulated in high-grade gliomas, correlating with a poor prognosis. We demonstrated that ectopic expression of Sufu inhibited cell proliferation, invasion and vasculogenic mimicry. In addition, overexpression of Sufu reduced Gli reporter gene transcription activity and prevented Gli1 nuclear accumulation, whereas knockdown of Sufu reversed these effects. Furthermore, overexpressed Sufu sensitized glioblastoma to Temozolomide and Cyclopamine. Thus, Sufu is potential tumor suppressor and therapeutic target in glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , Cell Nucleus/metabolism , Glioma/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Adult , Animals , Cell Line, Tumor , Cell Proliferation , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Disease Progression , Down-Regulation , Female , Glioma/drug therapy , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Transplantation , Prognosis , RNA, Small Interfering/metabolism , Temozolomide , Treatment Outcome , Veratrum Alkaloids/administration & dosage , Zinc Finger Protein GLI1
12.
Oncotarget ; 5(9): 2551-61, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24810474

ABSTRACT

Astrocytic tumors are the most common primary brain tumors in adults. ATRX mutations have been identified in gliomas and are correlated with its loss of expression, which causes alternative lengthening of telomeres (ALT) leading to genomic instability. In this study, we aimed to explore the role of ATRX mRNA expression alteration in the progression and subclassification of astrocytic tumors and examine its impact on clinical outcome. We investigated ATRX mRNA expression and its association with IDH1 and IDH2 mutations in 169 adult astrocytic tumors using whole transcriptome sequencing. In our cohort, low ATRX mRNA expression was detected in 68% of astrocytomas, 50% of anaplastic astrocytomas and 41.6% of glioblastomas. Low ATRX expression closely overlapped with mutations in IDH1/2 (P<0.0001) in astrocytic tumors across WHO grades II-IV. Significant association between low ATRX expression and longer overall survival was identified in our cohort (P<0.01). ATRX combined with IDH1/2 and Ki-67 was used to re-classify patients with astrocytic tumors: group A1 containing IDH1/2 mutations and low ATRX expression predicted a better prognostic outcome, whereas group A3 carrying wild-type IDH1/2 and high Ki-67 expression had the shortest overall survival; IDH-mutant tumors with low ATRX expression and IDH-wild-type tumors with high Ki-67 expression were grouped into group A2. In summary, our results showed that ATRX in cooperation with IDH1/2 and Ki-67 defines three subgroups of astrocytic tumors regardless of the conventional WHO grades consensus. The molecular stratification in astrocytic tumors may aid in treatment strategy selection, therapeutic trial design, and clinical prognosis evaluation.


Subject(s)
Astrocytoma/classification , DNA Helicases/genetics , Gene Expression Profiling , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Nuclear Proteins/genetics , RNA, Messenger/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Astrocytoma/genetics , Astrocytoma/mortality , Biomarkers, Tumor/genetics , Cohort Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Grading , Oligonucleotide Array Sequence Analysis , Prognosis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , X-linked Nuclear Protein , Young Adult
13.
PLoS One ; 9(2): e88096, 2014.
Article in English | MEDLINE | ID: mdl-24516588

ABSTRACT

BACKGROUND: ABO blood groups have been suggested to contribute to the development of certain tumors; however, the associations between ABO blood groups and the incidence and metastases of cutaneous malignant melanomas have not been fully evaluated in Chinese populations. Thus, we investigated these associations with a case-controlled study in northeast of China. METHODS: A total of 482 patients with cutaneous malignant melanoma and 3,068 healthy- controls were enrolled for the study between 2001 and 2012 at The Tumor Hospital of Harbin Medical University. A multivariate logistic model was used to evaluate adjusted odds ratios (ORs) and 95% confidence intervals (CI) for the incidence and metastases of cutaneous malignant melanoma. RESULTS: Blood type A individuals had higher tumor incidence and metastasis compared to those with blood type O (OR = 1.575; 95% CI = 1.208-2.053, p = 0.001; OR = 2.004; 95% CI = 1.032-3.889, p = 0.040), after adjusting for age, gender, smoking status and alcohol consumption. CONCLUSIONS: Blood type A was associated with higher incidence and metastasis of cutaneous malignant melanoma but future studies are needed to examine the mechanisms linking cutaneous malignant melanoma to ABO blood types.


Subject(s)
ABO Blood-Group System , Melanoma/epidemiology , Skin Neoplasms/epidemiology , Aged , Case-Control Studies , China/epidemiology , Female , Humans , Incidence , Male , Melanoma/blood , Melanoma/secondary , Middle Aged , Retrospective Studies , Skin Neoplasms/blood , Skin Neoplasms/pathology
14.
Carcinogenesis ; 35(8): 1698-706, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24480809

ABSTRACT

Bone morphogenetic protein-2 (BMP-2), a member of the transforming growth factor-ß family, plays critical roles in cell differentiation, modeling and regeneration processes in several tissues. BMP-2 is also closely associated with various malignant tumors. microRNAs negatively and posttranscriptionally regulate gene expression and function as oncogenes or tumor suppressors. Herein, we report that miR-656 expression was significantly downregulated in glioma cell lines and tissues. We identified and confirmed that BMP receptor, type 1A (BMPR1A) is a direct target of miR-656. The expression of BMPR1A was negatively correlated with that of miR-656 in human glioma tissues. We further demonstrated that miR-656 suppressed glioma cell proliferation, neurosphere formation, migration and invasion with or without exogenous BMP-2. Engineered knockdown of BMPR1A diminished the antiproliferation effect of miR-656 in vitro. Moreover, the canonical BMP/Smad and non-canonical BMP/mitogen-activated protein kinase (MAPK) pathways were inhibited by miR-656 overexpression. Several cancer-related signaling molecules, including cyclin B, cyclin D1, matrix metalloproteinase-9, p21 and p27, were also involved in miR-656 function in glioma cells. The tumor-suppressing function of miR-656 was validated using an in vivo intracranial xenograft mouse model. Notably, ectopic expression of miR-656 markedly reduced tumor size and prolonged the survival of mice treated with or without BMP-2. These results elucidate the function of miR-656 in glioma progression and suggest a promising application for glioma treatment.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/metabolism , Brain Neoplasms/genetics , Brain/metabolism , Cell Transformation, Neoplastic/genetics , Glioma/genetics , MicroRNAs/genetics , Animals , Blotting, Western , Bone Morphogenetic Protein Receptors, Type I/antagonists & inhibitors , Bone Morphogenetic Protein Receptors, Type I/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Case-Control Studies , Cell Cycle , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Glioma/metabolism , Glioma/pathology , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Grading , Neoplasm Invasiveness , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Wound Healing , Xenograft Model Antitumor Assays
15.
Asian Pac J Cancer Prev ; 15(23): 10407-12, 2014.
Article in English | MEDLINE | ID: mdl-25556484

ABSTRACT

BACKGROUND: ß-elemene, extracted from herb medicine Curcuma wenyujin has potent anti-tumor effects in various cancer cell lines. However, the activity of ß-elemene against glioma cells remains unclear. In the present study, we assessed effects of ß-elemene on human glioma cells and explored the underlying mechanism. MATERIALS AND METHODS: Human glioma U87 cells were used. Cell proliferation was determined with MTT assay and colony formation assay to detect the effect of ß-elemene at different doses and times. Fluorescence microscopy was used to observe cell apoptosis with Hoechst 33258 staining and change of glioma apoptosis and cell cycling were analyzed by flow cytometry. Real-time quantitative PCR and Western-blotting assay were performed to investigated the influence of ß-elemene on expression levels of Fas/FasL, caspase-3, Bcl-2 and Bax. The experiment was divided into two groups: the blank control group and ß-elemne treatment group. RESULTS: With increase in the concentration of ß-elemene, cytotoxic effects were enhanced in the glioma cell line and the concentration of inhibited cell viability (IC50) was 48.5 µg/mL for 24h. ß-elemene could induce cell cycle arrest in the G0/G1 phase. With Hoechst 33258 staining, apoptotic nuclear morphological changes were observed. Activation of caspase-3,-8 and -9 was increased and the pro-apoptotic factors Fas/FasL and Bax were upregulated, while the anti-apoptotic Bcl-2 was downregulated after treatment with ß-elemene at both mRNA and protein levels. Furthermore, proliferation and colony formation by U87 cells were inhibited by ß-elemene in a time and does- dependent manner. CONCLUSIONS: Our results indicate that ß-elemene inhibits growth and induces apoptosis of human glioma cells in vitro. The induction of apoptosis appears to be related with the upregulation of Fas/FasL and Bax, activation of caspase-3,-8 and -9 and downregulation of Bcl-2, which then trigger major apoptotic cascades.


