Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Pancreatology ; 20(2): 278-287, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31831391

ABSTRACT

BACKGROUND/OBJECTIVES: Fibromodulin (FMOD) expression in chronic pancreatitis (CP) tissues and its effect on PSC was unknown. Our aim was to investigate the role of FMOD in regulating PSC profibrogenic phenotype and the molecular mechanism of CP. METHODS: Rat CP models were induced by dibutyltin dichloride. Pancreatic fibrosis was evaluated by Sirius Red staining. The expression of FMOD and α-SMA was measured, the correlation between FMOD expression and fibrosis was investigated in CP models and CP patients. The effects of FMOD on PSCs were examined by CCK-8 and migration assays. We investigated the mechanisms underlying FMOD expression using MND and a MAPK pathway inhibitor. Luciferase reporter and chromatin immunoprecipitation assays were used to investigate the effects of AP-1 on FMOD expression. RESULTS: Sirius Red staining revealed high collagen deposition in model rats. Higher expression of FMOD and α-SMA was observed in fibrotic tissues, and the expression of FMOD was correlated with that of α-SMA and the areas of Sirius Red staining. Upregulation of FMOD increased the expression of collagen I and α-SMA and the proliferation and migration of PSCs. MND induced FMOD and α-SMA expression, and knockdown of FMOD abated α-SMA expression. ERK and JNK inhibitors attenuated FMOD expression as induced by MND. AP-1 upregulated the expression of FMOD. AP-1 binds to the FMOD promoter and transcriptionally regulates FMOD expression. CONCLUSION: FMOD levels are upregulated in fibrosis tissues in CP and it is a critical downstream mediator of oxidative stress. FMOD induces PSC activation and maintains the fibrosis phenotype of PSCs.


Subject(s)
Fibromodulin/genetics , MAP Kinase Signaling System/genetics , Oxidative Stress , Pancreatic Stellate Cells/metabolism , Signal Transduction/genetics , Transcription Factor AP-1/metabolism , Actins/metabolism , Aged , Animals , Cells, Cultured , Fibromodulin/biosynthesis , Fibrosis/pathology , Humans , Male , Middle Aged , Rats , Rats, Wistar , Transcription Factor AP-1/genetics , Up-Regulation
2.
Med Sci Monit ; 24: 6137-6143, 2018 Sep 03.
Article in English | MEDLINE | ID: mdl-30176167

ABSTRACT

BACKGROUND Angiogenesis plays an important role in the progression of glioblastoma, with a high degree of malignancy. Previous studies have proved that glial cell line-derived neurotrophic factor (GDNF) and fibromodulin (FMOD) are strongly expressed in human glioblastoma. The purpose of this study was to explore the roles of GDNF and FMOD in angiogenesis and the molecular mechanisms underlying these roles in human glioblastoma. MATERIAL AND METHODS The effects of GDNF on the expression and secretion of vascular endothelial growth factor (VEGF) in human glioblastoma cell line U251 and angiogenesis in human umbilical vein endothelial cells (HUVECs) were investigated. The molecular mechanism of GDNF-induced expression of FMOD was explored. The roles of FMOD in GDNF-induced expression and secretion of VEGF and angiogenesis were also examined. RESULTS In the present study, we showed that GDNF promoted the expression and secretion of VEGF in U251 cells. VEGF mediated GDNF-induced angiogenesis in human glioblastoma. In addition, GDNF significantly upregulated the expression of FMOD in U251 cells. Mechanistically, the results of luciferase reporter assay and methylation-specific PCR (MSP) demonstrated that GDNF facilitated the demethylation of the FMOD promoter. More importantly, we found that FMOD acted as an important mediator in VEGF expression and angiogenesis induced by GDNF in human glioblastoma. CONCLUSIONS Collectively, our data show that GDNF promotes angiogenesis through demethylation of the FMOD promoter in human glioblastoma, indicating that GDNF and FMOD may be potential therapeutic targets for glioblastoma.


Subject(s)
DNA Methylation , Fibromodulin/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glioblastoma/blood supply , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Demethylation , Fibromodulin/biosynthesis , Fibromodulin/metabolism , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glioblastoma/genetics , Glioblastoma/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Promoter Regions, Genetic , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/metabolism
3.
PLoS One ; 13(7): e0201422, 2018.
Article in English | MEDLINE | ID: mdl-30052659

ABSTRACT

Pressure overload of the heart leads to cardiac remodeling that may progress into heart failure, a common, morbid and mortal condition. Increased mechanistic insight into remodeling is instrumental for development of novel heart failure treatment. Cardiac remodeling comprises cardiomyocyte hypertrophic growth, extracellular matrix alterations including fibrosis, and inflammation. Fibromodulin is a small leucine-rich proteoglycan that regulates collagen fibrillogenesis. Fibromodulin is expressed in the cardiac extracellular matrix, however its role in the heart remains largely unknown. We investigated fibromodulin levels in myocardial biopsies from heart failure patients and mice, subjected fibromodulin knock-out (FMOD-KO) mice to pressure overload by aortic banding, and overexpressed fibromodulin in cultured cardiomyocytes and cardiac fibroblasts using adenovirus. Fibromodulin was 3-10-fold upregulated in hearts of heart failure patients and mice. Both cardiomyocytes and cardiac fibroblasts expressed fibromodulin, and its expression was increased by pro-inflammatory stimuli. Without stress, FMOD-KO mice showed no cardiac phenotype. Upon aortic banding, left ventricles of FMOD-KO mice developed mildly exacerbated hypertrophic remodeling compared to wild-type mice, with increased cardiomyocyte size and altered infiltration of leukocytes. There were no differences in mortality, left ventricle dilatation, dysfunction or expression of heart failure markers. Although collagen amount and cross-linking were comparable in FMOD-KO and wild-type, overexpression of fibromodulin in cardiac fibroblasts in vitro decreased their migratory capacity and expression of fibrosis-associated molecules, i.e. the collagen-cross linking enzyme lysyl oxidase, transglutaminase 2 and periostin. In conclusion, despite a robust fibromodulin upregulation in clinical and experimental heart failure, FMOD-KO mice showed a relatively mild hypertrophic phenotype. In cultured cardiac fibroblasts, fibromodulin has anti-fibrotic effects.


Subject(s)
Cardiomegaly/metabolism , Extracellular Matrix/metabolism , Fibromodulin/biosynthesis , Heart Failure/metabolism , Myocardium/metabolism , Animals , Biomarkers , Cardiomegaly/genetics , Cardiomegaly/pathology , Disease Models, Animal , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fibromodulin/genetics , Heart Failure/genetics , Heart Failure/pathology , Humans , Male , Mice , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology
4.
Int J Biol Markers ; 31(2): e153-62, 2016 May 28.
Article in English | MEDLINE | ID: mdl-26689247

ABSTRACT

BACKGROUND: The aim of this study was to evaluate the gene expression profiles of a set of prostate cancer-associated genes in prostate cancer cell lines, to determine their association with different cancer phenotypes and identify potential novel biomarkers for this disease. METHODS: Quantitative real-time PCR was used to determine the expression profiles of 21 prostate cancer-associated genes in the human prostate cancer cell lines PC-3 and LNCaP, using the nontumorigenic cell line PWR-1E as control cell line. Genes evaluated were ESM-1, SERPINE2, CLU, BGN, A2M, PENK, FMOD, CD81, DCN, TSPAN8, KBTBD10, F2RL1, TMSB4X, SNCG, CXXC5, FOXQ1, PDPN, SPN, CAV1, CD24 and KLK3. A potential biomarker from this set of genes, the FMOD gene, encoding the small leucine-rich proteoglycan fibromodulin, was selected for further evaluation in clinical samples from patients diagnosed with benign or malignant prostatic disease. RESULTS: Several of the evaluated genes showed significantly altered expression in the prostate cancer cell lines, compared with nontumorigenic PWR-1E cells. Further evaluation of FMOD transcript in prostate clinical samples from patients diagnosed with benign or malignant prostatic disease identified a significant difference in the expression levels of this proteoglycan between benign and malignant tissue (p<0.05). CONCLUSIONS: A number of gene transcripts were differentially expressed by the cell lines assayed. Among them, FMOD was further evaluated in clinical samples and was found to be differentially expressed between benign and prostate cancer tissue. Further validation of FMOD transcript in a larger population is required to ascertain its usefulness as biomarker for prostate cancer.


Subject(s)
Biomarkers, Tumor/genetics , Fibromodulin/genetics , Prostatic Neoplasms/genetics , Biomarkers, Tumor/biosynthesis , Cell Line, Tumor , Fibromodulin/biosynthesis , Gene Expression Profiling , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...