Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Med Rep ; 27(4)2023 Apr.
Article in English | MEDLINE | ID: mdl-36866735

ABSTRACT

Following the publication of this paper, it was drawn to the Editors' attention by a concerned reader that certain of the scratch-wound data shown in Fig. 3A were strikingly similar to data appearing in different form in another article by different authors. Owing to the fact that the contentious data in the above article had already been published elsewhere prior to its submission to Molecular Medicine Reports, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience caused. [Molecular Medicine Reports 13: 1558­1662, 2016; DOI: 10.3892/mmr.2015.4721].

2.
World Neurosurg ; 117: 109-114, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29890279

ABSTRACT

BACKGROUND: Giant serpentine aneurysms (GSAs) are a subgroup of giant intracranial aneurysms, distinct from saccular and fusiform varieties, that are defined as partially thrombosed giant aneurysms with tortuous internal vascular channel. Clinicopathologic characteristics of middle cerebral artery GSAs have been rarely reported in the literature, with discussion of radiologic characteristics only. We clarify patient clinical and neuroradiologic features and discuss the mechanism of formation and progression. CASE DESCRIPTION: A 43-year-old woman presented with a GSA arising from the middle cerebral artery. There was a separate inflow and outflow channel of the aneurysm, with the outflow channel feeding the distal branches of the parent artery and supplying normal brain parenchyma. The GSA was treated successfully by aneurysmectomy and superficial temporal artery-middle cerebral artery bypass followed by proximal occlusion and vascular reconstruction. An aneurysm specimen was examined to correlate pathologic findings and morphologic characteristics. RESULT: Pathologic results showed that thickness of the aneurysmal wall was typically increased and varied, and no internal elastic lamina or endothelial lining could be identified. The sac contained thrombi of various ages with recanalizing vessel formation and chronic inflammation infiltration. Intimal hyperplasia and neoangiogenesis in the wall and hyaline degeneration of the media were observed. Vessels coursing in their adventitia showed mucoid changes, which are responsible for the contrast enhancement of the aneurysmal rim on computed tomography scan. CONCLUSIONS: GSAs are a specific pathologic entity with unique morphologic and pathologic characteristics that can affect intracranial blood vessels. The pathogenic mechanisms are unclear; this report suggests that GSAs may be associated with degeneration of the vascular wall.


Subject(s)
Intracranial Aneurysm/diagnosis , Intracranial Aneurysm/pathology , Middle Cerebral Artery/diagnostic imaging , Middle Cerebral Artery/pathology , Adult , Diagnosis, Differential , Female , Humans , Intracranial Aneurysm/physiopathology , Intracranial Aneurysm/surgery , Middle Cerebral Artery/surgery
3.
Mol Med Rep ; 13(2): 1558-62, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26707955

ABSTRACT

The poor prognosis of gliomas is to a large extent attributed to the markedly proliferative and invasive nature of the disease. Endocannabinoids have emerged as novel potential anti-tumor agents. The present study aimed to investigate the anti-carcinogenic activity of anandamide (AEA), an endocannabinoid, on glioma cells. To assess the functional role of AEA in glioma, the effects of AEA on cell proliferation, migration, invasion, apoptosis and the cell cycle in vitro, and tumor growth in vivo, were investigated. AEA markedly inhibited the proliferation of U251 cells in a dose- and time-dependent manner. Flow cytometric assays revealed that the apoptosis rate of U251 cells upon treatment with AEA was increased. AEA also suppressed the adhesion, migration and invasion capabilities of the U251 cells. Furthermore, AEA inhibited tumor growth in vivo. These results highlighted the potential role of AEA in the tumorigenesis and progression of glioma, and suggested that AEA exhibits therapeutic potential in the management of human glioma.


Subject(s)
Arachidonic Acids/administration & dosage , Carcinogenesis/drug effects , Cell Proliferation/drug effects , Endocannabinoids/administration & dosage , Glioma/drug therapy , Polyunsaturated Alkamides/administration & dosage , Animals , Apoptosis/drug effects , Cell Cycle , Cell Line, Tumor , Cell Movement/drug effects , Glioma/pathology , Humans , Mice , Neoplasm Invasiveness/pathology , Xenograft Model Antitumor Assays
4.
Oncol Lett ; 8(3): 1195-1201, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25120686

ABSTRACT

The B7 family consists of activating and inhibitory molecules that regulate immune responses. Recent research demonstrated the roles of soluble B7-H3 (sB7-H3) and soluble B7-H1 (sB7-H1) in the blood serum of various tumors; however, none of these studies investigated the expression of these proteins in the cerebral spinal fluid (CSF) and blood serum of patients with glioma. The aim of the present study was to investigate the expression of B7-H3 and B7-H1 in the CSF, blood serum and tissues of patients with glioma and their correlation with clinicopathological data. Between January 2012 and November 2012, samples were obtained from 78 patients with glioma, four CSF samples were obtained from patients with a moderate traumatic brain injury, four brain tissue samples were obtained from patients with a traumatic brain injury and 40 blood serum samples were obtained from healthy individuals. The expression of B7-H3 and B7-H1 in the CSF, blood serum and tumor samples of the patients with high-grade glioma was found to be higher than that in the patients with low-grade glioma. However, no significant differences in sB7-H3 and sB7-H1 expression were observed in the blood serum of the patients with glioma compared with the healthy control subjects. In addition, the expression of sB7-H3 and sB7-H1 in the CSF of the patients with glioma was higher than that in the CSF of the patients with a moderate traumatic brain injury. Furthermore, in the patients with glioma, B7-H3 and B7-H1 expression in the CSF and tumor tissue, although not in the blood serum, correlated with the glioma grade.

5.
Exp Ther Med ; 6(1): 140-146, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23935735

ABSTRACT

Spinal cord injury (SCI) is a severe neurological disease. An effective strategy for the treatment of SCI is urgently required. Stem cell transplantation has emerged as a viable therapeutic option with great potential for restoring neurological function lost following SCI. From 2009 to 2010, a total of 20 SCI patients were enrolled in a clinical trial by Wuhan Hongqiao Brain Hospital; all patients completed and signed informed consent prior to autologous bone marrow-derived mesenchymal stem cell transplantation. Analysis of subsequent treatment results indicated significant improvements in sensory, motor and autonomic nerve function as assessed by the American Spinal Injury Association's impairment scale. Thirty days after transplantation, a total of 15 patients (75%) demonstrated improvement, including four of the eight patients (50%) with grade A SCI, three of the four patients (75%) with grade B injury and all eight patients (100%) with grade C injury. The most common adverse events, fever and headache, disappeared within 24-48 h without treatment.

6.
J Transl Med ; 11: 69, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23514245

ABSTRACT

BACKGROUND: Our previous study showed that SLC22A18 downregulation and promoter methylation were associated with the development and progression of glioma and the elevated expression of SLC22A18 was found to increase the sensitivity of glioma U251 cells to the anticancer drug 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU). In this study, we investigated the predictive value of SLC22A18 promoter methylation and protein expression in glioblastoma multiforme (GBM) patients receiving temozolomide (TMZ) therapy. PATIENTS AND METHODS: SLC22A18 promoter methylation and protein expression were examined by methylation-specific polymerase chain reaction (MSP) and Western blotting respectively, then we compared SLC22A18 promoter methylation and protein expression in tumor cell explants in regard to prediction of TMZ response and survival time of 86 GBM patients. RESULTS: SLC22A18 promoter methylation was detected in 61 of 86 (71%) samples, whereas 36 of 86 (42%) cases were scored positive for SLC22A18 protein expression. Overall SLC22A18 promoter methylation was significantly related to SLC22A18 protein expression, but a subgroup of cases did not follow this association. Multivariate Cox regression analysis indicated that SLC22A18 protein expression, but not promoter methylation, was significantly correlated with TMZ therapy. SLC22A18 protein expression predicted a significantly shorter overall survival in 51 patients receiving TMZ therapy, whereas no differences in overall survival were observed in 35 patients without TMZ therapy. CONCLUSIONS: These results show that lack of SLC22A18 protein expression is superior to promoter methylation as a predictive tumor biomarker in GBM patients receiving temozolomide therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/metabolism , Dacarbazine/analogs & derivatives , Glioblastoma/metabolism , Organic Cation Transport Proteins/metabolism , Blotting, Western , Brain Neoplasms/drug therapy , DNA Methylation , Dacarbazine/therapeutic use , Female , Glioblastoma/drug therapy , Humans , Male , Organic Cation Transport Proteins/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic , Temozolomide
7.
J Clin Neurosci ; 20(4): 543-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23317753

ABSTRACT

Special AT-rich-sequence-binding protein 1 (SATB1), a new type of gene regulator, has been reported to be expressed in various human cancers and may be associated with malignancy. The aim of this study was to investigate the expression of SATB1 in astrocytoma and to determine its prognostic value for the overall survival of patients with astrocytoma. The expression of SATB1 protein and messenger RNA (mRNA) in human astrocytoma specimens was examined using immunohistochemistry and semi-quantitative reverse transcription polymerase chain reaction (RT-PCR). The relationship between SATB1 expression and O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status was also investigated. Spearman's correlation coefficient was used to describe the association between SATB1 expression and the clinical parameters of astrocytoma patients. SATB1 protein and mRNA were expressed at significant levels in astrocytoma specimens. SATB1 expression was positively correlated with astrocytoma pathological grade but negatively correlated with the life span of astrocytoma patients. SATB1 expression was also significantly lower in astrocytoma specimens with MGMT promoter methylation than in those without MGMT promoter methylation. Our findings suggest that SATB1 may have an important role as a positive regulator of astrocytoma development and progression and that SATB1 might be a useful molecular marker for predicting the prognosis of patients with astrocytoma and could be a novel target for treating astrocytoma.


Subject(s)
Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Matrix Attachment Region Binding Proteins/genetics , Adult , Aged , Astrocytoma/surgery , Biomarkers, Tumor/genetics , Brain Neoplasms/surgery , Carcinogens , DNA Methylation , Female , Humans , Immunohistochemistry , Male , Middle Aged , Multivariate Analysis , Neurosurgical Procedures , O(6)-Methylguanine-DNA Methyltransferase/genetics , Prognosis , RNA/biosynthesis , RNA/isolation & purification , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Real-Time Polymerase Chain Reaction , Regression Analysis , Survival , Survival Analysis
8.
Mol Med Rep ; 7(1): 139-43, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23076250

ABSTRACT

Special AT-rich sequence-binding protein-1 (SATB1) has been reported to be overexpressed in numerous human tumors. The aim of the present study was to determine the correlation and clinical significance between the expression of SATB1 and B-cell lymphoma 2 (Bcl-2) in human glioblastoma multiforme (GBM). Samples from 70 patients with GBMs were analyzed and 10 normal brain tissues were used as the control group. In situ hybridization was used to detect SATB1 mRNA expression and immunohistochemistry was used to detect Bcl-2 and proliferating cell nuclear antigen (PCNA) protein expression. Apoptosis was detected with flow cytometry. The SATB1 mRNA and Bcl-2 protein levels were found to be significantly higher in GBM tissues than in normal brain and their levels were associated with patient survival, but not associated with patient gender, age and tumor size and site. A positive correlation was observed between SATB1 mRNA and Bcl-2 protein and between SATB1 mRNA and PCNA. A negative correlation was observed between SATB1 mRNA and apoptosis and between Bcl-2 and apoptosis. A positive correlation existed between Bcl-2 and PCNA. Patients with GBM identified as SATB1 mRNA (+) and Bcl-2 (+) were associated with a poor prognosis. Therefore, assessment of SATB1 and Bcl-2 co-expression may provide important information for the diagnosis, therapy and prognosis of GBM.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Matrix Attachment Region Binding Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Adult , Aged , Apoptosis/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Female , Flow Cytometry , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Matrix Attachment Region Binding Proteins/metabolism , Middle Aged , Prognosis , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
9.
J Transl Med ; 10: 149, 2012 Jul 28.
Article in English | MEDLINE | ID: mdl-22839214

ABSTRACT

BACKGROUND: Special AT-rich sequence-binding protein-1 (SATB1) has been reported to be expressed in several human cancers and may have malignant potential. This study was aimed at investigating the expression and potential role of SATB1 in human glioma. METHOD: The relationship between SATB1 expression, clinicopathological parameters, Ki67 expression and MGMT promoter methylation status was evaluated, and the prognostic value of SATB1 expression in patients with gliomas was analyzed. SATB1-specific shRNA sequences were synthesized, and U251 cells were transfected with SATB1 RNAi plasmids. Expression of SATB1 mRNA and protein was investigated by RT-PCR and immunofluoresence staining and western blotting. The expression of c-Met, SLC22A18, caspase-3 and bcl-2 protein was determined by western blotting. U251 cell growth and adherence was detected by methyl thiazole tetrazolium assay. The apoptosis of U251 cells was examined with a flow cytometer. The adherence, invasion, and in vitro angiogenesis assays of U251 cells were done. The growth and angiogenesis of SATB1 low expressing U251 cells was measured in an in vivo xenograft model. RESULTS: Of 70 tumors, 44 (62.9%) were positive for SATB1 expression. SATB1 expression was significantly associated with a high histological grade and with poor survival in univariate and multivariate analyses. SATB1 expression was also positively correlated with Ki67 expression but negatively with MGMT promoter methylation in glioma tissues. SATB1 shRNA expression vectors could efficiently induce the expression of SLC22A18 protein, increase the caspase-3 protein, inhibit the expression of SATB1, c-Met and bcl-2 protein, the growth, invasion, metastasis and angiogenesis of U251 cells, and induce apoptosis in vitro. Furthermore, the tumor growth of U251 cells expressing SATB1 shRNA were inhibited in vivo, and immunohistochemical analyses of tumor sections revealed a decreased vessel density in the animals where shRNA against SATB1 were expressed. CONCLUSIONS: SATB1 may have an important role as a positive regulator of glioma development and progression, and that SATB1 might be a useful molecular marker for predicting the prognosis of glioma.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Up-Regulation , Animals , Blotting, Western , Brain Neoplasms/pathology , Cell Adhesion , Cell Line, Tumor , DNA Methylation , Disease Progression , Fluorescent Antibody Technique , Glioma/pathology , Humans , Immunohistochemistry , Matrix Attachment Region Binding Proteins/genetics , Mice , Neoplasm Invasiveness , Neovascularization, Pathologic , Promoter Regions, Genetic , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
10.
J Clin Neurosci ; 19(1): 95-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22153794

ABSTRACT

We investigated the expression of the putative tumor suppressor SLC22A18 to evaluate it as a prognostic marker in glioma patients. Immunohistochemical and Western blot analyses of clinical tissue samples obtained from 120 patients with glioma were performed. Low expression of SLC22A18 was observed in 71.7% of patients. Loss of SLC22A18 expression in glioma was significantly related to pathological grade (p = 0.003). High pathological grade (World Health Organization III-IV) was correlated with negative (low or absent) expression of SLC22A18, which was correlated with a significantly shorter overall patient survival than in those with positive (high) expression (p = 0.007). Multivariate Cox regression analysis indicated that SLC22A18 expression level is an independent survival prognostic factor for patients with glioma (p = 0.011). Western blotting analysis confirmed decreased expression of SLC22A18 in glioma tissues compared with adjacent brain tissues. This study suggests that SLC22A18 functions as a tumor suppressor in glioma and represents a candidate biomarker for long-term survival in this disease.


Subject(s)
Biomarkers, Tumor/deficiency , Brain Neoplasms/diagnosis , Brain Neoplasms/metabolism , Down-Regulation/physiology , Glioma/diagnosis , Glioma/metabolism , Organic Cation Transport Proteins/deficiency , Adult , Aged , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/physiology , Brain Neoplasms/mortality , Female , Glioma/mortality , Humans , Male , Middle Aged , Organic Cation Transport Proteins/biosynthesis , Organic Cation Transport Proteins/physiology , Prognosis , Survival Rate , Tumor Suppressor Proteins/deficiency , Young Adult
11.
J Transl Med ; 9: 156, 2011 Sep 21.
Article in English | MEDLINE | ID: mdl-21936894

ABSTRACT

BACKGROUND: Downregulation of the putative tumor suppressor gene SLC22A18 has been reported in a number of human cancers. The aim of this study was to investigate the relationship between SLC22A18 downregulation, promoter methylation and the development and progression of human glioma. METHOD: SLC22A18 expression and promoter methylation was examined in human gliomas and the adjacent normal tissues. U251 glioma cells stably overexpressing SLC22A18 were generated to investigate the effect of SLC22A18 on cell growth and adherence in vitro using the methyl thiazole tetrazolium assay. Apoptosis was quantified using flow cytometry and the growth of SLC22A18 overexpressing U251 cells was measured in an in vivo xenograft model. RESULTS: SLC22A18 protein expression is significantly decreased in human gliomas compared to the adjacent normal brain tissues. SLC22A18 protein expression is significantly lower in gliomas which recurred within six months after surgery than gliomas which did not recur within six months. SLC22A18 promoter methylation was detected in 50% of the gliomas, but not in the adjacent normal tissues of any patient. SLC22A18 expression was significantly decreased in gliomas with SLC22A18 promoter methylation, compared to gliomas without methylation. The SLC22A18 promoter is methylated in U251 cells and treatment with the demethylating agent 5-aza-2-deoxycytidine increased SLC22A18 expression and reduced cell proliferation. Stable overexpression of SLC22A18 inhibited growth and adherence, induced apoptosis in vitro and reduced in vivo tumor growth of U251 cells. CONCLUSION: SLC22A18 downregulation via promoter methylation is associated with the development and progression of glioma, suggesting that SLC22A18 is an important tumor suppressor in glioma.


Subject(s)
DNA Methylation/genetics , Disease Progression , Down-Regulation/genetics , Glioma/genetics , Glioma/pathology , Organic Cation Transport Proteins/genetics , Promoter Regions, Genetic , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Azacitidine/pharmacology , Blotting, Western , Brain/drug effects , Brain/metabolism , Cell Adhesion/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Methylation/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Grading , Neurons/drug effects , Neurons/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Organic Cation Transport Proteins/metabolism , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recurrence
12.
J Neurooncol ; 80(2): 143-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16648987

ABSTRACT

C-Met, a receptor tyrosine kinase, and its ligand, hepatocyte growth factor (HGF), are critical in cellular proliferation, motility, and invasion, and are known to be overexpressed in gliomas, which are related to the repair of damaged DNA. In this study, we investigated both in vitro and in vivo whether inhibition of the c-Met gene by antisense oligonucleotides (ODNs) enhances the cytotoxic effect of radiation on human U251 gliomas. A volume of 100 nM of c-Met antisense ODNs inhibited the level of mRNA by more than 95% and reduced the protein expression by about 70%. Treatment of human U251 glioma cells with 100 nM of c-Met antisense ODNs significantly enhanced the radiation-induced cell kill compared to control cells, and cells treated with nonsense ODNs. When the glioma cells were implanted in the cisterna magna of nude mice followed by treatment with c-Met antisense ODNs, the survival time of the nude mice was markedly prolonged compared to that of the untreated group (P < 0.001, logrank test). In addition, the combination of antisense ODNs and irradiation extended the survival time of the glioma-bearing nude mice much longer than could be achieved with radiation alone (P < 0.0001, logrank test). These results suggest that inhibition of c-Met can be expected to serve as a novel potentiator for radiation therapy in human U251 gliomas.


Subject(s)
Central Nervous System Neoplasms/therapy , Glioma/therapy , Oligonucleotides, Antisense/therapeutic use , Proto-Oncogene Proteins c-met/genetics , Animals , Blotting, Western , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/radiotherapy , Combined Modality Therapy , Glioma/drug therapy , Glioma/radiotherapy , Hepatocyte Growth Factor/genetics , Mice , Mice, Nude , Microscopy, Fluorescence , Neoplasm Transplantation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis
13.
J Neurooncol ; 77(2): 117-23, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16314954

ABSTRACT

To examine the role of focal adhesion kinase in human glioma cells, we studied its effects on proliferation and apoptosis using FAK antisense oligonucleotide. U251 MG cells were transfected with ODNs, sense FAK, mismatch FAK and antisense-FAK, respectively. Expression of FAK proteins were detected by Western blots and Immnofluoressence. Cell apoptosis and mitochondrial membrane potential were analyzed by flow cytometry. Caspase-3 activity was measured by spectrofluorometer. MTT assay was used to examine changes in cell proliferation. The protein expression of FAK in U251 MG cells decreased in antisense-FAK ODNs group significantly. Caspase-3 activity increased in cells treated with antisense-FAK and down-regulated when treated with caspase-3 inhibitor. The level of cell apoptosis and loss of mitochondrial membrane potential in antisense-FAK group was higher than in the mismatch sense group. Cells proliferation was inhibited by antisense-FAK, and the effects were clearly additive when antisense oligonuceotides were added to cells treated with the anticancer agents. The results suggest that antisense-FAK ODNs inhibit U251 MG cells proliferation and induce their apoptosis. It is possible that FAK via mitochondrial and caspase-3 inhibits U251 MG cells apoptosis. And antisense oligonucleotide treatment enhances U251 MG cells sensitivity to chemotherapy.


Subject(s)
Apoptosis/drug effects , Brain Neoplasms/enzymology , Cell Proliferation/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glioma/enzymology , Oligonucleotides, Antisense/pharmacology , Antineoplastic Agents/pharmacology , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , Drug Synergism , Flow Cytometry , Focal Adhesion Protein-Tyrosine Kinases/genetics , Humans , Immunohistochemistry , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Transfection
14.
Zhonghua Yi Xue Za Zhi ; 85(12): 835-8, 2005 Mar 30.
Article in Chinese | MEDLINE | ID: mdl-15949402

ABSTRACT

OBJECTIVE: To investigate the expression of hepatocyte growth factor (HGF) mRNA and its receptor (c-Met) mRNA in brain astrocytomas and their relationships with tumor proliferation, angiogenesis, clinical pathology and prognosis. METHODS: The expression of HGF mRNA, c-Met mRNA in the resected tumor tissues of 76 patients with brain astrocytomas, 43 males and 33 females, aged 20 - 71, were detected by in situ hybridization. Immunohistochemistry technique was used to test the expression of proliferating cell nuclear antigen (PCNA) protein and the microvessel density (MVD) was determined by immunohistochemistry with monoclonal antibody against CD34. RESULTS: The positive rates of expression of HGF, c-Met and PCNA in low pathologic grades of brain astrocytoma were 34.5%, 44.8% and 15% +/- 9% respectively, and in high pathologic grades of brain astrocytoma were 34.5%, 44.8% and 48% +/- 12% respectively (P < 0.05). MVD in low and high pathologic grades of brain astrocytoma were 17 +/- 7 and 31 +/- 13 respectively (P < 0.05). The expression of HGF, c-Met, PCNA and CD34 was not related to sex, age, position of tumor and diameter of tumor. The expression of c-Met was related to the expression of HGF, PCNA and the MVD in the tumor tissues of these patients. The pathological grade, position of tumor, HGF, c-Met, PCNA, MVD had a significant influence on the survival time. CONCLUSION: HGF/c-Met plays an important role in the formation and progression of the brain astrocytoma and can promote tumor proliferation and intratumoral microvascular formation, and is closely related to the prognosis of the patients.


Subject(s)
Astrocytoma/metabolism , Brain Neoplasms/metabolism , Hepatocyte Growth Factor/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Receptors, Growth Factor/biosynthesis , Adult , Aged , Biomarkers, Tumor , Female , Hepatocyte Growth Factor/genetics , Humans , Male , Middle Aged , Neovascularization, Pathologic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-met , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Growth Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...