Subject(s)
Apoptosis/drug effects , Fas Ligand Protein/drug effects , Glioma/genetics , Proto-Oncogene Proteins c-bcl-2/drug effects , RNA, Messenger/drug effects , Sesquiterpenes/pharmacology , bcl-2-Associated X Protein/drug effects , fas Receptor/drug effects , Apoptosis/genetics , Blotting, Western , Caspases/drug effects , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Glioma/metabolism , Humans , In Vitro Techniques , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , fas Receptor/genetics , fas Receptor/metabolism
16.
CNS Neurosci Ther ; 19(12): 926-36, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24165291

ABSTRACT

AIMS: To study the role of curcumin on glioma cells via the SHH/GLI1 pathway in vitro and vivo. METHODS: The effects of curcumin on proliferation, migration, apoptosis, SHH/GLI1 signaling, and GLI1 target genes expression were evaluated in multiple glioma cell lines in vitro. A U87-implanted nude mice model was used to study the role of curcumin on tumor volume and the suppression efficacy of GLI1. RESULTS: Curcumin showed cytotoxic effects on glioma cell lines in vitro. Both mRNA and protein levels of SHH/GLI1 signaling (Shh, Smo, GLI1) were downregulated in a dose- and time-dependent manner. Several GLI1-dependent target genes (CyclinD1, Bcl-2, Foxm1) were also downregulated. Curcumin treatment prevented GLI1 translocating into the cell nucleus and reduced the concentration of its reporter. Curcumin suppressed cell proliferation, colony formation, migration, and induced apoptosis which was mediated partly through the mitochondrial pathway after an increase in the ratio of Bax to Bcl2. Intraperitoneal injection of curcumin in vivo reduced tumor volume, GLI1 expression, the number of positively stained cells, and prolonged the survival period compared with the control group. CONCLUSION: This study shows that curcumin holds a great promise for SHH/GLI1 targeted therapy against gliomas.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Curcumin/therapeutic use , Glioma/drug therapy , Signal Transduction/drug effects , Animals , Brain Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colony-Forming Units Assay , Disease Models, Animal , Glioma/metabolism , Hedgehog Proteins/metabolism , Humans , Kaplan-Meier Estimate , Mice , Transcription Factors/metabolism , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1
17.
CNS Neurosci Ther ; 19(7): 477-83, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23551751

ABSTRACT

AIMS: Mcl-1, an antiapoptotic member of the Bcl-2 family, is overexpressed in human glioblastoma, conferring a survival advantage to tumor cells. The mechanisms underlying its dysregulation have not been clarified. In this study, we explored the involvement of micro-RNAs that acted as endogenous sequence-specific suppressors of gene expression. METHODS AND RESULTS: Using computational and TCGA analysis, we identified miR-139 as being downregulated in glioblastoma in comparison with human brain tissue, as well as possessing a putative target site in Mcl-1 mRNA. Overexpression of miR-139 led to a clear decrease in Mcl-1 expression in gliomas. Reporter assays revealed direct post-transcriptional regulation involving miR-139 and the 3'-untranslated region of Mcl-1. Human glioma tissues with low expression of miR-139 displayed higher expression of Mcl-1 protein than those with high expression, suggesting that low miR-139 contributes to Mcl-1 overexpression. In addition, upregulation of miR-139 suppressed the proliferation and enhanced temozolomide (TMZ)-induced apoptosis. Finally, we observed that Mcl-1 knockdown resulted in similar effects compared with miR-139 transfection. CONCLUSION: Our results suggested that miR-139 negatively regulated Mcl-1 and induced apoptosis in cooperation with an anticancer drug TMZ in glioma.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Brain Neoplasms/pathology , Dacarbazine/analogs & derivatives , Glioma/pathology , MicroRNAs/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , 3' Untranslated Regions , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Humans , Immunohistochemistry , Luciferases/genetics , Mice , Mice, Nude , MicroRNAs/metabolism , Neoplasm Transplantation , Oligonucleotides/genetics , Paraffin Embedding , Plasmids/genetics , Real-Time Polymerase Chain Reaction , Temozolomide
18.
Cancer Lett ; 329(2): 174-80, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23142218

ABSTRACT

MicroRNAs are strongly implicated as affecting glioma, but their specific roles and functions have yet to be fully elucidated. In this study, we defined the expression and function of miR-24, which we found to be upregulated in glioma samples and glioma cells by qRT-PCR. Downregulation of miR-24 in glioma cell lines inhibited proliferation and invasion and induced apoptosis. Using computational and expression analysis, ST7L was identified as a candidate target of miR-24. A reporter assay with the 3'UTR of ST7L cloned downstream of a luciferase gene showed increased luciferase activity in the absence of miR-24, providing strong evidence that miR-24 is a direct regulator of ST7L. Furthermore, we observed that restoration of ST7L activity resulted in effects that were similar to those from transfecting a miR-24 inhibitor into glioma cells. Mechanistic investigation revealed that the deletion of miR-24 suppressed ß-catenin/Tcf-4 transcription activity by targeting ST7L. In conclusion, our study demonstrates that miR-24 upregulation is common in glioma and that suppression of miR-24 expression inhibits cell proliferation and invasion, suggesting that miR-24 may act as an oncogene in glioma.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Brain Neoplasms/metabolism , Cell Proliferation , Glioblastoma/metabolism , MicroRNAs/genetics , RNA-Binding Proteins/genetics , Transcription Factors/metabolism , beta Catenin/metabolism , 3' Untranslated Regions , Animals , Apoptosis , Base Sequence , Binding Sites , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice , Mice, Nude , MicroRNAs/metabolism , MicroRNAs/physiology , Neoplasm Invasiveness , Neoplasm Transplantation , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Transcription Factor 4 , Tumor Suppressor Proteins
19.
CNS Neurosci Ther ; 19(2): 109-16, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23253187

ABSTRACT

AIMS: To examine a novel strategy to enhance the survival of grafted neural stem cells (NSCs) in stroke model. METHODS: Using a cell counting kit-8 (CCK-8) and TUNEL assay to test the protective effects of p53 inhibitor, pifithrin-α (PFT-α), on oxygen glucose deprivation (OGD) in NSCs. We compared the effects of vehicle + NSCs and FFT-α + NSCs on the efficacy of transplantation in stroke rat model using behavioral analysis, immunohistochemistry, etc. RESULTS: Pifithrin-α increased viability and decreased apoptosis in NSCs after OGD in vitro. By in vivo studies, we showed that the best recovery of neurological function in the stroke rats and the maximum survival of grafted NSCs were found in the PFT-α + NSCs group. Twelve hours after cell transplantation, p53 was localized to the nuclei of grafted NSCs in the vehicle + NSCs group but was primarily localized to the cytoplasm in the PFT-α + NSCs group. The p53-upregulated modulator of apoptosis (PUMA) was highly expressed among the grafted cells in the vehicle + NSCs group compared with that in the PFT-α + NSCs group. CONCLUSION: Our results indicate that PFT-α enhances the survival of grafted NSCs through the inhibition of p53 translocation into the nucleus.


Subject(s)
Benzothiazoles/administration & dosage , Neural Stem Cells/transplantation , Stem Cell Transplantation/methods , Stroke/therapy , Toluene/analogs & derivatives , Tumor Suppressor Protein p53/antagonists & inhibitors , Up-Regulation/physiology , Animals , Cell Nucleus/drug effects , Cell Nucleus/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Combined Modality Therapy/methods , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Recovery of Function/physiology , Stroke/genetics , Stroke/metabolism , Toluene/administration & dosage , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/physiology
20.
Int J Biochem Cell Biol ; 44(11): 1711-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22750473

ABSTRACT

MET, the receptor for hepatocyte growth factor receptor (HGF), has been reported to trigger multiple and sometimes opposing cellular responses in various types of tumor cells. It has been implicated in the regulation of tumor-cell survival, proliferation, angiogenesis, invasion and metastasis. However, the MET regulatory mechanism in glioma is not well known. MicroRNAs are a class of small noncoding RNAs that play important roles in a variety of biological processes including human cancers. In this study, we used computational and expressional analysis to identify that the 'seed sequence' of miR-410 matched the 3' UTR of the MET mRNA. Besides, the expression of miR-410 was inversely associated with MET in human glioma tissues. Using luciferase and western blot assay, we certified that miR-410 directly targeted MET in glioma cells. While restoring expression of miR-410 led to proliferation inhibition and reduced invasive capability in glioma cells. Furthermore, we showed that miR-410 played an important role in regulating MET-induced AKT signal transduction. While downregulation of MET by RNAi, we observed that MET knockdown resulted in effects similar to that with miR-410 transfection in glioma cells. Our findings suggest that miR-410, a direct regulator of MET, may function as a tumor suppressor in human gliomas.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioma/genetics , Glioma/pathology , MicroRNAs/metabolism , Proto-Oncogene Proteins c-met/genetics , 3' Untranslated Regions/genetics , Animals , Antineoplastic Agents , Base Sequence , Binding Sites/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Mice , MicroRNAs/genetics , Molecular Sequence Data , Neoplasm Invasiveness , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...