Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Type of study
Language
Publication year range
1.
Cells ; 8(8)2019 08 12.
Article in English | MEDLINE | ID: mdl-31409057

ABSTRACT

Background: Group VIA calcium-independent phospholipase A2 (iPla2ß) regulates homeostasis and remodeling of phospholipids (PL). We previously showed that iPla2ß-/- mice fed with a methionine-choline-deficient diet (MCD) exhibited exaggerated liver fibrosis. As iPla2ß is located in the endoplasmic reticulum (ER), we investigated the mechanisms for this by focusing on hepatic ER unfolded protein response (UPR), ER PL, and enterohepatic bile acids (BA). Methods: Female WT (wild-type) and iPla2ß-/- mice were fed with chow or MCD for 5 weeks. PL and BA profiles were measured by liquid chromatography-mass spectrometry. Gene expression analyses were performed. Results: MCD feeding of WT mice caused a decrease of ER PL subclasses, which were further decreased by iPla2ß deficiency. This deficiency alone or combined with MCD downregulated the expression of liver ER UPR proteins and farnesoid X-activated receptor. The downregulation under MCD was concomitant with an elevation of BA in the liver and peripheral blood and an increase of biliary epithelial cell proliferation measured by cytokeratin 19. Conclusion: iPla2ß deficiency combined with MCD severely disturbed ER PL composition and caused inactivation of UPR, leading to downregulated Fxr, exacerbated BA, and ductular proliferation. Our study provides insights into iPla2ß inactivation for injury susceptibility under normal conditions and liver fibrosis and cholangiopathies during MCD feeding.


Subject(s)
Endoplasmic Reticulum/metabolism , Fibrosis/metabolism , Group VI Phospholipases A2/physiology , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Bile Acids and Salts/metabolism , Cell Proliferation , Disease Models, Animal , Endoplasmic Reticulum/pathology , Female , Liver/pathology , Mice , Mice, Inbred C57BL , Phospholipids/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Unfolded Protein Response
2.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(5): 677-687, 2019 05.
Article in English | MEDLINE | ID: mdl-30735854

ABSTRACT

Group VIA calcium-independent phospholipase A2 (iPla2ß) is among modifier genes of non-alcoholic fatty liver disease which leads to non-alcoholic steatohepatitis (NASH). Consistently, iPla2ß deletion protects hepatic steatosis and obesity in genetic ob/ob and obese mice chronically fed with high-fat diet by replenishing the loss of hepatic phospholipids (PL). As mouse feeding with methionine- and choline-deficient (MCD) diet is a model of lean NASH, we tested whether iPla2ß-null mice could still be protected since PL syntheses are disturbed. MCD-diet feeding of female wild-type for 5 weeks induced hepatic steatosis with a severe reduction of body and visceral fat weights concomitant with a decrease of hepatic phosphatidylcholine. These parameters were not altered in MCD-fed iPla2ß-null mice. However, iPla2ß deficiency attenuated MCD-induced elevation of serum transaminase activities and hepatic expression of fatty-acid translocase Cd36, fatty-acid binding protein-4, peroxisome-proliferator activated receptorγ, and HDL-uptake scavenger receptor B type 1. The reduction of lipid uptake genes was consistent with a decrease of hepatic esterified and unesterified fatty acids and cholesterol esters. On the contrary, iPla2ß deficiency under MCD did not have any effects on inflammasomes and pro-inflammatory markers but exacerbated hepatic expression of myofibroblast α-smooth muscle actin and vimentin. Thus, without any rescue of PL loss, iPla2ß inactivation attenuated hepatocellular injury in MCD-induced NASH with a novel mechanism of lipid uptake inhibition. Taken together, we have shown that iPla2ß mediates hepatic steatosis and lipotoxicity in hepatocytes in both obese and lean NASH, but elicits exacerbated liver fibrosis in lean NASH likely by affecting other cell types.


Subject(s)
Group VI Phospholipases A2/genetics , Non-alcoholic Fatty Liver Disease/genetics , Phospholipids/metabolism , Animals , Choline Deficiency/complications , Diet/adverse effects , Female , Gene Deletion , Group VI Phospholipases A2/metabolism , Methionine/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Phospholipids/genetics
3.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(5): 662-676, 2019 05.
Article in English | MEDLINE | ID: mdl-30735855

ABSTRACT

A defect of hepatic remodeling of phospholipids (PL) is seen in non-alcoholic fatty liver disease and steatohepatitis (NASH) indicating pivotal role of PL metabolism in this disease. The deletion of group VIA calcium-independent phospholipase A2 (iPla2ß) protects ob/ob mice from hepatic steatosis (BBAlip 1861, 2016, 440-461), however its role in high-fat diet (HFD)-induced NASH is still elusive. Here, wild-type and iPla2ß-null mice were subjected to chronic feeding with HFD for 6 months. We showed that protection was observed in iPla2ß-null mice with an attenuation of diet-induced body and liver-weight gains, liver enzymes, serum free fatty acids as well as hepatic TG and steatosis scores. iPla2ß deficiency under HFD attenuated the levels of 1-stearoyl lysophosphatidylcholine (LPC), lysophosphatidylethanolamine (LPE), and lysophosphatidylinositol (LPI) as well as elevation of hepatic arachidonate, arachidonate-containing cholesterol esters and prostaglandin E2. More importantly, this deficiency rescued a defect in PL remodeling and attenuated the ratio of saturated and unsaturated PL. The protection by iPla2ß deficiency was not observed during short-term HFD feeding of 3 or 5 weeks which showed no PL remodeling defect. In addition to PC/PE, this deficiency reversed the suppression of PC/PI and PE/PI among monounsaturated PL. However, this deficiency did not modulate hepatic PL contents and PL ratios in ER fractions, ER stress, fibrosis, and inflammation markers. Hence, iPla2ß inactivation protected mice against hepatic steatosis and obesity during chronic dietary NASH by correcting PL remodeling defect and PI composition relative to PC and PE.


Subject(s)
Gene Deletion , Group VI Phospholipases A2/genetics , Non-alcoholic Fatty Liver Disease/genetics , Phospholipids/metabolism , Animals , Diet, High-Fat/adverse effects , Group VI Phospholipases A2/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Phospholipids/genetics , Protective Factors
4.
Nat Commun ; 9(1): 3760, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30218046

ABSTRACT

Interactions between the gut microbial ecosystem and host lipid homeostasis are highly relevant to host physiology and metabolic diseases. We present a comprehensive multi-omics view of the effect of intestinal microbial colonization on hepatic lipid metabolism, integrating transcriptomic, proteomic, phosphoproteomic, and lipidomic analyses of liver and plasma samples from germfree and specific pathogen-free mice. Microbes induce monounsaturated fatty acid generation by stearoyl-CoA desaturase 1 and polyunsaturated fatty acid elongation by fatty acid elongase 5, leading to significant alterations in glycerophospholipid acyl-chain profiles. A composite classification score calculated from the observed alterations in fatty acid profiles in germfree mice clearly differentiates antibiotic-treated mice from untreated controls with high sensitivity. Mechanistic investigations reveal that acetate originating from gut microbial degradation of dietary fiber serves as precursor for hepatic synthesis of C16 and C18 fatty acids and their related glycerophospholipid species that are also released into the circulation.


Subject(s)
Acetates/metabolism , Acetyltransferases/metabolism , Dietary Fiber/metabolism , Fatty Acids/metabolism , Gastrointestinal Microbiome/physiology , Liver/metabolism , Stearoyl-CoA Desaturase/metabolism , Animals , Fatty Acid Elongases , Fatty Acids, Monounsaturated/metabolism , Fatty Acids, Unsaturated/metabolism , Gene Expression Profiling , Germ-Free Life , Lipid Metabolism , Mice , Proteomics
SELECTION OF CITATIONS
SEARCH DETAIL