Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Mol Metab ; 63: 101537, 2022 09.
Article in English | MEDLINE | ID: mdl-35772644

ABSTRACT

INTRODUCTION: Primary mitochondrial diseases (PMD) are a large, heterogeneous group of genetic disorders affecting mitochondrial function, mostly by disrupting the oxidative phosphorylation (OXPHOS) system. Understanding the cellular metabolic re-wiring occurring in PMD is crucial for the development of novel diagnostic tools and treatments, as PMD are often complex to diagnose and most of them currently have no effective therapy. OBJECTIVES: To characterize the cellular metabolic consequences of OXPHOS dysfunction and based on the metabolic signature, to design new diagnostic and therapeutic strategies. METHODS: In vitro assays were performed in skin-derived fibroblasts obtained from patients with diverse PMD and validated in pharmacological models of OXPHOS dysfunction. Proliferation was assessed using the Incucyte technology. Steady-state glucose and glutamine tracing studies were performed with LC-MS quantification of cellular metabolites. The therapeutic potential of nutritional supplements was evaluated by assessing their effect on proliferation and on the metabolomics profile. Successful therapies were then tested in a in vivo lethal rotenone model in zebrafish. RESULTS: OXPHOS dysfunction has a unique metabolic signature linked to an NAD+/NADH imbalance including depletion of TCA intermediates and aspartate, and increased levels of glycerol-3-phosphate. Supplementation with pyruvate and uridine fully rescues this altered metabolic profile and the subsequent proliferation deficit. Additionally, in zebrafish, the same nutritional treatment increases the survival after rotenone exposure. CONCLUSIONS: Our findings reinforce the importance of the NAD+/NADH imbalance following OXPHOS dysfunction in PMD and open the door to new diagnostic and therapeutic tools for PMD.


Subject(s)
Mitochondrial Diseases , Oxidative Phosphorylation , Animals , Metabolome , Mitochondrial Diseases/metabolism , NAD/metabolism , Pyruvic Acid/metabolism , Rotenone , Uridine/metabolism , Uridine/pharmacology , Zebrafish/metabolism
2.
Metabolites ; 9(10)2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31658717

ABSTRACT

Primary mitochondrial disease (PMD) is a large group of genetic disorders directly affecting mitochondrial function. Although next generation sequencing technologies have revolutionized the diagnosis of these disorders, biochemical tests remain essential and functional confirmation of the critical genetic diagnosis. While enzymological testing of the mitochondrial oxidative phosphorylation (OXPHOS) complexes remains the gold standard, oxygraphy could offer several advantages. To this end, we compared the diagnostic performance of both techniques in a cohort of 34 genetically defined PMD patient fibroblast cell lines. We observed that oxygraphy slightly outperformed enzymology for sensitivity (79 ± 17% versus 68 ± 15%, mean and 95% CI), and had a better discriminatory power, identifying 58 ± 17% versus 35 ± 17% as "very likely" for oxygraphy and enzymology, respectively. The techniques did, however, offer synergistic diagnostic prediction, as the sensitivity rose to 88 ± 11% when considered together. Similarly, the techniques offered varying defect specific information, such as the ability of enzymology to identify isolated OXPHOS deficiencies, while oxygraphy pinpointed PDHC mutations and captured POLG mutations that were otherwise missed by enzymology. In summary, oxygraphy provides useful information for the diagnosis of PMD, and should be considered in conjunction with enzymology for the diagnosis of PMD.

3.
Am J Hum Genet ; 104(5): 835-846, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30982613

ABSTRACT

Phosphoglucomutase 1 (PGM1) encodes the metabolic enzyme that interconverts glucose-6-P and glucose-1-P. Mutations in PGM1 cause impairment in glycogen metabolism and glycosylation, the latter manifesting as a congenital disorder of glycosylation (CDG). This unique metabolic defect leads to abnormal N-glycan synthesis in the endoplasmic reticulum (ER) and the Golgi apparatus (GA). On the basis of the decreased galactosylation in glycan chains, galactose was administered to individuals with PGM1-CDG and was shown to markedly reverse most disease-related laboratory abnormalities. The disease and treatment mechanisms, however, have remained largely elusive. Here, we confirm the clinical benefit of galactose supplementation in PGM1-CDG-affected individuals and obtain significant insights into the functional and biochemical regulation of glycosylation. We report here that, by using tracer-based metabolomics, we found that galactose treatment of PGM1-CDG fibroblasts metabolically re-wires their sugar metabolism, and as such replenishes the depleted levels of galactose-1-P, as well as the levels of UDP-glucose and UDP-galactose, the nucleotide sugars that are required for ER- and GA-linked glycosylation, respectively. To this end, we further show that the galactose in UDP-galactose is incorporated into mature, de novo glycans. Our results also allude to the potential of monosaccharide therapy for several other CDG.


Subject(s)
Congenital Disorders of Glycosylation/metabolism , Fibroblasts/metabolism , Galactose/administration & dosage , Phosphoglucomutase/deficiency , Uridine Diphosphate Galactose/metabolism , Uridine Diphosphate Glucose/metabolism , Cells, Cultured , Cohort Studies , Congenital Disorders of Glycosylation/drug therapy , Congenital Disorders of Glycosylation/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Glycosylation , Humans
4.
Peptides ; 109: 33-38, 2018 11.
Article in English | MEDLINE | ID: mdl-30176261

ABSTRACT

We report here on the structure-activity relationship study of a 14 amino acid fragment of the cathelicidin-related antimicrobial peptide (CRAMP), CRAMP20-33 (KKIGQKIKNFFQKL). It showed activity against Escherichia coli and filamentous fungi with IC50 values below 30 µM and 10 µM, respectively. CRAMP20-33 variants with glycine at position 23 substituted by phenylalanine, leucine or tryptophan showed 2- to 4-fold improved activity against E. coli but not against filamentous fungi. Furthermore, the most active single-substituted peptide, CRAMP20-33 G23 W (IC50 = 2.3 µM against E. coli), showed broad-spectrum activity against Candida albicans, Staphylococcus epidermidis and Salmonella Typhimurium. Introduction of additional arginine substitutions in CRAMP20-33 G23 W, more specifically in CRAMP20-33 G23 W N28R or CRAMP20-33 G23 W Q31R, resulted in 3-fold increased activity against S. epidermidis (IC50 = 4 µM and 4.8 µM, respectively) as compared to CRAMP20-33 G23 W (IC50 = 15.1 µM) but not against the other pathogens tested. In general, double-substituted variants were non-toxic for human HepG2 cells, pointing to their therapeutic potential.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Amino Acid Sequence , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/metabolism , Bacteria/drug effects , Fungi/drug effects , Hep G2 Cells , Humans , Microbial Sensitivity Tests , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Structure-Activity Relationship , Cathelicidins
5.
World J Gastroenterol ; 23(30): 5499-5507, 2017 Aug 14.
Article in English | MEDLINE | ID: mdl-28852309

ABSTRACT

AIM: To develop a MRI-based method for accurate determination of liver volume (LV) and to explore the effect of long-term everolimus (EVR) treatment on LV in PCK rats with hepatomegaly. METHODS: Thirty-one female PCK rats (model for polycystic-liver-disease: PCLD) were randomized into 3 groups and treatment was started at 16 wk, at the moment of extensive hepatomegaly (comparable to what is done in the human disease). Animals received: controls (n = 14), lanreotide (LAN: 3 mg/kg per 2 wk) (n = 10) or everolimus (EVR: 1 mg/kg per day) (n = 7). LV was measured at week 16, 24, 28. At week 28, all rats were sacrificed and liver tissue was harvested. Fibrosis was evaluated using quantitative image analysis. In addition, gene (quantitative RT-PCR) and protein expression (by Western blot) of the PI3K/AkT/mTOR signaling pathway was investigated. RESULTS: LV determination by MRI correlated excellent with the ex vivo measurements (r = 0.99, P < 0.001). The relative changes in LV at the end of treatment were: (controls) +31.8%; (LAN) +5.1% and (EVR) +8.8%, indicating a significantly halt of LV progression compared with controls (respectively, P = 0.01 and P = 0.04). Furthermore, EVR significantly reduced the amount of liver fibrosis (P = 0.004) thus might also prevent the development of portal hypertension. There was no difference in phosphorylation of Akt (Threonine 308) between LAN-treated PCK rats control PCK rats, whereas S6 was significantly more phosphorylated in the LAN group. Phosphorylation of Akt was not different between controls and EVR treated rats, however, for S6 there was significantly less phosphorylation in the EVR treated rats. Thus, both drugs interact with the PI3K/AkT/mTOR signaling cascade but acting at different molecular levels. CONCLUSION: Everolimus halts cyst growth comparable to lanreotide and reduces the development of fibrosis. mTOR-inhibition should be further explored in PCLD patients especially those that need immunosuppression.


Subject(s)
Cysts/drug therapy , Everolimus/therapeutic use , Liver Cirrhosis/drug therapy , Liver Diseases/drug therapy , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cysts/diagnostic imaging , Cysts/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Gene Expression Profiling , Humans , Hypertension, Portal/prevention & control , Liver/diagnostic imaging , Liver/drug effects , Liver/pathology , Liver Cirrhosis/diagnostic imaging , Liver Cirrhosis/pathology , Liver Diseases/diagnostic imaging , Liver Diseases/pathology , Magnetic Resonance Imaging , Peptides, Cyclic/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Rats , Real-Time Polymerase Chain Reaction , Ribosomal Protein S6/metabolism , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , TOR Serine-Threonine Kinases/metabolism
6.
Lipids Health Dis ; 16(1): 46, 2017 Feb 23.
Article in English | MEDLINE | ID: mdl-28231800

ABSTRACT

BACKGROUND: Dietary intervention is the cornerstone of non-alcoholic steatohepatitis (NASH) treatment. However, histological evidence of its efficacy is limited and its impact on hepatic pathways involved in NASH is underreported. The efficacy of the angiotensin receptor type 1 blocker losartan is controversial because of varying results in a few animal and human studies. We evaluated the effect of dietary intervention versus losartan on NASH and associated systemic metabolic features in a representative mouse model. METHODS: Male C57BL/6 J mice with high fat-high sucrose diet (HF-HSD) induced NASH, obesity, insulin resistance and hypercholesterolemia were subjected to dietary intervention (switch from HF-HSD to normal chow diet (NCD)) (n = 9), continuation HF-HSD together with losartan (30 mg/kg/day) (n = 9) or continuation HF-HSD only (n = 9) for 8 weeks. 9 mice received NCD during the entire experiment (20 weeks). We assessed the systemic metabolic effects and performed a detailed hepatic histological and molecular profiling. A P-value of < 0.05, using the group with continuation of HF-HSD only as control, was considered as statistically significant. RESULTS: Dietary intervention normalized obesity, insulin resistance, and hypercholesterolemia (for all P < 0.001), and remarkably, completely reversed all histological features of pre-existent NASH (for all P < 0.001), including fibrosis measured by quantification of collagen proportional area (P < 0.01). At the hepatic molecular level, dietary intervention targeted fibrogenesis with a normalization of collagen type I alpha 1, transforming growth factor ß1, tissue inhibitor of metalloproteinase 1 mRNA levels (for all P < 0.01), lipid metabolism with a normalization of fatty acid translocase/CD36, fatty acid transport protein 5, fatty acid synthase mRNA levels (P < 0.05) and markers related to mitochondrial function with a normalization of hepatic ATP content (P < 0.05) together with sirtuin1 and uncoupling protein 2 mRNA levels (for both P < 0.001). Dietary intervention abolished p62 accumulation (P < 0.01), suggesting a restoration of autophagic flux. Losartan did not significantly affect obesity, insulin resistance, hypercholesterolemia or any histological NASH feature. CONCLUSIONS: Dietary intervention, and not losartan, completely restores the metabolic phenotype in a representative mouse model with pre-existent NASH, obesity, insulin resistance and hypercholesterolemia.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Losartan/pharmacology , Non-alcoholic Fatty Liver Disease/diet therapy , Obesity/diet therapy , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , Collagen Type I, alpha 1 Chain , Drug Evaluation, Preclinical , Gene Expression , Insulin Resistance , Male , Mice, Inbred C57BL , Obesity/drug therapy , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
7.
Transl Oncol ; 10(1): 59-69, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27987431

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is linked to tumor invasion, drug resistance and aggressive disease and this is largely dependent on the cell's microenvironment. Acriflavine (ACF) is an old antibacterial drug recently also suggested as anticancer agent and HIF inhibitor. We wanted to study the effect of acriflavine on EMT in different human cancer models. Pancreatic cancer cells (Panc-1) were exposed to TGF-ß1 or cobalt chloride (to mimick severe hypoxia) to induce EMT. For our third model we exposed HepG2 liver cancer cells to sorafenib which resulted in development of acquired drug resistance with strong features of EMT and aggressive behavior. These models were morphologically and functionally (invasion assay) characterized. Markers of EMT were determined using qRT-PCR and Western blotting. Transcriptome analysis was performed following gene expression determination and combining the iRegulon tool and Gene Set Enrichment Analysis (GSEA). We made the following observations: (1) acriflavine inhibited EMT based on changes in cell morphology, invasive capacities and markers of EMT (at protein and gene expression level). (2) Transcriptome analysis revealed potent inhibition of ATF4 target genes and of the unfolded protein response. We showed that acriflavine blocked eIF2a phosphorylation and reduced ATF4 translation thereby inhibiting the PERK/eIF2a/ATF4 UPR pathway. (3) ACF restored drug sensitivity of cells that obtained acquired resistance. CONCLUSIONS: We identified acriflavine as a potent inhibitor of EMT and the UPR, thereby re-sensitizing the cancer cells to antineoplastic drugs.

8.
PLoS One ; 11(12): e0166048, 2016.
Article in English | MEDLINE | ID: mdl-27992443

ABSTRACT

Intestinal dysbiosis and elevated lipopolysaccharides (LPS) levels have been implicated in the development of obesity, insulin resistance and non-alcoholic steatohepatitis (NASH). In order to determine if LPS levels are elevated in patients with NASH compared to patients with non-alcoholic fatty liver (NAFL) and, if elevated LPS levels correlated with histological severity of non-alcoholic fatty liver disease (NAFLD) we compared LPS, markers of LPS bioactivity and pro-inflammatory cytokines/chemokines in patients undergoing bariatric surgery. At the time of surgery a liver biopsy was taken allowing the stratification into well-delineated subgroups including: No NAFL/NAFL; NASH; NASH with fibrosis and NASH cirrhotics, using the NAFLD Activity Score (NAS). Anthropometric data and plasma were collected for assessment of LPS, lipopolysaccharide binding protein (LBP), soluble CD14 (sCD14), intestinal-type fatty acid binding protein (iFABP), Toll-like receptors 2 and 4 (TLR2, 4) and a panel of cytokines/chemokines. Similar analysis was performed on plasma from a cohort of healthy controls. Our data indicate elevated levels of LPS, LBP, sCD14, iFABP and TLR2,4 in obese patients compared to healthy controls, however, these parameters remained unaltered within patients with limited liver disease (NAFL) compared to NASH/NASH with fibrosis subgroups. Hierarchic cluster analysis using endotoxin-related parameters failed to discriminate between lean controls, NAFLD. While similar cluster analysis implementing inflammation-related parameters clearly distinguished lean controls, NALFD subgroups and NASH cirrhotics. In addition, LPS levels was not associated with disease severity while TNFα, IL8, and CCL3 featured a clear correlation with transaminase levels and the histological severity of NALFD. In conclusion our data indicate a stronger correlation for circulating inflammatory- rather than endotoxin-related parameters in progression of NAFLD and highlights the need for additional larger studies in unravelling further mechanistic insights.


Subject(s)
Carrier Proteins/blood , Chemokines/blood , Cytokines/blood , Lipopolysaccharides/blood , Membrane Glycoproteins/blood , Non-alcoholic Fatty Liver Disease/pathology , Obesity/surgery , Acute-Phase Proteins , Adult , Bariatric Surgery , Cluster Analysis , Endotoxins/metabolism , Female , Humans , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/immunology , Obesity/metabolism , Prospective Studies
9.
Sci Rep ; 6: 33453, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27634375

ABSTRACT

Hepatic inflammation drives hepatic stellate cells (HSC), resulting in liver fibrosis. The Farnesoid-X receptor (FXR) antagonizes inflammation through NF-κB inhibition. We investigated preventive and therapeutic effects of FXR agonist obeticholic acid (OCA) on hepatic inflammation and fibrosis in toxic cirrhotic rats. Cirrhosis was induced by thioacetamide (TAA) intoxication. OCA was given during or after intoxication with vehicle-treated rats as controls. At sacrifice, fibrosis, hemodynamic and biochemical parameters were assessed. HSC activation, cell turn-over, hepatic NF-κB activation, pro-inflammatory and pro-fibrotic cytokines were determined. The effect of OCA was further evaluated in isolated HSC, Kupffer cells, hepatocytes and liver sinusoidal endothelial cells (LSEC). OCA decreased hepatic inflammation and fibrogenesis during TAA-administration and reversed fibrosis in established cirrhosis. Portal pressure decreased through reduced intrahepatic vascular resistance. This was paralleled by decreased expression of pro-fibrotic cytokines (transforming growth-factor ß, connective tissue growth factor, platelet-derived growth factor ß-receptor) as well as markers of hepatic cell turn-over, by blunting effects of pro-inflammatory cytokines (e.g. monocyte chemo-attractant protein-1). In vitro, OCA inhibited both LSEC and Kupffer cell activation; while HSC remained unaffected. This related to NF-κB inhibition via up-regulated IκBα. In conclusion, OCA inhibits hepatic inflammation in toxic cirrhotic rats resulting in decreased HSC activation and fibrosis.


Subject(s)
Chenodeoxycholic Acid/analogs & derivatives , Inflammation/drug therapy , Liver Cirrhosis/drug therapy , Liver/pathology , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Chenodeoxycholic Acid/pharmacology , Chenodeoxycholic Acid/therapeutic use , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hemodynamics/drug effects , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Inflammation/complications , Inflammation/pathology , Inflammation/physiopathology , Inflammation Mediators/metabolism , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Kupffer Cells/pathology , Lipopolysaccharides/pharmacology , Liver Cirrhosis/complications , Liver Cirrhosis/pathology , Liver Cirrhosis/physiopathology , Male , Mice , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Portal Pressure/drug effects , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/metabolism , Thioacetamide , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Vascular Resistance/drug effects
10.
Transl Oncol ; 9(2): 139-146, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27084430

ABSTRACT

OBJECTIVES: Recurrence of hepatocellular carcinoma can arise from the primary tumor ("early recurrence") or de novo from tumor formation in a cirrhotic environment ("late recurrence"). We aimed to develop one simple gene expression score applicable in both the tumor and the surrounding liver that can predict the recurrence risk. METHODS: We determined differentially expressed genes in a cell model of cancer aggressiveness. These genes were first validated in three large published data sets of hepatocellular carcinoma from which we developed a seven-gene risk score. RESULTS: The gene score was applied on two independent large patient cohorts. In the first cohort, with only tumor data available, it could predict the recurrence risk at 3 years after resection (68 ± 10% vs 35 ± 7%, P = .03). In the second cohort, when applied on the tumor, this gene score predicted early recurrence (62 ± 5% vs 37 ± 4%, P < .001), and when applied on the surrounding liver tissue, the same genes also correlated with late recurrence. Four patient classes with each different time patterns and rates of recurrence could be identified based on combining tumor and liver scores. In a multivariate Cox regression analysis, our gene score remained significantly associated with recurrence, independent from other important cofactors such as disease stage (P = .007). CONCLUSIONS: We developed a Global Risk Score that is able to simultaneously predict the risk of early recurrence when applied on the tumor itself, as well as the risk of late recurrence when applied on the surrounding liver tissue.

11.
Gastroenterology ; 149(3): 635-48.e14, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26028579

ABSTRACT

BACKGROUND & AIMS: The prevalence of nonalcoholic fatty liver disease (NAFLD) has increased with the obesity pandemic. We analyzed the transcriptional profiles of subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT), and phenotypes and functional characteristics of adipocyte tissue macrophages (ATMs), in obese patients undergoing bariatric surgery. METHODS: We collected anthropometric data; plasma samples; and SAT, VAT, and liver tissues from 113 obese patients undergoing bariatric surgery at academic hospitals in Europe (Antwerp and Leuven) and South Africa. Based on clinical and histologic features, patients were assigned to the following groups: obese, NAFLD, nonalcoholic steatohepatitis (NASH), or NASH with fibrosis. Microarray analyses were performed to identify genes expressed differentially among groups. We measured levels of cytokines and chemokines in plasma samples and levels of RNAs in adipose tissues by quantitative reverse-transcription polymerase chain reaction. ATMs were isolated from patients and 13 lean individuals undergoing cholecystectomy (controls), analyzed by flow cytometry, and cultured; immunophenotypes and levels of cytokines and chemokines in supernatants were determined. RESULTS: We observed increased expression of genes that regulate inflammation in adipose tissues from patients with NAFLD and NASH; expression of these genes increased as disease progressed from NAFLD to NASH. We found 111 genes associated with inflammation that were expressed differentially between VAT and SAT. Serum levels of interleukin 8, chemokine (C-C motif) ligand 3, and tumor necrosis factor-α correlated with liver inflammation and NAFLD activity score. We developed 2 models that could be used to determine patients' liver histology based on gene expression in VAT and SAT. Flow cytometry showed increased proportions of CD11c+CD206+ and CCR2+ macrophages in VAT from patients with NASH, and supernatants of cultured macrophages had increased levels of cytokines and chemokines compared with controls. CONCLUSIONS: VAT and SAT from patients with NAFLD and NASH have an increased expression of genes that regulate inflammation, and ATM produce increased levels of inflammatory cytokines, compared with adipose tissues from controls. We identified an expression profile of 5 genes in SAT that accurately predict liver histology in these patients. Transcript profiling: accession numbers: GSE58979 and GSE59045.


Subject(s)
Cytokines/immunology , Inflammation Mediators/immunology , Intra-Abdominal Fat/immunology , Macrophages/immunology , Non-alcoholic Fatty Liver Disease/immunology , Obesity/complications , Panniculitis/immunology , Subcutaneous Fat/immunology , Adult , Bariatric Surgery , Belgium , Biomarkers/blood , Biopsy , Cells, Cultured , Cytokines/blood , Cytokines/genetics , Disease Progression , Female , Gene Expression Profiling/methods , Gene Regulatory Networks , Humans , Immunophenotyping/methods , Inflammation Mediators/blood , Intra-Abdominal Fat/metabolism , Liver Cirrhosis/immunology , Macrophages/metabolism , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/diagnosis , Obesity/diagnosis , Obesity/surgery , Oligonucleotide Array Sequence Analysis , Panniculitis/blood , Panniculitis/diagnosis , Phenotype , Predictive Value of Tests , Severity of Illness Index , South Africa , Subcutaneous Fat/metabolism
12.
Am J Pathol ; 185(2): 409-19, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25592258

ABSTRACT

Bacterial translocation (BTL) drives pathogenesis and complications of cirrhosis. Farnesoid X-activated receptor (FXR) is a key transcription regulator in hepatic and intestinal bile metabolism. We studied potential intestinal FXR dysfunction in a rat model of cholestatic liver injury and evaluated effects of obeticholic acid (INT-747), an FXR agonist, on gut permeability, inflammation, and BTL. Rats were gavaged with INT-747 or vehicle during 10 days after bile-duct ligation and then were assessed for changes in gut permeability, BTL, and tight-junction protein expression, immune cell recruitment, and cytokine expression in ileum, mesenteric lymph nodes, and spleen. Auxiliary in vitro BTL-mimicking experiments were performed with Transwell supports. Vehicle-treated bile duct-ligated rats exhibited decreased FXR pathway expression in both jejunum and ileum, in association with increased gut permeability through increased claudin-2 expression and related to local and systemic recruitment of natural killer cells resulting in increased interferon-γ expression and BTL. After INT-747 treatment, natural killer cells and interferon-γ expression markedly decreased, in association with normalized permeability selectively in ileum (up-regulated claudin-1 and occludin) and a significant reduction in BTL. In vitro, interferon-γ induced increased Escherichia coli translocation, which remained unaffected by INT-747. In experimental cholestasis, FXR agonism improved ileal barrier function by attenuating intestinal inflammation, leading to reduced BTL and thus demonstrating a crucial protective role for FXR in the gut-liver axis.


Subject(s)
Bacterial Translocation/drug effects , Chenodeoxycholic Acid/analogs & derivatives , Cholestasis/microbiology , Escherichia coli/physiology , Ileum/microbiology , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Chenodeoxycholic Acid/pharmacology , Cholestasis/metabolism , Cholestasis/pathology , Cytokines/biosynthesis , Gene Expression Regulation/drug effects , Ileum/metabolism , Ileum/pathology , Male , Rats , Rats, Wistar
13.
Gut ; 64(4): 673-83, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24917551

ABSTRACT

OBJECTIVE: No therapy for non-alcoholic steatohepatitis (NASH) has been approved so far. Roux-en-y gastric bypass (RYGB) is emerging as a therapeutic option, although its effect on NASH and related hepatic molecular pathways is unclear from human studies. We studied the effect of RYGB on pre-existent NASH and hepatic mitochondrial dysfunction-a key player in NASH pathogenesis-in a novel diet-induced mouse model nicely mimicking human disease. DESIGN: C57BL/6J mice were fed a high-fat high-sucrose diet (HF-HSD). RESULTS: HF-HSD led to early obesity, insulin resistance and hypercholesterolaemia. HF-HSD consistently induced NASH (steatosis, hepatocyte ballooning and inflammation) with fibrosis already after 12-week feeding. NASH was accompanied by hepatic mitochondrial dysfunction, characterised by decreased mitochondrial respiratory chain (MRC) complex I and IV activity, ATP depletion, ultrastructural abnormalities, together with higher 4-hydroxynonenal (HNE) levels, increased uncoupling protein 2 (UCP2) and tumour necrosis factor-α (TNF-α) mRNA and free cholesterol accumulation. In our model of NASH and acquired mitochondrial dysfunction, RYGB induced sustained weight loss, improved insulin resistance and inhibited progression of NASH, with a marked reversal of fibrosis. In parallel, RYGB preserved hepatic MRC complex I activity, restored ATP levels, limited HNE production and decreased TNF-α mRNA. CONCLUSIONS: Progression of NASH and NASH-related hepatic mitochondrial dysfunction can be prevented by RYGB. RYGB preserves respiratory chain complex activity, thereby restoring energy output, probably by limiting the amount of oxidative stress and TNF-α. These data suggest that modulation of hepatic mitochondrial function contributes to the favourable effect of RYBG on established NASH.


Subject(s)
Gastric Bypass , Mitochondria, Liver/physiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/surgery , Animals , Humans , Insulin Resistance , Liver Diseases , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/physiopathology , Tumor Necrosis Factor-alpha
14.
Molecules ; 19(9): 15088-102, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25244288

ABSTRACT

We investigated the effect of the Arabidopsis thaliana-derived decapeptide OSIP108 on human cell tolerance to the chemotherapeutic agent cisplatin (Cp), which induces apoptosis and mitochondrial dysfunction. We found that OSIP108 increases the tolerance of HepG2 cells to Cp and prevents Cp-induced changes in basic cellular metabolism. More specifically, we demonstrate that OSIP108 reduces Cp-induced inhibition of respiration, decreases glycolysis and prevents Cp-uptake in HepG2 cells. Apart from its protective action against Cp in human cells, OSIP108 also increases the yeast Saccharomyces cerevisiae tolerance to Cp. A limited yeast-based study of OSIP108 analogs showed that cyclization does not severely affect its activity, which was further confirmed in HepG2 cells. Furthermore, the similarity in the activity of the D-stereoisomer (mirror image) form of OSIP108 with the L-stereoisomer suggests that its mode of action does not involve binding to a stereospecific receptor. In addition, as OSIP108 decreases Cp uptake in HepG2 cells and the anti-Cp activity of OSIP108 analogs without free cysteine is reduced, OSIP108 seems to protect against Cp-induced toxicity only partly via complexation. Taken together, our data indicate that OSIP108 and its cyclic derivatives can protect against Cp-induced toxicity and, thus, show potential as treatment options for mitochondrial dysfunction- and apoptosis-related conditions.


Subject(s)
Antineoplastic Agents/toxicity , Arabidopsis Proteins/pharmacology , Arabidopsis/physiology , Cisplatin/toxicity , Mitochondria/drug effects , Hep G2 Cells , Humans , Mitochondria/physiology
15.
Hepatology ; 59(6): 2286-98, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24259407

ABSTRACT

UNLABELLED: The farnesoid X receptor (FXR) is a nuclear bile acid receptor involved in bile acid homeostasis, hepatic and intestinal inflammation, liver fibrosis, and cardiovascular disease. We studied the effect of short-term treatment with obeticholic acid (INT-747), a potent selective FXR agonist, on intrahepatic hemodynamic dysfunction and signaling pathways in different rat models of cirrhotic portal hypertension (PHT). For this, thioacetamide (TAA)-intoxicated and bile-duct-ligated (BDL) rats were used as models. After gavage of two doses of 30 mg/kg of INT-747 or vehicle within 24 hours, in vivo hemodynamics were assessed. Additionally, we evaluated the direct effect of INT-747 on total intrahepatic vascular resistance (IHVR) and intrahepatic vascular tone (endothelial dysfunction and hyperresponsiveness to methoxamine) by means of an in situ liver perfusion system and on hepatic stellate cell contraction in vitro. FXR expression and involved intrahepatic vasoactive pathways (e.g., endothelial nitric oxide synthase [eNOS], Rho-kinase, and dimethylarginine dimethylaminohydrolase [DDAH]) were analyzed by immunohistochemistry, reverse-transcriptase polymerase chain reaction, or western blotting. In both cirrhotic models, FXR expression was decreased. Treatment with INT-747 in TAA and BDL reactivated the FXR downstream signaling pathway and decreased portal pressure by lowering total IHVR without deleterious systemic hypotension. In the perfused TAA and BDL cirrhotic liver, INT-747 improved endothelial vasorelaxation capacity, but not hyperresponsiveness. In both groups, this was associated with an increased eNOS activity, which, in TAA, related to down-regulation of Rho-kinase and in BDL to up-regulation of DDAH-2. CONCLUSION: FXR agonist INT-747 improves PHT in two different rat models of cirrhosis by decreasing IHVR. This hemodynamic effect relates to increased intrahepatic eNOS activity by pathways that differ depending on the etiology of cirrhosis.


Subject(s)
Chenodeoxycholic Acid/analogs & derivatives , Hypertension, Portal/drug therapy , Signal Transduction/physiology , Animals , Chenodeoxycholic Acid/pharmacology , Chenodeoxycholic Acid/therapeutic use , Disease Models, Animal , Down-Regulation , Drug Evaluation, Preclinical , Hypertension, Portal/etiology , Hypertension, Portal/physiopathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/complications , Liver Cirrhosis/physiopathology , Male , Nitric Oxide Synthase Type III/metabolism , Random Allocation , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/agonists
16.
Exp Toxicol Pathol ; 65(7-8): 1025-33, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23578882

ABSTRACT

Heavy metals can accumulate in organisms via various pathways, including respiration, adsorption and ingestion. They are known to generate free radicals and induce oxidative and/or nitrosative stress with depletion of anti-oxidants. Tuna by-product meal (TBM) is rich in proteins and can, therefore, offer an attractive protein source for animals. This study was undertaken to assess the effects of metals present in TBM, namely cadmium (Cd), lead (Pb), and mercury (Hg), separately or in combination with oxidative stress, on cell viability. Three cell models: rat liver FTO2B, human hepatoma HepG2, and human hepatic WRL-68, were used. Cell viability was determined following exposure to various concentrations of the metals. Two antioxidant genes, catalase (CAT) and superoxide dismutase (SOD), were measured to obtain a better understanding of oxidative stress-associated gene expression. Among the metals present in TBM, only Cd at a concentration of 30µM was noted to exhibit cytotoxic effects. This cytotoxicity was even more pronounced after co-stimulation with H2O2, used to mimic systemic oxidative stress. At non-toxic concentrations, Hg and Pb were noted to aggravate oxidative stress toxicity. The results further revealed that exposure to Cd, Pb, and a co-stimulation of H2O2 with Hg resulted in the increased expression of antioxidant gene SOD. A risk assessment of toxic contaminants in TBM indicated that food safety objectives should consider the human health impacts of foods derived from animals fed on contaminated meal and that much care should be taken when TBM is used in animal diet.


Subject(s)
Food Contamination , Heavy Metal Poisoning , Hepatocytes/drug effects , Tuna , Animal Feed/toxicity , Animals , Cadmium/toxicity , Catalase/metabolism , Cell Line , Cell Survival , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/metabolism , Humans , Lead/toxicity , Mercury/toxicity , Metals, Heavy/adverse effects , Metals, Heavy/metabolism , Oxidative Stress/physiology , Poisoning/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism
17.
Cancer Lett ; 329(1): 74-83, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23111106

ABSTRACT

Sorafenib leads to a survival benefit in patients with advanced hepatocellular carcinoma but its use is hampered by the occurrence of drug resistance. To investigate the molecular mechanisms involved we developed five resistant human liver cell lines in which we studied morphology, gene expression and invasive potential. The cells changed their appearance, lost E-cadherin and KRT19 and showed high expression of vimentin, indicating epithelial-to-mesenchymal transition. Resistant cells showed reduced adherent growth, became more invasive and lost liver-specific gene expression. Furthermore, following withdrawal of sorafenib, the resistant cells showed rebound growth, a phenomenon also found in patients. This cell model was further used to investigate strategies for restoration of sensitivity to sorafenib.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition/drug effects , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Anilides/pharmacology , Cadherins/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Chromones/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Keratin-19/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Morpholines/pharmacology , Niacinamide/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/pharmacology , Sorafenib , Vimentin/metabolism
18.
Cancer Lett ; 315(2): 178-88, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22088439

ABSTRACT

We demonstrated in an in vitro model (human HepG2 liver cells) that chronic hypoxia induced gene expression is associated with an aggressive phenotype in patients with hepatocellular carcinoma (HCC). The aim of this study was to characterize this model further using gene expression microarray, real-time PCR and immunocytochemistry. Subsequently, pathway analysis software was used to identify relevant processes. After examination, we selected 2% O2 during 72 h as conditions to study chronic hypoxia. The most affected signaling is centered on TGF-ß1 and PPARα/RXRα. Cells at 2% O2 showed a shift in expression of Epithelial-to-Mesenchymal-Transition (EMT) related genes. Furthermore, a downregulation of liver specific detoxification pathways including cytochrome P450's and glutathione-S-transferases was observed. Both up- and downregulation events within different signaling cascades indicated a cellular adaptation and the onset of a new equilibrium. The prominent role of TGF-ß1- and PPARα/RXRα signaling and cell motility pathways warrants their further investigation for therapeutic targets in HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Culture Techniques , Hypoxia , Liver Neoplasms/pathology , Models, Biological , Gene Expression Regulation , Humans , Immunohistochemistry , Microarray Analysis , Real-Time Polymerase Chain Reaction , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL