Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cells ; 8(9)2019 09 06.
Article in English | MEDLINE | ID: mdl-31489941

ABSTRACT

Liver damage upon exposure to ionizing radiation, whether accidental or because of therapy can contribute to liver dysfunction. Currently, radiation therapy is used for various cancers including hepatocellular carcinoma; however, the treatment dose is limited by poor liver tolerance to radiation. Furthermore, reliable biomarkers to predict liver damage and associated side-effects are unavailable. Here, we investigated fibrinogen-like 1 (FGL1)-expression in the liver and plasma after radiation exposure. We found that 30 Gy of liver irradiation (IR) induced cell death including apoptosis, necrosis, and autophagy, with fibrotic changes in the liver occurring during the acute and subacute phase in mice. Moreover, FGL1 expression pattern in the liver following IR was associated with liver damage represented by injury-related proteins and oxidative stress markers. We confirmed the association between FGL1 expression and hepatocellular injury by exposing human hepatocytes to radiation. To determine its suitability, as a potential biomarker for radiation-induced liver injury, we measured FGL1 in the liver tissue and the plasma of mice following total body irradiation (TBI) or liver IR. In TBI, FGL1 showed the highest elevation in the liver compared to other major internal organs including the heart, lung, kidney, and intestine. Notably, plasma FGL1 showed good correlation with radiation dose by liver IR. Our data revealed that FGL1 upregulation indicates hepatocellular injury in response to IR. These results suggest that plasma FGL1 may represent a potential biomarker for acute and subacute radiation exposure to the liver.


Subject(s)
Fibrinogen/metabolism , Liver Cirrhosis/blood , Liver/radiation effects , Radiation Injuries, Experimental/blood , Animals , Apoptosis , Autophagy , Biomarkers/blood , Cells, Cultured , Hepatocytes/metabolism , Hepatocytes/radiation effects , Humans , Liver/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred BALB C , Radiation Injuries, Experimental/pathology , Radiation, Ionizing
2.
Nat Commun ; 7: 13593, 2016 12 14.
Article in English | MEDLINE | ID: mdl-27966538

ABSTRACT

Overexpression of NQO1 is associated with poor prognosis in human cancers including breast, colon, cervix, lung and pancreas. Yet, the molecular mechanisms underlying the pro-tumorigenic capacities of NQO1 have not been fully elucidated. Here we show a previously undescribed function for NQO1 in stabilizing HIF-1α, a master transcription factor of oxygen homeostasis that has been implicated in the survival, proliferation and malignant progression of cancers. We demonstrate that NQO1 directly binds to the oxygen-dependent domain of HIF-1α and inhibits the proteasome-mediated degradation of HIF-1α by preventing PHDs from interacting with HIF-1α. NQO1 knockdown in human colorectal and breast cancer cell lines suppresses HIF-1 signalling and tumour growth. Consistent with this pro-tumorigenic function for NQO1, high NQO1 expression levels correlate with increased HIF-1α expression and poor colorectal cancer patient survival. These results collectively reveal a function of NQO1 in the oxygen-sensing mechanism that regulates HIF-1α stability in cancers.


Subject(s)
Breast Neoplasms/genetics , Colorectal Neoplasms/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , NAD(P)H Dehydrogenase (Quinone)/physiology , Proteasome Endopeptidase Complex/physiology , Breast Neoplasms/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Gene Knockdown Techniques , Homeostasis , Humans , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxygen/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Stability
3.
Mol Med Rep ; 13(6): 4666-70, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27082939

ABSTRACT

Radiation-induced lung injury (RILI) involves pneumonitis and fibrosis, and results in pulmonary dysfunction. Moreover, RILI can be a fatal complication of thoracic radiotherapy. The present study investigated the protective effect of geranylgeranlyacetone (GGA), an inducer of heat shock protein (HSP)70, on RILI using a C57BL/6 mouse model of RILI developing 6 months subsequent to exposure to 12.5 Gy thoracic radiation. GGA was administered 5 times orally prior and subsequent to radiation exposure, and the results were assessed by histological analysis and western blotting. The results show that late RILI was alleviated by GGA treatment, possibly through the suppression of epithelial­to­mesenchymal transition (EMT) marker expression. Based on histological examination, orally administered GGA during the acute phase of radiation injury not only significantly inhibited pro­surfactant protein C (pro­SPC) and vimentin expression, but also preserved E­cadherin expression 6 months after irradiation­induced injury of the lungs. GGA induced HSP70 and inhibited EMT marker expression in L132 human lung epithelial cells following IR. These data suggest that the prevention of EMT signaling is a key cytoprotective effect in the context of RILI. Thus, HSP70­inducing drugs, such as GGA, could be beneficial for protection against RILI.


Subject(s)
Diterpenes/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Radiation Pneumonitis/metabolism , Radiation Pneumonitis/pathology , Signal Transduction/drug effects , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/radiation effects , Animals , Cell Line , Disease Models, Animal , Female , Gene Expression , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Mice , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Radiation Injuries/drug therapy , Radiation Injuries/metabolism , Radiation Injuries/pathology , Radiation Pneumonitis/drug therapy
4.
PLoS One ; 10(6): e0128552, 2015.
Article in English | MEDLINE | ID: mdl-26029925

ABSTRACT

Radiation enteropathy is a common complication in cancer patients. The aim of this study was to investigate whether radiation-induced intestinal injury could be alleviated by coniferyl aldehyde (CA), an HSF1-inducing agent that increases cellular HSP70 expression. We systemically administered CA to mice with radiation enteropathy following abdominal irradiation (IR) to demonstrate the protective effects of CA against radiation-induced gastrointestinal injury. CA clearly alleviated acute radiation-induced intestinal damage, as reflected by the histopathological data and it also attenuated sub-acute enteritis. CA prevented intestinal crypt cell death and protected the microvasculature in the lamina propria during the acute and sub-acute phases of damage. CA induced HSF1 and HSP70 expression in both intestinal epithelial cells and endothelial cells in vitro. Additionally, CA protected against not only the apoptotic cell death of both endothelial and epithelial cells but also the loss of endothelial cell function following IR, indicating that CA has beneficial effects on the intestine. Our results provide novel insight into the effects of CA and suggest its role as a therapeutic candidate for radiation-induced enteropathy due to its ability to promote rapid re-proliferation of the intestinal epithelium by the synergic effects of the inhibition of cell death and the promotion of endothelial cell function.


Subject(s)
Acrolein/analogs & derivatives , Cell Death/drug effects , Endothelial Cells/drug effects , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/pharmacology , Acrolein/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Death/radiation effects , Cell Line , Cell Line, Tumor , Endothelial Cells/radiation effects , Enteritis/drug therapy , Epithelial Cells/drug effects , Epithelial Cells/radiation effects , Human Umbilical Vein Endothelial Cells , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/radiation effects , Intestines/drug effects , Intestines/radiation effects , Mice , Mice, Inbred C3H , Radiation , Rats
5.
Mol Cancer Res ; 7(12): 2011-21, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19996305

ABSTRACT

Although multiple studies have revealed that gallic acid plays an important role in the inhibition of malignant transformation, cancer development, and inflammation, the molecular mechanism of gallic acid in inflammatory diseases is still unclear. In this study, we identified gallic acid from Rosa rugosa as a histone acetyltransferase (HAT) inhibitor with global specificity for the majority of HAT enzymes, but with no activity toward epigenetic enzymes including sirtuin (silent mating type information regulation 2 homologue) 1 (S. cerevisiae), histone deacetylase, and histone methyltransferase. Enzyme kinetic studies indicated that gallic acid uncompetitively inhibits p300/CBP-dependent HAT activities. We found that gallic acid inhibits p300-induced p65 acetylation, both in vitro and in vivo, increases the level of cytosolic IkappaBalpha, prevents lipopolysaccharide (LPS)-induced p65 translocation to the nucleus, and suppresses LPS-induced nuclear factor-kappaB activation in A549 lung cancer cells. We have also shown that gallic acid treatment inhibits the acetylation of p65 and the LPS-induced serum levels of interleukin-6 in vivo. Importantly, gallic acid generally inhibited inflammatory responses caused by other stimuli, including LPS, IFN-gamma, and interleukin-1beta, and further downregulated the expression of nuclear factor-kappaB-regulated antiapoptotic genes. These results show the crucial role of acetylation in the development of inflammatory diseases.


Subject(s)
Gallic Acid/pharmacology , Lipopolysaccharides/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Acetylation/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , E1A-Associated p300 Protein/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Histone Acetyltransferases/antagonists & inhibitors , Humans , Inflammation/pathology , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Male , Mice , Mice, Inbred BALB C , Rosaceae/chemistry
6.
Cancer Res ; 69(2): 583-92, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19147572

ABSTRACT

Because the p300/CBP-mediated hyperacetylation of RelA (p65) is critical for nuclear factor-kappaB (NF-kappaB) activation, the attenuation of p65 acetylation is a potential molecular target for the prevention of chronic inflammation. During our ongoing screening study to identify natural compounds with histone acetyltransferase inhibitor (HATi) activity, we identified epigallocatechin-3-gallate (EGCG) as a novel HATi with global specificity for the majority of HAT enzymes but with no activity toward epigenetic enzymes including HDAC, SIRT1, and HMTase. At a dose of 100 micromol/L, EGCG abrogates p300-induced p65 acetylation in vitro and in vivo, increases the level of cytosolic IkappaBalpha, and suppresses tumor necrosis factor alpha (TNFalpha)-induced NF-kappaB activation. We also showed that EGCG prevents TNFalpha-induced p65 translocation to the nucleus, confirming that hyperacetylation is critical for NF-kappaB translocation as well as activity. Furthermore, EGCG treatment inhibited the acetylation of p65 and the expression of NF-kappaB target genes in response to diverse stimuli. Finally, EGCG reduced the binding of p300 to the promoter region of interleukin-6 gene with an increased recruitment of HDAC3, which highlights the importance of the balance between HATs and histone deacetylases in the NF-kappaB-mediated inflammatory signaling pathway. Importantly, EGCG at 50 micromol/L dose completely blocks EBV infection-induced cytokine expression and subsequently the EBV-induced B lymphocyte transformation. These results show the crucial role of acetylation in the development of inflammatory-related diseases.


Subject(s)
B-Lymphocytes/virology , Catechin/analogs & derivatives , Cell Transformation, Viral/drug effects , Herpesvirus 4, Human/physiology , Histone Acetyltransferases/antagonists & inhibitors , Transcription Factor RelA/metabolism , Acetylation/drug effects , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Catechin/pharmacology , Cell Line, Tumor , Down-Regulation/drug effects , HeLa Cells , Humans , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology
7.
J Ethnopharmacol ; 118(3): 412-7, 2008 Aug 13.
Article in English | MEDLINE | ID: mdl-18562138

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Rosa rugosa Thunb. (Rosaceae) has been traditionally used for treatments of diabetes, chronic inflammatory diseases, pain, and anticancer in Korea. AIM OF STUDY: We investigate the inhibitory effect of histone acetyltransferase activity from the methanol extract of stems of Rosa rugosa on androgen receptor-mediated transcriptional regulation. MATERIALS AND METHODS: For the present study, Rosa rugosa methanol extract (RRME) was obtained from stem part of Rosa rugosa using methanol extraction. Histone acetyltransferase assay were performed to measure the inhibitory effect on acetylation, reporter assay, real-time PCR and ChIP assay were performed to measure androgen receptor-mediated transcriptional regulation, and MTT test were performed to measure cell viability. RESULTS: RRME inhibited both p300 and CBP (60-70% at 100 microg/ml) activity. We show RRME mediates agonist-dependent androgen receptor (AR) activation and suppresses antagonist-dependent inhibition. RRME treatment also decreased transcription of AR regulated genes and also reduced histone H3 and AR acetylation in the promoters of prostate-specific antigen (PSA) and beta-2-microglobulin (B2M). Finally, RRME treatment reduced the growth of LNCaP, a human prostate cancer cell line. CONCLUSION: These results demonstrate RRME is a potent HAT inhibitor, which reduced AR and histone acetylation leading to decreased AR-mediated transcription and reduced LNCaP cell growth.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Acetyltransferases/antagonists & inhibitors , Plant Extracts/pharmacology , Receptors, Androgen/physiology , Rosa , Apoptosis/drug effects , Cell Line, Tumor , Female , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Rosa/chemistry , Transcription, Genetic
8.
Biosci Biotechnol Biochem ; 71(11): 2712-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17986787

ABSTRACT

Histone acetylation depends on the activity of two enzyme families, histone acetyltransferase (HAT) and deacetylase (HDAC). In this study, we screened various plant extracts to find potent HAT inhibitors. Hot water extracts of allspice inhibited HAT activity, especially p300 and CBP (40% at 100 microg/ml). The mRNA levels of two androgen receptor (AR) regulated genes, PSA and TSC22, decreased with allspice treatment (100 microg/ml). Importantly, in IP western analysis, AR acetylation was dramatically decreased by allspice treatment.Furthermore, chromatin immunoprecipitation indicated that the acetylation of histone H3 in the PSA and B2M promoter regions was also repressed. Finally, allspice treatment reduced the growth of human prostate cancer cells, LNCaP (50% growth inhibition at 200 microg/ml). Taken together, our data indicate that the potent HAT inhibitory activity of allspice reduced AR and histone acetylation and led to decreased transcription of AR target genes, resulting in inhibition of prostate cancer cell growth.


Subject(s)
Androgen Receptor Antagonists , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Acetyltransferases/antagonists & inhibitors , Pimenta , Prostatic Neoplasms/enzymology , Acetylation , Carboxypeptidase B/antagonists & inhibitors , Carboxypeptidase B/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Histones/metabolism , Humans , Male , Plant Extracts/pharmacology , Receptors, Androgen/metabolism , Transcriptional Activation/drug effects , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...