Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Int J Mol Sci ; 21(21)2020 Oct 28.
Article in English | MEDLINE | ID: mdl-33126685

ABSTRACT

Bile acids (BAs) have been implicated in the development of oesophagitis, Barrett's oesophagus and oesophageal adenocarcinoma (OAC). However, whether BAs promote cancer invasiveness has not been elucidated. We evaluated the role of BAs, in particular deoxycholic acid (DCA), in OAC invasion. Migration and invasiveness in untreated and BA-treated oesophageal SKGT-4 cancer cells were evaluated. Activity and expression of different matrix metalloproteinases (MMPs) were determined by zymography, ELISA, PCR and Western blot. Finally, human OAC tissues were stained for MMP-10 by immunohistochemistry. It was found that SKGT-4 cells incubated with low concentrations of DCA had a significant increase in invasion. In addition, MMP-10 mRNA and protein expression were also increased in the presence of DCA. MMP-10 was found to be highly expressed both in-vitro and in-vivo in neoplastic OAC cells relative to non-neoplastic squamous epithelial cells. Our results show that DCA promotes OAC invasion and MMP-10 overexpression. This study will advance our understanding of the pathophysiological mechanisms involved in human OAC and shows promise for the development of new therapeutic strategies.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , Deoxycholic Acid/pharmacology , Esophageal Neoplasms/pathology , Esophagus/pathology , Gene Expression Regulation, Enzymologic/drug effects , Matrix Metalloproteinase 10/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Apoptosis , Case-Control Studies , Cell Movement , Cell Proliferation , Cholagogues and Choleretics/pharmacology , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/enzymology , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/enzymology , Esophageal Squamous Cell Carcinoma/pathology , Esophagus/drug effects , Esophagus/enzymology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Invasiveness , Prognosis , Tumor Cells, Cultured
2.
J Cell Mol Med ; 21(12): 3612-3625, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28941013

ABSTRACT

The fundamental mechanisms underlying erosive oesophagitis and subsequent development of Barrett's oesophagus (BO) are poorly understood. Here, we investigated the contribution of specific components of the gastric refluxate on adhesion molecules involved in epithelial barrier maintenance. Cell line models of squamous epithelium (HET-1A) and BO (QH) were used to examine the effects of bile acids on cell adhesion to extracellular matrix proteins (Collagen, laminin, vitronectin, fibronectin) and expression of integrin ligands (α3 , α4, α5 , α6 and αν ). Experimental findings were validated in human explant oesophageal biopsies, a rat model of gastroesophageal reflux disease (GORD) and in patient tissue microarrays. The bile acid deoxycholic acid (DCA) specifically reduced adhesion of HET-1A cells to vitronectin and reduced cell-surface expression of integrin-αν via effects on endocytic recycling processes. Increased expression of integrin-αv was observed in ulcerated tissue in a rat model of GORD and in oesophagitis and Barrett's intestinal metaplasia patient tissue compared to normal squamous epithelium. Increased expression of integrin-αν was observed in QH BO cells compared to HET-1A cells. QH cells were resistant to DCA-mediated loss of adhesion and reduction in cell-surface expression of integrin-αν . We demonstrated that a specific component of the gastric refluxate, DCA, affects the epithelial barrier through modulation of integrin αν expression, providing a novel mechanism for bile acid-mediated erosion of oesophageal squamous epithelium and promotion of BO. Strategies aimed at preventing bile acid-mediated erosion should be considered in the clinical management of patients with GORD.


Subject(s)
Barrett Esophagus/metabolism , Deoxycholic Acid/pharmacology , Epithelial Cells/drug effects , Esophagitis/metabolism , Gastroesophageal Reflux/metabolism , Integrin alphaV/genetics , Animals , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Cell Adhesion , Cell Line , Collagen/chemistry , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Esophagitis/genetics , Esophagitis/pathology , Fibronectins/chemistry , Gastroesophageal Reflux/genetics , Gastroesophageal Reflux/pathology , Gene Expression Profiling , Gene Expression Regulation , Humans , Integrin alphaV/metabolism , Integrins/genetics , Integrins/metabolism , Laminin/chemistry , Permeability/drug effects , Protein Transport , Rats , Tissue Array Analysis , Vitronectin/chemistry
3.
Bioorg Med Chem Lett ; 26(21): 5369-5372, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27729186

ABSTRACT

Tauroursodeoxycholic acid (TUDCA) is a cytoprotective ER stress inhibitor and chemical chaperone. It has therapeutic potential in a wide array of diseases but a specific macromolecular target or molecular mechanism of action remains obscure. This Letter describes an effective new synthetic approach to taurine conjugation of bile acids which we used to prepare 3α-dansyl TUDCA (4) as a probe for TUDCA actions. As a model of ER stress we used the hepatocarcinoma cell line HUH7 and stimulation with either deoxycholic acid (DCA, 200µM) or tunicamycin (5µg/ml) and measured levels of Bip/GRP78, ATF4, CHOP and XBP1s/XBP1u. Compound 4 was more effective than UDCA at inhibiting ER stress markers and had similar effects to TUDCA. In a model of cholestasis using the cytotoxic DCA to induce apoptosis, pretreatment with 4 prevented cell death similarly to TUDCA whereas the unconjugated clinically used UDCA had no effect. 3α-Dansyl TUDCA (4) appears to be a suitable reporter for TUDCA effects on ER stress and related cytoprotective activity.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Fluorescent Dyes/chemistry , Taurochenodeoxycholic Acid/pharmacology , Animals , Endoplasmic Reticulum Chaperone BiP , Humans
4.
Rheumatol Int ; 36(3): 311-24, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26563338

ABSTRACT

The objective of this study was to systematically review the evidence from randomised controlled trials (RCTs) evaluating the effectiveness of multidisciplinary team (MDT) care for the management of disability, disease activity and quality of life (QoL) in adults with rheumatoid arthritis (RA). Data sources identified published (MEDLINE, PsychINFO, EMBASE, CINAHL, Web of Science, CENTRAL) and unpublished (OpenGrey) literature. Independent data extraction and quality assessment, using the Cochrane risk of bias tool, were conducted by two reviewers. The primary outcome was change in disability at 12 months; secondary outcomes included disability at other time points and disease activity and QoL at 12 months. Where possible, the pooled effect sizes were calculated for inpatient or outpatient MDT interventions. Four hundred and fifteen studies were retrieved. Twelve manuscripts, which reported 10 RCTs, representing 1147 participants were included. Only data from five high- or moderate-quality trials were pooled according to clinical setting. There was no difference in disability between inpatient MDT care and any comparison group [mean difference (95% confidence intervals) 0.04, -0.13 to 0.20] or between outpatient MDT care and comparison groups (0.09, -0.07 to 0.25) at 12 months. There was no difference in disability at 2 years or <12 months or disease activity and QoL at 12 months between MDT care and any comparison group. There is limited evidence evaluating the effect of MDT care on disability, disease activity or QoL in people with RA. There is likely to be no effect of MDT care on disability at 12 months or other time points.


Subject(s)
Arthritis, Rheumatoid/therapy , Delivery of Health Care, Integrated , Patient Care Team , Adult , Aged , Aged, 80 and over , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/physiopathology , Arthritis, Rheumatoid/psychology , Chi-Square Distribution , Combined Modality Therapy , Disability Evaluation , Female , Humans , Male , Middle Aged , Quality of Life , Remission Induction , Severity of Illness Index , Time Factors , Treatment Outcome , Young Adult
5.
Br J Cancer ; 113(9): 1332-42, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26461057

ABSTRACT

BACKGROUND: The aetiology of Barrett's oesophagus (BO) and oesophageal cancer is poorly understood. We previously demonstrated that Golgi structure and function is altered in oesophageal cancer cells. A Golgi-associated protein, GOLPH2, was previously established as a tissue biomarker for BO. Cellular functions for GOLPH2 are currently unknown, therefore in this study we sought to investigate functional roles for this Golgi-associated protein in oesophageal disease. METHODS: Expression, intracellular localisation and secretion of GOLPH2 were identified by immunofluorescence, immunohistochemistry and western blot. GOLPH2 expression constructs and siRNA were used to identify cellular functions for GOLPH2. RESULTS: We demonstrate that the structure of the Golgi is fragmented and the intracellular localisation of GOLPH2 is altered in BO and oesophageal adenocarcinoma tissue. GOLPH2 is secreted by oesophageal cancer cells and GOLPH2 expression, cleavage and secretion facilitate cell migration and invasion. Furthermore, exposure of cells to DCA, a bile acid component of gastric refluxate and known tumour promoter for oesophageal cancer, causes disassembly of the Golgi structure into ministacks, resulting in cleavage and secretion of GOLPH2. CONCLUSIONS: This study demonstrates that GOLPH2 may be a useful tissue biomarker for oesophageal disease. We provide a novel mechanistic insight into the aetiology of oesophageal cancer and reveal novel functions for GOLPH2 in regulating tumour cell migration and invasion, important functions for the metastatic process in oesophageal cancer.


Subject(s)
Barrett Esophagus/genetics , Barrett Esophagus/pathology , Bile Acids and Salts/genetics , Cell Movement/genetics , Membrane Proteins/genetics , Neoplasm Invasiveness/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Barrett Esophagus/metabolism , Bile Acids and Salts/metabolism , Biomarkers, Tumor/genetics , Cell Line, Tumor , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , Humans , Neoplasm Invasiveness/pathology
6.
Glycobiology ; 22(5): 638-48, 2012 May.
Article in English | MEDLINE | ID: mdl-22223758

ABSTRACT

It is generally accepted that esophageal adenocarcinoma arises from a Barrett's metaplastic lesion. Altered glycoprotein expression has been demonstrated in tissue from patients with Barrett's esophagus and esophageal cancer but the mechanisms regarding such changes are unknown. The bile acid deoxycholic acid (DCA) alters many cell signaling pathways and is implicated in esophageal cancer progression. We have demonstrated that DCA disrupts Golgi structure and affects protein secretion and glycosylation processes in cell lines derived from normal squamous epithelium (HET-1A) and Barrett's metaplastic epithelium (QH). Cell surface expression of glycans was identified using carbohydrate-specific probes (wheat germ agglutinate, conconavalin A, peanut agglutinin, lithocholic acid and Ulex europaeus agglutinin) that monitored N-glycosylation, O-glycosylation and core fucosylation in resting and DCA-treated cells. DCA altered intracellular localization and reduced cell surface expression of N-acetyl-D-glucosamine, α-methyl-mannopyranoside (Man/Glc) and fucose in both cell lines. Furthermore, DCA reduced the expression of epithelial growth factor receptor and E-cadherin in a manner analogous to treatment of cells with the N-glycan biosynthesis inhibitor tunicamycin. This is the first study to identify an altered Golgi structure and glycomic profile in response to DCA in esophageal epithelial cells, a process which could potentially contribute to metaplasia, dysplasia and cancer of the esophagus.


Subject(s)
Deoxycholic Acid/pharmacology , Esophagus/drug effects , Fucose/metabolism , Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Cadherins/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Esophagus/cytology , Esophagus/metabolism , Glycosylation , Golgi Apparatus/drug effects , Humans
7.
Carcinogenesis ; 31(4): 737-44, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20093383

ABSTRACT

Deoxycholic acid (DCA) is a secondary bile acid that modulates signalling pathways in epithelial cells. DCA has been implicated in pathogenesis of colon carcinoma, particularly by activation of the protein kinase C (PKC) pathway. Ursodeoxycholic acid (UDCA), a tertiary bile acid, has been observed to have chemopreventive effects. The aim of this study was to investigate the effect of DCA and UDCA on the subcellular localization and activity of PKCeta and its downstream effects on Golgi structure in a colon cancer cell model. PKCeta expression was localized to the Golgi in HCT116 colon cancer cells. DCA induced fragmentation of the Golgi in these cells following activation of PKCeta and its downstream effector protein kinase D (PKD). Pretreatment of cells with UDCA or a glucocorticoid, dexamethasone, inhibited DCA-induced PKCeta/PKD activation and Golgi fragmentation. Knockdown of glucocorticoid receptor (GR) expression using small interfering RNA or inhibition using the GR antagonist mifepristone attenuated the inhibitory effect of UDCA on Golgi fragmentation. Elevated serum and faecal levels of DCA have been previously reported in patients with ulcerative colitis (UC) and colon cancer. Analysis of Golgi architecture in vivo using tissue microarrays revealed Golgi fragmentation in UC and colorectal cancer tissue. We have demonstrated that DCA can disrupt the structure of the Golgi, an organelle critical for normal cell function. Inhibition of this DCA-induced Golgi fragmentation by UDCA was mediated via the GR. This represents a potential mechanism of observed chemopreventive effects of UDCA in benign and malignant disease of the colon.


Subject(s)
Bile Acids and Salts/pharmacology , Golgi Apparatus/drug effects , Protein Kinase C/physiology , Colitis, Ulcerative/pathology , Colonic Neoplasms/pathology , Deoxycholic Acid/pharmacology , Dexamethasone/pharmacology , Golgi Apparatus/ultrastructure , HCT116 Cells , Humans , Phosphorylation , Protein Kinase C/analysis , Receptors, Glucocorticoid/physiology , Ursodeoxycholic Acid/pharmacology
8.
Oncotarget ; 8(1): 967-978, 2017 Jan 03.
Article in English | MEDLINE | ID: mdl-27888615

ABSTRACT

Bile acids are components of gastro-duodenal refluxate and regarded as causative agents in oesophageal disease but the precise mechanisms are unknown. Here we demonstrate that a specific subset of physiological bile acids affect the protein secretory pathway by inducing ER stress, activating the Unfolded Protein Response (UPR) and causing disassembly of the Golgi apparatus in oesophageal cells. Deoxycholic acid (DCA), Chemodeoxycholic acid (CDCA) and Lithocholic acid (LCA) activated the PERK arm of the UPR, via phosphorylation of eIF2α and up-regulation of ATF3, CHOP and BiP/GRP78. UPR activation by these bile acids is mechanistically linked with Golgi fragmentation, as modulating the UPR using a PERK inhibitor (GSK2606414) or salubrinal attenuated bile acid-induced effects on Golgi structure. Furthermore we demonstrate that DCA, CDCA and LA activate Src kinase and that inhibition of this kinase attenuated both bile acid-induced BiP/GRP78 expression and Golgi fragmentation. This study highlights a novel mechanism whereby environmental factors (bile acids) impact important cellular processes regulating cell homeostasis, including the UPR and Golgi structure, which may contribute to cancer progression in the oesophagus.


Subject(s)
Bile Acids and Salts/pharmacology , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Unfolded Protein Response/drug effects , src-Family Kinases/metabolism , Bile Acids and Salts/metabolism , Cell Line, Tumor , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Esophageal Mucosa/metabolism , Humans , Models, Biological , Signal Transduction/drug effects , Unfolded Protein Response/genetics , eIF-2 Kinase/metabolism
9.
J Med Chem ; 54(1): 122-30, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21158453

ABSTRACT

Ursodeoxycholic acid (UDCA) is used for the treatment of hepatic inflammatory diseases. Recent studies have shown that UDCA's biological effects are partly glucocorticoid receptor (GR) mediated. UDCA derivatives were synthesized and screened for ability to induce GR translocation in a high content analysis assay using the esophageal cancer SKGT-4 cell line. UDCA derivatives induced GR translocation in a time dependent manner with equal efficacy to that of dexamethasone (Dex) and with greatly increased potency relative to UDCA. The cyclopropylamide 1a suppressed TNF-α induced NF-κB activity and it induced GRE transactivation. 1a was unable to displace Dex from the GR ligand binding domain (LBD) in a competition experiment but was capable of coactivator recruitment in a time-resolved fluorescence energy transfer assay (TR-FRET). This represents a novel mechanism of action for a GR modulator. These derivatives could result in a new class of GR modulators.


Subject(s)
Amides/chemical synthesis , Receptors, Glucocorticoid/metabolism , Ursodeoxycholic Acid/analogs & derivatives , Ursodeoxycholic Acid/chemical synthesis , Amides/pharmacology , Binding Sites , Binding, Competitive , Cell Line, Tumor , Dexamethasone/metabolism , Dexamethasone/pharmacology , Esophageal Neoplasms , Fluorescence Resonance Energy Transfer , Glucocorticoids/metabolism , Glucocorticoids/pharmacology , HEK293 Cells , Humans , Ligands , Models, Molecular , NF-kappa B/metabolism , Protein Transport , Radioligand Assay , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/genetics , Response Elements , Structure-Activity Relationship , Transcriptional Activation , Tumor Necrosis Factor-alpha/pharmacology , Ursodeoxycholic Acid/pharmacology
10.
J Cell Mol Med ; 9(4): 777-94, 2005.
Article in English | MEDLINE | ID: mdl-16364190

ABSTRACT

Vascular endothelial growth factor (VEGF) was originally identified as an endothelial cell specific growth factor stimulating angiogenesis and vascular permeability. Some family members, VEGF C and D, are specifically involved in lymphangiogenesis. It now appears that VEGF also has autocrine functions acting as a survival factor for tumour cells protecting them from stresses such as hypoxia, chemotherapy and radiotherapy. The mechanisms of action of VEGF are still being investigated with emerging insights into overlapping pathways and cross-talk between other receptors such as the neuropilins which were not previously associated with angiogenesis. VEGF plays an important role in embryonic development and angiogenesis during wound healing and menstrual cycle in the healthy adult. VEGF is also important in a number of both malignant and non-malignant pathologies. As it plays a limited role in normal human physiology, VEGF is an attractive therapeutic target in diseases where VEGF plays a key role. It was originally thought that in pathological conditions such as cancer, VEGF functioned solely as an angiogenic factor, stimulating new vessel formation and increasing vascular permeability. It has since emerged it plays a multifunctional role where it can also have autocrine pro-survival effects and contribute to tumour cell chemoresistance. In this review we discuss the established role of VEGF in angiogenesis and the underlying mechanisms. We discuss its role as a survival factor and mechanisms whereby angiogenesis inhibition improves efficacy of chemotherapy regimes. Finally, we discuss the therapeutic implications of targeting angiogenesis and VEGF receptors, particularly in cancer therapy.


Subject(s)
Neovascularization, Pathologic , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/physiology , Animals , Antineoplastic Agents/pharmacology , Arthritis, Rheumatoid/metabolism , Cell Movement , Cell Proliferation , Cell Survival , Diabetes Mellitus/metabolism , Exons , Gene Expression Regulation , Humans , Hypoxia , Ligands , Models, Biological , Neoplasms/metabolism , Protein Binding , Protein Isoforms , Psoriasis/metabolism , Reproduction , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Wound Healing
11.
Int J Cancer ; 101(5): 415-22, 2002 Oct 10.
Article in English | MEDLINE | ID: mdl-12216068

ABSTRACT

Endotoxin/lipopolysaccharide (LPS), a cell wall component of Gram-negative bacteria, is a potent inflammatory stimulus. We previously reported that LPS increased the growth of experimental metastases in a murine tumor model. Here, we examined the effect of LPS exposure on key determinants of metastasis-angiogenesis, tumor cell invasion, vascular permeability, nitric oxide synthase (NOS) and matrix metalloproteinase 2 (MMP2) expression. BALB/c mice bearing 4T1 lung metastases were given an intraperitoneal (i.p.) injection of 10 microg LPS or saline. LPS exposure resulted in increased lung weight and incidence of pleural lesions. LPS increased angiogenesis both in vivo and in vitro. Vascular permeability in lung tissue was increased 18 hr after LPS injection. LPS increased inducible nitric oxide synthase (iNOS) and MMP2 expression in lung tumor nodules. 4T1 cells transfected with green fluorescent protein (4T1-GFP) were injected via lateral tail vein. LPS exposure resulted in increased numbers of 4T1-GFP cells in mouse lung tissue compared to saline controls, an effect blocked by the competitive NOS inhibitor, N(G) methyl-L-arginine (NMA). LPS-induced growth and metastasis of 4T1 experimental lung metastases is associated with increased angiogenesis, vascular permeability and tumor cell invasion/migration with iNOS expression implicated in LPS-induced metastasis.


Subject(s)
Cell Division/drug effects , Lipopolysaccharides/toxicity , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic/chemically induced , Animals , Cell Movement , Disease Models, Animal , Endothelial Growth Factors/pharmacology , Female , Guanosine Triphosphate/metabolism , Lung Neoplasms/pathology , Lymphokines/pharmacology , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL