Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
Pancreas ; 50(7): 994-999, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34629450

ABSTRACT

OBJECTIVES: The aim was to evaluate the relationship between cytoglobin (Cygb) expression and both clinicopathologic factors and prognosis in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS: Seventy-five patients with PDAC who underwent pancreatectomy between 2009 and 2014 at our department were included. Diagnosis was based on World Health Organization standards, with staging by TNM classification of Union for International Cancer Control. Expressions of Cygb, phosphoinositide-3 kinase, phosphorylated protein kinase B, interleukin-6, and vascular endothelial growth factor were evaluated by immunohistochemical staining of resected surgical specimens and densitometrical analysis. RESULTS: Elevated expression of Cygb was found mainly in carcinoma cells of PDAC. Patients with low expression of Cygb showed significantly shorter disease-free survival and disease-specific survival than those with high expression. There was also a significant negative correlation between Cygb expression and the expressions of phosphoinositide 3-kinase, phosphorylated protein kinase B, interleukin-6, and vascular endothelial growth factor. In univariate analysis, Cygb expression, clinical stage, histologic tumor grade, lymphatic invasion, and vascular invasion were prognostic factors. In multivariate analysis, Cygb expression and the clinical stage were independent prognostic factors. CONCLUSIONS: Loss of Cygb may contribute to tumor recurrence and poor prognosis of PDAC by increases in angiogenic factor.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cytoglobin/biosynthesis , Pancreatic Neoplasms/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Aged , Aged, 80 and over , Carcinoma, Pancreatic Ductal/pathology , Female , Humans , Immunohistochemistry/methods , Interleukin-6/biosynthesis , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/biosynthesis , Prognosis , Proto-Oncogene Proteins c-akt/biosynthesis , Retrospective Studies
2.
J Hepatol ; 73(4): 882-895, 2020 10.
Article in English | MEDLINE | ID: mdl-32330605

ABSTRACT

BACKGROUND & AIMS: Cytoglobin (CYGB) is a respiratory protein that acts as a scavenger of reactive oxygen species. The molecular role of CYGB in human hepatic stellate cell (HSC) activation and human liver disease remains uncharacterised. The aim of this study was to reveal the mechanism by which the TGF-ß1/SMAD2 pathway regulates the human CYGB promoter and the pathophysiological function of CYGB in human non-alcoholic steatohepatitis (NASH). METHODS: Immunohistochemical staining was performed using human NASH biopsy specimens. Molecular and biochemical analyses were performed by western blotting, quantitative PCR, and luciferase and immunoprecipitation assays. Hydroxyl radicals (•OH) and oxidative DNA damage were measured using an •OH-detectable probe and 8-hydroxy-2'-deoxyguanosine (8-OHdG) ELISA. RESULTS: In culture, TGF-ß1-pretreated human HSCs exhibited lower CYGB levels - together with increased NADPH oxidase 4 (NOX4) expression - and were primed for H2O2-triggered •OH production and 8-OHdG generation; overexpression of human CYGB in human HSCs reversed these effects. Electron spin resonance demonstrated the direct •OH scavenging activity of recombinant human CYGB. Mechanistically, pSMAD2 reduced CYGB transcription by recruiting the M1 repressor isoform of SP3 to the human CYGB promoter at nucleotide positions +2-+13 from the transcription start site. The same repression did not occur on the mouse Cygb promoter. TGF-ß1/SMAD3 mediated αSMA and collagen expression. Consistent with observations in cultured human HSCs, CYGB expression was negligible, but 8-OHdG was abundant, in activated αSMA+pSMAD2+- and αSMA+NOX4+-positive hepatic stellate cells from patients with NASH and advanced fibrosis. CONCLUSIONS: Downregulation of CYGB by the TGF-ß1/pSMAD2/SP3-M1 pathway brings about •OH-dependent oxidative DNA damage in activated hepatic stellate cells from patients with NASH. LAY SUMMARY: Cytoglobin (CYGB) is a respiratory protein that acts as a scavenger of reactive oxygen species and protects cells from oxidative DNA damage. Herein, we show that the cytokine TGF-ß1 downregulates human CYGB expression. This leads to oxidative DNA damage in activated hepatic stellate cells. Our findings provide new insights into the relationship between CYGB expression and the pathophysiology of fibrosis in patients with non-alcoholic steatohepatitis.


Subject(s)
Cytoglobin/genetics , Gene Expression Regulation , Hepatic Stellate Cells/metabolism , NADPH Oxidase 4/genetics , Non-alcoholic Fatty Liver Disease/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta1/metabolism , Biopsy , Cells, Cultured , Cytoglobin/biosynthesis , Down-Regulation , Female , Hepatic Stellate Cells/pathology , Humans , Liver/metabolism , Liver/pathology , Male , Middle Aged , NADPH Oxidase 4/biosynthesis , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress/genetics , Smad3 Protein/biosynthesis
3.
Sci Rep ; 8(1): 17860, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30552362

ABSTRACT

Cytoglobin (CYGB), discovered in hepatic stellate cells (HSCs), is known to possess a radical scavenger function, but its pathophysiological roles remain unclear. Here, for the first time, we generated a new transgenic (TG) mouse line in which both Cygb and mCherry reporter gene expression were under the control of the native Cygb gene promoter. We demonstrated that the expression of Cygb-mCherry was related to endogenous Cygb in adult tissues by tracing mCherry fluorescence together with DNA, mRNA, and protein analyses. Administration of a single dose (50 mg/kg) of thioacetamide (TAA) in Cygb-TG mice resulted in lower levels of alanine transaminase and oxidative stress than those in WT mice. After 10 weeks of TAA administration, Cygb-TG livers exhibited reduced neutrophil accumulation, cytokine expression and fibrosis but high levels of quiescent HSCs. Primary HSCs isolated from Cygb-TG mice (HSCCygb-TG) exhibited significantly decreased mRNA levels of α-smooth muscle actin (αSMA), collagen 1α1, and transforming growth factor ß-3 after 4 days in culture relative to WT cells. HSCsCygb-TG were resistant to H2O2-induced αSMA expression. Thus, cell-specific overexpression of Cygb attenuates HSC activation and protects mice against TAA-induced liver fibrosis presumably by maintaining HSC quiescence. Cygb is a potential new target for antifibrotic approaches.


Subject(s)
Cytoglobin/biosynthesis , Gene Expression , Hepatic Stellate Cells/enzymology , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/prevention & control , Thioacetamide/toxicity , Animals , Artificial Gene Fusion , Genes, Reporter , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Promoter Regions, Genetic , Thioacetamide/administration & dosage , Red Fluorescent Protein
4.
J Exp Clin Cancer Res ; 37(1): 313, 2018 Dec 13.
Article in English | MEDLINE | ID: mdl-30545372

ABSTRACT

BACKGROUND: Recent studies suggested the globin family member cytoglobin (CYGB) as a potential tumor suppressor; however, the mechanism by which CYGB suppresses cancer is elusive. We investigated the role and mechanism of CYGB in suppressing breast cancer. METHODS: CYGB expression was examined by reverse transcription PCR, quantitative reverse transcription PCR and open database analysis. Promoter methylation was examined by methylation-specific PCR. Metabolomics and proteomics were analyzed by gas chromatography-mass spectrometry and isobaric tags for relative and absolute quantitation, respectively. The effects and mechanisms of ectopic CYGB expression in breast cancer cells were assessed with molecular biological and cellular approaches in vitro and with a xenograft tumor model in nude mice. RESULTS: CYGB expression was downregulated in breast cancer tissues and cell lines, which was associated with promoter methylation. Ectopic CYGB expression suppressed proliferation, migration, invasion and induced apoptosis in breast cancer cell lines MCF7 (p53WT) and MB231 (p53mt) in vitro, and inhibited xenograft tumor growth in vivo. By proteomics and metabolomics analysis, glucose metabolism was found to be one of the main pathways suppressed by CYGB. The CYGB-expressing cells had lower ATP and compromised glycolysis. Additionally, CYGB suppressed key glucose metabolism factors including GLUT1 and HXK2 in p53-dependent and -independent manners. Restoration of GLUT1 or HXK2 expression attenuated CYGB-mediated proliferation suppression and apoptosis induction. CONCLUSIONS: CYGB is a potential tumor suppressor in breast cancer that is epigenetically suppressed. The results for the first time suggest that CYGB suppresses breast cancer through inhibiting glucose metabolism, which could be exploited for breast cancer prevention and therapy.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cytoglobin/metabolism , Glucose/antagonists & inhibitors , Glucose/metabolism , Animals , Apoptosis/physiology , Breast Neoplasms/pathology , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Cytoglobin/biosynthesis , Cytoglobin/genetics , Down-Regulation , Female , Heterografts , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude
5.
Nat Commun ; 9(1): 1021, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523849

ABSTRACT

Hyperproliferative keratinocytes induced by trauma, hyperkeratosis and/or inflammation display molecular signatures similar to those of palmoplantar epidermis. Inherited gain-of-function mutations in RHBDF2 (encoding iRHOM2) are associated with a hyperproliferative palmoplantar keratoderma and squamous oesophageal cancer syndrome (termed TOC). In contrast, genetic ablation of rhbdf2 in mice leads to a thinning of the mammalian footpad, and reduces keratinocyte hyperproliferation and migration. Here, we report that iRHOM2 is a novel target gene of p63 and that both p63 and iRHOM2 differentially regulate cellular stress-associated signalling pathways in normal and hyperproliferative keratinocytes. We demonstrate that p63-iRHOM2 regulates cell survival and response to oxidative stress via modulation of SURVIVIN and Cytoglobin, respectively. Furthermore, the antioxidant compound Sulforaphane downregulates p63-iRHOM2 expression, leading to reduced proliferation, inflammation, survival and ROS production. These findings elucidate a novel p63-associated pathway that identifies iRHOM2 modulation as a potential therapeutic target to treat hyperproliferative skin disease and neoplasia.


Subject(s)
Carrier Proteins/metabolism , Cell Proliferation/genetics , Esophageal Squamous Cell Carcinoma/pathology , Keratinocytes/metabolism , Oxidative Stress/genetics , Phosphoproteins/metabolism , Trans-Activators/metabolism , Animals , Apoptosis/genetics , Carrier Proteins/genetics , Cell Line , Cell Survival/genetics , Cytoglobin/biosynthesis , Female , HEK293 Cells , Humans , Isothiocyanates/pharmacology , Mice , Mice, Knockout , Phosphoproteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Skin Diseases/pathology , Sulfoxides , Survivin/biosynthesis , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL