Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt B): 1308-1318, 2018 04.
Article in English | MEDLINE | ID: mdl-28965883

ABSTRACT

The nuclear receptor farnesoid X receptor (FXR) is the master regulator of bile acids (BAs) homeostasis since it transcriptionally drives modulation of BA synthesis, influx, efflux, and detoxification along the enterohepatic axis. Due to its crucial role, FXR alterations are involved in the progression of a plethora of BAs associated inflammatory disorders in the liver and in the gut. The involvement of the FXR pathway in cholestasis development and management has been elucidated so far with a direct role of FXR activating therapy in this condition. However, the recent identification of a new type of genetic progressive familial intrahepatic cholestasis (PFIC) linked to FXR mutations has strengthen also the bona fide beneficial effects of target therapies that by-pass FXR activation, directly promoting the action of its target, namely the enterokine FGF19, in the repression of hepatic BAs synthesis with reduction of total BA levels in the liver and serum, accomplishing one of the major goals in cholestasis. This article is part of a Special Issue entitled: Cholangiocytes in Health and Diseaseedited by Jesus Banales, Marco Marzioni and Peter Jansen.


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis, Intrahepatic/genetics , Epithelial Cells/metabolism , Fibroblast Growth Factors/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Bile Acids and Salts/blood , Cholagogues and Choleretics/pharmacology , Cholagogues and Choleretics/therapeutic use , Cholestasis, Intrahepatic/blood , Cholestasis, Intrahepatic/drug therapy , Cholestasis, Intrahepatic/pathology , Clinical Trials as Topic , Disease Models, Animal , Epithelial Cells/drug effects , Humans , Intestinal Mucosa/metabolism , Liver/metabolism , Mice , Mice, Transgenic , Mutation , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
2.
Hepatology ; 65(6): 2031-2044, 2017 06.
Article in English | MEDLINE | ID: mdl-28142199

ABSTRACT

In cirrhosis, increased intrahepatic vascular resistance (IHVR) is the primary factor for portal hypertension (PH) development. Hepatic stellate cells (HSCs) play a major role increasing IHVR because, when activated, they are contractile and promote fibrogenesis. Protease-activated receptors (PARs) can activate HSCs through thrombin and factor Xa, which are known PAR agonists, and cause microthrombosis in liver microcirculation. This study investigates the effects of the oral anticoagulant, rivaroxaban (RVXB), a direct antifactor Xa, on HSC phenotype, liver fibrosis (LF), liver microthrombosis, and PH in cirrhotic rats. Hepatic and systemic hemodynamic, nitric oxide (NO) bioavailability, LF, HSC activation, and microthrombosis were evaluated in CCl4 and thioacetamide-cirrhotic rats treated with RVXB (20 mg/kg/day) or its vehicle for 2 weeks. RVXB significantly decreased portal pressure (PP) in both models of cirrhosis without changes in portal blood flow, suggesting a reduction in IHVR. RVXB reduced oxidative stress, improved NO bioavailability, and ameliorated endothelial dysfunction. Rivaroxaban deactivated HSC, with decreased alpha-smooth muscle actin and mRNA expression of other HSC activation markers. Despite this marked improvement in HSC phenotype, no significant changes in LF were identified. RVXB markedly reduced fibrin deposition, suggesting reduced intrahepatic microthrombosis. CONCLUSION: RVXB decreases PP in two rat models of cirrhosis. This effect is mostly associated with decreased IHVR, enhanced NO bioavailability, HSC deactivation, and reduced intrahepatic microthrombosis. Our findings suggest that RVXB deserves further evaluation as a potential treatment for cirrhotic PH. (Hepatology 2017;65:2031-2044).


Subject(s)
Anticoagulants/pharmacology , Hypertension, Portal/drug therapy , Liver Cirrhosis/drug therapy , Rivaroxaban/pharmacology , Vascular Resistance/drug effects , Administration, Oral , Animals , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/drug effects , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , Hypertension, Portal/etiology , Liver Cirrhosis/complications , Liver Cirrhosis/pathology , Male , Oxidative Stress/drug effects , Portal Pressure/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reference Values , Statistics, Nonparametric , Treatment Outcome
3.
J Hepatol ; 64(4): 834-42, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26686269

ABSTRACT

BACKGROUND & AIMS: Increased hepatic vascular resistance due to fibrosis and elevated hepatic vascular tone is the primary factor in the development of portal hypertension. Heparin may decrease fibrosis by inhibiting intrahepatic microthrombosis and thrombin-mediated hepatic stellate cell activation. In addition, heparin enhances eNOS activity, which may reduce hepatic vascular tone. Our study aimed at evaluating the effects of acute, short-, long-term and preventive enoxaparin administration on hepatic and systemic hemodynamics, liver fibrosis and nitric oxide availability in cirrhotic rats. METHODS: Enoxaparin (1.8 mg/kg subcutaneously), or its vehicle, was administered to CCl4-cirrhotic rats 24h and 1h before the study (acute), daily for 1 week (short-term) or daily for 3 weeks (long-term) and to thioacetamide-cirrhotic rats daily for 3 weeks with/without thioacetamide (preventive/long-term, respectively). Mean arterial pressure, portal pressure, portal blood flow, hepatic vascular resistance and molecular/cellular mechanisms were evaluated. RESULTS: No significant changes in hemodynamic parameters were observed in acute administration. However, one-week, three-week and preventive treatments significantly decreased portal pressure mainly due to a decrease in hepatic vascular resistance without significant changes in mean arterial pressure. These findings were associated with significant reductions in liver fibrosis, hepatic stellate cell activation, and desmin expression. Moreover, a reduction in fibrin deposition was observed in enoxaparin-treated rats, suggesting reduced intrahepatic microthrombosis. CONCLUSION: Enoxaparin reduces portal pressure in cirrhotic rats by improving the structural component of increased liver resistance. These findings describe the potentially beneficial effects of enoxaparin beyond the treatment/prevention of portal vein thrombosis in cirrhosis, which deserve further investigation.


Subject(s)
Enoxaparin/pharmacology , Liver Cirrhosis, Experimental/drug therapy , Portal Pressure/drug effects , Vascular Resistance/drug effects , Animals , Liver Cirrhosis, Experimental/physiopathology , Male , Rats , Rats, Wistar
4.
Hepatology ; 62(1): 166-78, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25808184

ABSTRACT

UNLABELLED: Matrix metalloproteinases (MMPs) participate in tissue repair after acute injury, but also participate in cancer by promoting a protumorigenic microenvironment. Previously, we reported on a key role for MMP10 in mouse liver regeneration. Herein, we investigated MMP10 expression and function in human hepatocellular carcinoma (HCC) and diethylnitrosamine (DEN)-induced mouse hepatocarcinogenesis. MMP10 was induced in human and murine HCC tissues and cells. MMP10-deficient mice showed less HCC incidence, smaller histological lesions, reduced tumor vascularization, and less lung metastases. Importantly, expression of the protumorigenic, C-X-C chemokine receptor-4 (CXCR4), was reduced in DEN-induced MMP10-deficient mice livers. Human HCC cells stably expressing MMP10 had increased CXCR4 expression and migratory capacity. Pharmacological inhibition of CXCR4 significantly reduced MMP10-stimulated HCC cell migration. Furthermore, MMP10 expression in HCC cells was induced by hypoxia and the CXCR4 ligand, stromal-derived factor-1 (SDF1), through the extracellular signal-regulated kinase 1/2 pathway, involving an activator protein 1 site in MMP10 gene promoter. CONCLUSION: MMP10 contributes to HCC development, participating in tumor angiogenesis, growth, and dissemination. We identified a new reciprocal crosstalk between MMP10 and the CXCR4/SDF1 axis contributing to HCC progression and metastasis. To our knowledge, this is the first report addressing the role of a MMP in hepatocarcinogenesis in the corresponding genetic mouse model.


Subject(s)
Chemokine CXCL12/metabolism , Liver Neoplasms, Experimental/etiology , Matrix Metalloproteinase 10/metabolism , Receptors, CXCR4/metabolism , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Hypoxia/metabolism , Liver Neoplasms, Experimental/enzymology , Male , Mice, Inbred C57BL , Receptor Cross-Talk
5.
Int J Cancer ; 136(10): 2469-75, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25346390

ABSTRACT

Fibroblast growth factor 15 (FGF15), FGF19 in humans, is a gut-derived hormone and a key regulator of bile acids and carbohydrate metabolism. FGF15 also participates in liver regeneration after partial hepatectomy inducing hepatocellular proliferation. FGF19 is overexpressed in a significant proportion of human hepatocellular carcinomas (HCC), and activation of its receptor FGFR4 promotes HCC cell growth. Here we addressed for the first time the role of endogenous Fgf15 in hepatocarcinogenesis. Fgf15(+/+) and Fgf15(-/-) mice were subjected to a clinically relevant model of liver inflammation and fibrosis-associated carcinogenesis. Fgf15(-/-) mice showed less and smaller tumors, and histological neoplastic lesions were also smaller than in Fgf15(+/+) animals. Importantly, ileal Fgf15 mRNA expression was enhanced in mice undergoing carcinogenesis, but at variance with human HCC it was not detected in liver or HCC tissues, while circulating FGF15 protein was clearly upregulated. Hepatocellular proliferation was also reduced in Fgf15(-/-) mice, which also expressed lower levels of the HCC marker alpha-fetoprotein (AFP). Interestingly, lack of FGF15 resulted in attenuated fibrogenesis. However, in vitro experiments showed that liver fibrogenic stellate cells were not direct targets for FGF15/FGF19. Conversely we demonstrate that FGF15/FGF19 induces the expression of the pro-fibrogenic and pro-tumorigenic connective tissue growth factor (CTGF) in hepatocytes. These findings suggest the existence of an FGF15-triggered CTGF-mediated paracrine action on stellate cells, and an amplification mechanism for the hepatocarcinogenic effects of FGF15 via CTGF production. In summary, our observations indicate that ileal FGF15 may contribute to HCC development in a context of chronic liver injury and fibrosis.


Subject(s)
Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Ileum/metabolism , Liver Cirrhosis, Experimental/metabolism , Liver Neoplasms, Experimental/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Fibroblast Growth Factors/blood , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Hep G2 Cells , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/blood , Liver Cirrhosis, Experimental/pathology , Liver Neoplasms, Experimental/pathology , Mice
6.
Liver Int ; 34(7): e257-70, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24119197

ABSTRACT

BACKGROUND & AIMS: Upon tissue injury, the liver mounts a potent reparative and regenerative response. A role for proteases, including serine and matrix metalloproteinases (MMPs), in this process is increasingly recognized. We have evaluated the expression and function of MMP10 (stromelysin-2) in liver wound healing and regeneration. METHODS: The hepatic expression of MMP10 was examined in two murine models: liver regeneration after two-thirds partial hepatectomy (PH) and bile duct ligation (BDL). MMP10 was detected in liver tissues by qPCR, western blotting and immunohistochemistry. The effect of growth factors and toll-like receptor 4 (TLR4) agonists on MMP10 expression was studied in cultured parenchymal and biliary epithelial cells and macrophages respectively. The role of MMP10 was evaluated by comparing the response of Mmp10+/+ and Mmp10-/- mice to PH and BDL. The intrahepatic turnover of the extracellular matrix proteins fibrin (ogen) and fibronectin was examined. RESULTS: MMP10 mRNA was readily induced after PH and BDL. MMP10 protein was detected in hepatocytes, cholangiocytes and macrophages. In cultured liver epithelial cells, MMP10 expression was additively induced by transforming growth factor-ß and epidermal growth factor receptor ligands. TLR4 ligands also stimulated MMP10 expression in macrophages. Lack of MMP10 resulted in increased liver injury upon PH and BDL. Resolution of necrotic areas was impaired, and Mmp10-/- mice showed increased fibrogenesis and defective turnover of fibrin (ogen) and fibronectin. CONCLUSIONS: MMP10 expression is induced during mouse liver injury and participates in the hepatic wound healing response. The profibrinolytic activity of MMP10 may be essential in this novel hepatoprotective role.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Liver Diseases/physiopathology , Liver/physiology , Matrix Metalloproteinase 10/metabolism , Regeneration/physiology , Animals , Bile Ducts/physiopathology , Bile Ducts/surgery , Blotting, Western , Fibrinogen/metabolism , Hepatectomy , Immunohistochemistry , Ligation , Liver Diseases/enzymology , Mice , Mice, Knockout , Polymerase Chain Reaction , Toll-Like Receptor 4/antagonists & inhibitors
7.
Front Endocrinol (Lausanne) ; 14: 1159127, 2023.
Article in English | MEDLINE | ID: mdl-37409233

ABSTRACT

Objective: Increased Fibroblast Growth Factor-21 (FGF-21) circulating levels have been described in obesity. In this observational study, we analysed a group of subjects with metabolic disorders to unravel the putative link between visceral adiposity and FGF-21 serum levels. Methods: Total and intact serum FGF-21 concentration was measured with an ELISA assay respectively in 51 and 46 subjects, comparing FGF-21 levels in dysmetabolic conditions. We also tested Spearman's correlations between FGF-21 serum levels and biochemical and clinical metabolic parameters. Results: FGF-21 was not significantly increased in high-risk conditions such as visceral obesity, Metabolic Syndrome, diabetes, smoking, and atherosclerosis. Waist Circumference (WC), but not BMI, positively correlated with total FGF-21 levels (r=0.31, p <0.05), while HDL-cholesterol (r=-0.29, p <0.05) and 25-OH Vitamin D (r=-0.32, p <0.05) showed a significant negative correlation with total FGF-21. ROC analysis of FGF-21 in prediction of increased WC, showed that patients with total FGF-21 level over cut-off value of 161.47 pg/mL presented with impaired FPG. Conversely, serum levels of the intact form of FGF-21 did not correlate with WC and other metabolic biomarkers. Conclusion: Our newly calculated cut-off for total FGF-21 according to visceral adiposity identified subjects with fasting hyperglycemia. However, waist circumference correlates with total FGF-21 serum levels but does not correlate with intact FGF-21, suggesting that functional FGF-21 does not necessarily relate with obesity and metabolic features.


Subject(s)
Adiposity , Obesity, Abdominal , Humans , Obesity, Abdominal/metabolism , Body Mass Index , Obesity , Fibroblast Growth Factors/metabolism
8.
Hepatology ; 54(6): 2149-58, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21800344

ABSTRACT

UNLABELLED: The identification of molecular mechanisms involved in the maintenance of the transformed phenotype of hepatocellular carcinoma (HCC) cells is essential for the elucidation of therapeutic strategies. Here, we show that human HCC cells display an autocrine loop mediated by connective tissue growth factor (CTGF) that promotes DNA synthesis and cell survival. Expression of CTGF was stimulated by epidermal growth factor receptor (EGFR) ligands and was dependent on the expression of the transcriptional coactivator, Yes-associated protein (YAP). We identified elements in the CTGF gene proximal promoter that bound YAP-enclosing complexes and were responsible for basal and EGFR-stimulated CTGF expression. We also demonstrate that YAP expression can be up-regulated through EGFR activation not only in HCC cells, but also in primary human hepatocytes. CTGF contributed to HCC cell dedifferentiation, expression of inflammation-related genes involved in carcinogenesis, resistance toward doxorubicin, and in vivo HCC cell growth. Importantly, CTGF down-regulated tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor 2 expression and was involved in the reduced sensitivity of these cells toward TRAIL-mediated apoptosis. CONCLUSION: We have identified autocrine CTGF as a novel determinant of HCC cells' neoplastic behavior. Expression of CTGF can be stimulated through the EGFR-signaling system in HCC cells in a novel cross-talk with the oncoprotein YAP. Moreover, to our knowledge, this is the first study that identifies a signaling mechanism triggering YAP gene expression in healthy and transformed liver parenchymal cells.


Subject(s)
Autocrine Communication/physiology , Carcinoma, Hepatocellular/physiopathology , Connective Tissue Growth Factor/physiology , ErbB Receptors/physiology , Liver Neoplasms/physiopathology , Nuclear Proteins/physiology , Transcription Factors/physiology , Cell Cycle Proteins , Connective Tissue Growth Factor/biosynthesis , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Hepatocytes/metabolism , Humans , Nuclear Proteins/biosynthesis , Primary Cell Culture , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Transcription Factors/biosynthesis
9.
Dig Dis ; 30(5): 524-31, 2012.
Article in English | MEDLINE | ID: mdl-23108309

ABSTRACT

BACKGROUND/AIMS: Hepatocellular carcinoma (HCC) is a chemoresistant tumor strongly associated with chronic hepatitis. Identification of molecular links connecting inflammation with cell growth/survival, and characterization of pro-tumorigenic intracellular pathways is therefore of therapeutic interest. The epidermal growth factor receptor (EGFR) signaling system stands at a crossroad between inflammatory signals and intracellular pathways associated with hepatocarcinogenesis. We investigated the regulation and activity of different components of the EGFR system, including the EGFR ligand amphiregulin (AR) and its sheddase ADAM17, and the modulation of intracellular EGFR signaling by a novel mechanism involving protein methylation. METHODS: ADAM17 protein expression was examined in models of liver injury and carcinogenesis. Crosstalk between tumor necrosis factor (TNF)-α, AR and EGFR signaling was evaluated in human HCC cells and mouse hepatocytes. Modulation of EGFR signaling and biological responses by methylation reactions was evaluated in AML12 mouse hepatocytes. RESULTS: ADAM17 was upregulated in liver injury and hepatocarcinogenesis. TNF-α triggered AR shedding and EGFR transactivation in HCC cells. AR was necessary for TNF-α activation of ERK1/2 and Akt signaling in hepatocytes. Inhibition of methylation reactions increased the ERK1/2 signal amplitude triggered by AR/EGFR and reduced DNA synthesis in AML12 cells. CONCLUSIONS: Increased ADAM17 in pre-neoplastic liver injury further supports its implication in hepatocarcinogenesis. AR release and EGFR transactivation by TNF-α constitutes a novel link between inflammatory signals and pro-tumorigenic mechanisms in liver cells. Finally, the identification of a new mechanism controlling growth factor signaling, and biological responses, involving methylation reactions within the RAS/RAF/MEK/ERK pathway, exposes a new target for antineoplastic intervention.


Subject(s)
Carcinoma, Hepatocellular/metabolism , ErbB Receptors/metabolism , Inflammation/metabolism , Liver Neoplasms/metabolism , Signal Transduction/physiology , ADAM Proteins/metabolism , ADAM17 Protein , Amphiregulin , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , EGF Family of Proteins , Gene Expression Regulation/physiology , Glycoproteins/metabolism , Hepatocytes/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Liver/injuries , Liver/metabolism , MAP Kinase Signaling System/physiology , Methylation , Mice , Proto-Oncogene Proteins c-akt , Tumor Necrosis Factor-alpha/metabolism
10.
Hepatol Commun ; 6(10): 2937-2949, 2022 10.
Article in English | MEDLINE | ID: mdl-35903850

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is defined by a set of hepatic conditions ranging from steatosis to steatohepatitis (NASH), characterized by inflammation and fibrosis, eventually predisposing to hepatocellular carcinoma (HCC). Together with fatty acids (FAs) originated from adipose lipolysis and hepatic lipogenesis, intestinal-derived FAs are major contributors of steatosis. However, the role of mono-unsaturated FAs (MUFAs) in NAFLD development is still debated. We previously established the intestinal capacity to produce MUFAs, but its consequences in hepatic functions are still unknown. Here, we aimed to determine the role of the intestinal MUFA-synthetizing enzyme stearoyl-CoA desaturase 1 (SCD1) in NAFLD. We used intestinal-specific Scd1-KO (iScd1-/- ) mice and studied hepatic dysfunction in different models of steatosis, NASH, and HCC. Intestinal-specific Scd1 deletion decreased hepatic MUFA proportion. Compared with controls, iScd1-/- mice displayed increased hepatic triglyceride accumulation and derangement in cholesterol homeostasis when fed a MUFA-deprived diet. Then, on Western diet feeding, iScd1-/- mice triggered inflammation and fibrosis compared with their wild-type littermates. Finally, intestinal-Scd1 deletion predisposed mice to liver cancer. Conclusions: Collectively, these results highlight the major importance of intestinal MUFA metabolism in maintaining hepatic functions and show that gut-derived MUFAs are protective from NASH and HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Animals , Carcinoma, Hepatocellular/genetics , Cholesterol , Diet, Western , Fatty Acids , Fatty Acids, Monounsaturated/metabolism , Fibrosis , Inflammation , Liver Neoplasms/genetics , Mice , Non-alcoholic Fatty Liver Disease/genetics , Stearoyl-CoA Desaturase/genetics , Triglycerides/metabolism
11.
iScience ; 25(1): 103707, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35036884

ABSTRACT

Compelling evidence support an involvement of oxidative stress and intestinal inflammation as early events in the predisposition and development of obesity and its related comorbidities. Here, we show that deficiency of the major mitochondrial antioxidant enzyme superoxide dismutase 2 (SOD2) in the gastrointestinal tract drives spontaneous obesity. Intestinal epithelium-specific Sod2 ablation in mice induced adiposity and inflammation via phospholipase A2 (PLA2) activation and increased release of omega-6 polyunsaturated fatty acid arachidonic acid. Remarkably, this obese phenotype was rescued when fed an essential fatty acid-deficient diet, which abrogates de novo biosynthesis of arachidonic acid. Data from clinical samples revealed that the negative correlation between intestinal Sod2 mRNA levels and obesity features appears to be conserved between mice and humans. Collectively, our findings suggest a role of intestinal Sod2 levels, PLA2 activity, and arachidonic acid in obesity presenting new potential targets of therapeutic interest in the context of this metabolic disorder.

12.
EBioMedicine ; 54: 102719, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32259714

ABSTRACT

BACKGROUND: Disruption of bile acid (BA) homeostasis plays a key role in intestinal inflammation. The gut-liver axis is the main site for the regulation of BA synthesis and BA pool size via the combined action of the nuclear Farnesoid X Receptor (FXR) and the enterokine Fibroblast Growth Factor 19 (FGF19). Increasing evidence have linked derangement of BA metabolism with dysbiosis and mucosal inflammation. Thus, here we aimed to investigate the potential action of an FGF19 analogue on intestinal microbiota and inflammation. METHODS: A novel engineered non-tumorigenic variant of the FGF19 protein, M52-WO 2016/0168219 was generated. WT and FXRnull mice were injected with AAV-FGF19-M52 or the control AAV-GFP and subjected to Sodium Dextran Sulphate-induced colitis. FINDINGS: FGF19-M52 reduced BA synthesis and pool size, modulated its composition and protected mice from intestinal inflammation. These events were coupled with preservation of the intestinal epithelial barrier integrity, inhibition of inflammatory immune response and modulation of microbiota composition. Interestingly, FGF19-M52-driven systemic and local anti-inflammatory activity was completely abolished in Farnesoid X Receptor (FXR)null mice, thus underscoring the need of FXR to guarantee enterocytes' fitness and complement FGF19 anti-inflammatory activity. To provide a translational perspective, we also show that circulating FGF19 levels are reduced in patients with Crohn's disease. INTERPRETATION: Reactivation of the FXR-FGF19 axis in a murine model of intestinal inflammation could bona fide provide positive changes in BA metabolism with consequent reduction of intestinal inflammation and modulation of microbiota. These results point to the therapeutic potential of FGF19 in the treatment of intestinal inflammation with concomitant derangement of BA homeostasis. FUNDING: A. Moschetta is funded by MIUR-PRIN 2017 <- 2017J3E2W2; Italian Association for Cancer Research (AIRC, IG 23239); Interreg V-A Greece-Italy 2014-2020-SILVER WELLBEING, MIS5003627; HDHL-INTIMIC EuJPI-FATMAL; MIUR PON "R&I" 2014-2020-ARS01_01220. No money has been paid by NGM Biopharmaceuticals or any other agency to write this article.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis, Ulcerative/microbiology , Crohn Disease/microbiology , Fibroblast Growth Factors/metabolism , Gastrointestinal Microbiome , Peptides/therapeutic use , Animals , Bile Acids and Salts/metabolism , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Crohn Disease/drug therapy , Crohn Disease/metabolism , Female , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Peptides/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use
13.
Cell Metab ; 25(4): 763-764, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28380370

ABSTRACT

During conditions of impaired bile flow (cholestasis), increased serum bile acids (BAs) are prognostic markers of sepsis. In this issue, Hao et al. (2017) show that the BA receptor FXR binds NLRP3 inflammasome in macrophages and inhibits activation of inflammasome components, thus reducing endotoxemia in cholestasis.


Subject(s)
Bile Acids and Salts , Inflammasomes , Cholestasis , Endotoxemia , Humans , Receptors, Cytoplasmic and Nuclear
14.
Curr Protoc Mouse Biol ; 7(1): 13-28, 2017 Mar 02.
Article in English | MEDLINE | ID: mdl-28252200

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic multifactorial inflammatory disorder characterized by periods of activation and remission of intestinal inflammation, with potentially severe complications, that can lead to mortality. Experimental animal models of intestinal inflammation are crucial for understanding the pathogenesis of Crohn's disease (CD) and ulcerative colitis (UC), the two major human IBD phenotypes. Animal models have been instrumental in unveiling the molecular background of IBD, and although a single model is not able to capture the complexity of this disease, each of them provided valuable insight into its different aspects. Chemically induced models of intestinal inflammation, mainly dextran sodium sulfate (DSS)- and 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis, are widely used. This article describes DSS- and TNBS-induced colitis models and their relevance to IBD pathophysiology and pre-clinical therapeutic management. © 2017 by John Wiley & Sons, Inc.


Subject(s)
Cell Transformation, Neoplastic/chemically induced , Colitis/chemically induced , Disease Models, Animal , Inflammatory Bowel Diseases/chemically induced , Acute Disease , Animals , Azoxymethane , Chronic Disease , Dextran Sulfate , Humans , Mice , Trinitrobenzenesulfonic Acid
15.
Mol Aspects Med ; 56: 66-74, 2017 08.
Article in English | MEDLINE | ID: mdl-28400119

ABSTRACT

Diet and lifestyle habits have a profound impact on the pathophysiology of many diseases. Colorectal cancer (CRC) is the third most common cancer worldwide and its etiology is strongly influenced by nutrition and high fat/high carbohydrate Western-style diet. Human epidemiological and animal studies have shown that colonic cancer risk is also related to faecal bile acid concentration. Abnormally high levels of bile acids (BA) trigger the colonic mucosa with a plethora of detrimental effects such as DNA oxidative damage, inflammation and hyperproliferation that highly promote CRC progression in post-initiation phase. The Farnesoid X receptor (FXR) is a nuclear receptor that transcriptionally mediates the signalling activity of BAs. FXR regulates BA metabolism mainly maintaining BA concentrations within a physiological range, thereby preventing BA-induced cytotoxicity. In fact, loss of FXR is associated with higher BA concentrations and with a pro-tumorigenic phenotype. Here we explore the liaison connecting nutrition, intestinal epithelium renewal, BA and their nuclear receptor FXR in CRC. Moreover, we summarize evidence linking BA and CRC, as well as examine current understanding of the protumoral actions of BA and the bona fide antitumoral properties of FXR.


Subject(s)
Bile Acids and Salts/metabolism , Colon/metabolism , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Colon/pathology , Colonic Neoplasms/etiology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Constitutive Androstane Receptor , Diet, Western/adverse effects , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Liver/metabolism , Pregnane X Receptor , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Risk Factors , Signal Transduction
17.
Hepat Oncol ; 1(2): 241-252, 2014 Apr.
Article in English | MEDLINE | ID: mdl-30190958

ABSTRACT

Hepatocellular carcinoma (HCC) is a molecularly complex tumor that is resistant to standard and targeted therapies, and thus a deadly disease. In this context, the identification of key alterations driving HCC development is therefore essential. The implementation of next-generation sequencing techniques has underscored earlier realizations of the marked dysregulation of pre-mRNA splicing in HCC. Impairments in alternative splicing may lead to the expression of protumorigenic protein isoforms and to the generation of unstable mRNA species. Mechanistically, mutations in key nucleotides are responsible for many of these alterations in different types of tumors. However, changes in the expression of factors involved in the regulation of the splicing machinery are also important determinants in the derangement of pre-mRNA splicing. Here we discuss recent reports on the alteration of splicing factors in HCC, the pathological significance of these changes, and the identification of cell signaling pathways leading to the missplicing of genes in hepatocarcinogenesis.

18.
J Clin Invest ; 124(7): 2909-20, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24865429

ABSTRACT

A precise equilibrium between cellular differentiation and proliferation is fundamental for tissue homeostasis. Maintaining this balance is particularly important for the liver, a highly differentiated organ with systemic metabolic functions that is endowed with unparalleled regenerative potential. Carcinogenesis in the liver develops as the result of hepatocellular de-differentiation and uncontrolled proliferation. Here, we identified SLU7, which encodes a pre-mRNA splicing regulator that is inhibited in hepatocarcinoma, as a pivotal gene for hepatocellular homeostasis. SLU7 knockdown in human liver cells and mouse liver resulted in profound changes in pre-mRNA splicing and gene expression, leading to impaired glucose and lipid metabolism, refractoriness to key metabolic hormones, and reversion to a fetal-like gene expression pattern. Additionally, loss of SLU7 also increased hepatocellular proliferation and induced a switch to a tumor-like glycolytic phenotype. Slu7 governed the splicing and/or expression of multiple genes essential for hepatocellular differentiation, including serine/arginine-rich splicing factor 3 (Srsf3) and hepatocyte nuclear factor 4α (Hnf4α), and was critical for cAMP-regulated gene transcription. Together, out data indicate that SLU7 is central regulator of hepatocyte identity and quiescence.


Subject(s)
Liver/metabolism , RNA Splicing , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Carcinoma, Hepatocellular/etiology , Cell Differentiation , Gene Expression , Gene Knockdown Techniques , Glucose/metabolism , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Homeostasis , Humans , Lipid Metabolism , Liver Neoplasms/etiology , Male , Mice , Mice, Inbred C57BL , RNA Splicing Factors , Ribonucleoproteins, Small Nuclear/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors
19.
PLoS One ; 7(12): e52711, 2012.
Article in English | MEDLINE | ID: mdl-23285165

ABSTRACT

Hepatocellular carcinoma (HCC) is the most prevalent liver tumor and a deadly disease with limited therapeutic options. Dysregulation of cell signaling pathways is a common denominator in tumorigenesis, including hepatocarcinogenesis. The epidermal growth factor receptor (EGFR) signaling system is commonly activated in HCC, and is currently being evaluated as a therapeutic target in combination therapies. We and others have identified a central role for the EGFR ligand amphiregulin (AR) in the proliferation, survival and drug resistance of HCC cells. AR expression is frequently up-regulated in HCC tissues and cells through mechanisms not completely known. Here we identify the ß-catenin signaling pathway as a novel mechanism leading to transcriptional activation of the AR gene in human HCC cells. Activation of ß-catenin signaling, or expression of the T41A ß-catenin active mutant, led to the induction of AR expression involving three specific ß-catenin-Tcf responsive elements in its proximal promoter. We demonstrate that HCC cells expressing the T41A ß-catenin active mutant show enhanced proliferation that is dependent in part on AR expression and EGFR signaling. We also demonstrate here a novel cross-talk of the EGFR system with fibroblast growth factor 19 (FGF19). FGF19 is a recently identified driver gene in hepatocarcinogenesis and an activator of ß-catenin signaling in HCC and colon cancer cells. We show that FGF19 induced AR gene expression through the ß-catenin pathway in human HCC cells. Importantly, AR up-regulation and EGFR signaling participated in the induction of cyclin D1 and cell proliferation elicited by FGF19. Finally, we demonstrate a positive correlation between FGF19 and AR expression in human HCC tissues, therefore supporting in clinical samples our experimental observations. These findings identify the AR/EGFR system as a key mediator of FGF19 responses in HCC cells involving ß-catenin signaling, and suggest that combined targeting of FGF19 and AR/EGFR may enhance therapeutic efficacy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Signal Transduction , beta Catenin/metabolism , Amphiregulin , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , EGF Family of Proteins , ErbB Receptors/metabolism , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Neoplastic , Glycoproteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mutation , Prognosis , Promoter Regions, Genetic , Transcription Factor 4 , Transcription Factors/metabolism , beta Catenin/genetics
20.
Cancers (Basel) ; 3(2): 2444-61, 2011 May 18.
Article in English | MEDLINE | ID: mdl-24212818

ABSTRACT

Hepatocarcinogenesis is a complex multistep process in which many different molecular pathways have been implicated. Hepatocellular carcinoma (HCC) is refractory to conventional chemotherapeutic agents, and the new targeted therapies are meeting with limited success. Interreceptor crosstalk and the positive feedback between different signaling systems are emerging as mechanisms of targeted therapy resistance. The identification of such interactions is therefore of particular relevance to improve therapeutic efficacy. Among the different signaling pathways activated in hepatocarcinogenesis the epidermal growth factor receptor (EGFR) system plays a prominent role, being recognized as a "signaling hub" where different extracellular growth and survival signals converge. EGFR can be transactivated in response to multiple heterologous ligands through the physical interaction with multiple receptors, the activity of intracellular kinases or the shedding of EGFR-ligands. In this article we review the crosstalk between the EGFR and other signaling pathways that could be relevant to liver cancer development and treatment.

SELECTION OF CITATIONS
SEARCH DETAIL