Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Biochim Biophys Acta ; 1851(7): 946-55, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25732850

ABSTRACT

Although expression of the human liver fatty acid binding protein (FABP1) T94A variant alters serum lipoprotein cholesterol levels in human subjects, nothing is known whereby the variant elicits these effects. This issue was addressed by in vitro cholesterol binding assays using purified recombinant wild-type (WT) FABP1 T94T and T94A variant proteins and in cultured primary human hepatocytes expressing the FABP1 T94T (genotyped as TT) or T94A (genotyped as CC) proteins. The human FABP1 T94A variant protein had 3-fold higher cholesterol-binding affinity than the WT FABP1 T94T as shown by NBD-cholesterol fluorescence binding assays and by cholesterol isothermal titration microcalorimetry (ITC) binding assays. CC variant hepatocytes also exhibited 30% higher total FABP1 protein. HDL- and LDL-mediated NBD-cholesterol uptake was faster in CC variant than TT WT human hepatocytes. VLDL-mediated uptake of NBD-cholesterol did not differ between CC and TT human hepatocytes. The increased HDL- and LDL-mediated NBD-cholesterol uptake was not associated with any significant change in mRNA levels of SCARB1, LDLR, CETP, and LCAT encoding the key proteins in lipoprotein cholesterol uptake. Thus, the increased HDL- and LDL-mediated NBD-cholesterol uptake by CC hepatocytes may be associated with higher affinity of T94A protein for cholesterol and/or increased total T94A protein level.


Subject(s)
Cholesterol/metabolism , Fatty Acid-Binding Proteins/genetics , Hepatocytes/metabolism , Mutation, Missense , Alanine/genetics , Amino Acid Substitution , Animals , Biological Transport/genetics , Cells, Cultured , Fatty Acid-Binding Proteins/metabolism , Female , Humans , Lipid Metabolism/genetics , Middle Aged , Threonine/genetics
2.
Am J Physiol Gastrointest Liver Physiol ; 310(9): G726-38, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26968211

ABSTRACT

Hepatic inflammation and fibrosis are key elements in the pathogenesis of nonalcoholic steatohepatitis (NASH), a progressive liver disease initiated by excess hepatic lipid accumulation. Lipid droplet protein Perilipin 2 (Plin2) alleviates dietary-induced hepatic steatosis when globally ablated; however, its role in the progression of NASH remains unknown. To investigate this further, we challenged Plin2 liver-specific knockout mice (designated L-KO) and their respective wild-type (WT) controls with a methionine-choline-deficient (MCD) diet for 15 days to induce a NASH phenotype of increased hepatic triglyceride levels through impaired phosphatidylcholine (PC) synthesis and very-low-density lipoprotein (VLDL) secretion. Results on liver weights, body weights, fat tissue mass, and histology in WT and L-KO mice fed the MCD diet revealed signs of hepatic steatosis, fibrosis, and inflammation; however, these effects were blunted in L-KO mice. In addition, levels of PC and VLDL were unchanged, and hepatic steatosis was reduced in L-KO mice fed the MCD diet, due in part to an increase in remodeling of PE to PC via the enzyme phosphatidylethanolamine N-methyltransferase (PEMT). These mice also exhibited decreased hepatic expression of proinflammatory markers cyclooxygenase 2, IL-6, TNF-α, IL-1ß, and reduced expression of endoplasmic reticulum (ER) stress proteins C/EBP homologous protein and cleaved caspase-1. Taken together, these results suggest that Plin2 liver-specific ablation alleviates diet-induced hepatic steatosis and inflammation via a PEMT-mediated mechanism that involves compensatory changes in proteins involved in phospholipid remodeling, inflammation, and ER stress that work to alleviate diet-induced NASH. Overall, these findings support a role for Plin2 as a target for NASH therapy.


Subject(s)
Liver Cirrhosis/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Perilipin-2/genetics , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Caspase 1/genetics , Caspase 1/metabolism , Choline Deficiency/complications , Cytokines/genetics , Cytokines/metabolism , Lipoproteins, LDL/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/genetics , Methionine/deficiency , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Perilipin-2/metabolism , Phosphatidylcholines/metabolism , Phosphatidylethanolamine N-Methyltransferase/metabolism , Triglycerides/metabolism
3.
Arch Biochem Biophys ; 580: 41-9, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26116377

ABSTRACT

Although roles for both sterol carrier protein-2/sterol carrier protein-x (SCP-2/SCP-x) and liver fatty acid binding protein (L-FABP) have been proposed in hepatic lipid accumulation, individually ablating these genes has been complicated by concomitant alterations in the other gene product(s). For example, ablating SCP2/SCP-x induces upregulation of L-FABP in female mice. Therefore, the impact of ablating SCP-2/SCP-x (DKO) or L-FABP (LKO) individually or both together (TKO) was examined in female mice. Loss of SCP-2/SCP-x (DKO, TKO) more so than loss of L-FABP alone (LKO) increased hepatic total lipid and total cholesterol content, especially cholesteryl ester. Hepatic accumulation of nonesterified long chain fatty acids (LCFA) and phospholipids occurred only in DKO and TKO mice. Loss of SCP-2/SCP-x (DKO, TKO) increased serum total lipid primarily by increasing triglycerides. Altered hepatic level of proteins involved in cholesterol uptake, efflux, and/or secretion was observed, but did not compensate for the loss of L-FABP, SCP-2/SCP-x or both. However, synergistic responses were not seen with the combinatorial knock out animals-suggesting that inhibiting SCP-2/SCP-x is more correlative with hepatic dysfunction than L-FABP. The DKO- and TKO-induced hepatic accumulation of cholesterol and long chain fatty acids shared significant phenotypic similarities with non-alcoholic fatty liver disease (NAFLD).


Subject(s)
Carrier Proteins/genetics , Fatty Acid-Binding Proteins/genetics , Lipid Metabolism/genetics , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Carrier Proteins/metabolism , Cholesterol/blood , Cholesterol Esters/blood , Disease Models, Animal , Fatty Acid-Binding Proteins/deficiency , Fatty Acids, Nonesterified/blood , Female , Gene Deletion , Gene Expression , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Phospholipids/blood , Triglycerides/blood
4.
Arch Biochem Biophys ; 588: 25-32, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26541319

ABSTRACT

Both sterol carrier protein-2/sterol carrier protein-x (SCP-2/SCP-x) and liver fatty acid binding protein (L-FABP) have been proposed to function in hepatobiliary bile acid metabolism/accumulation. To begin to address this issue, the impact of ablating L-FABP (LKO) or SCP-2/SCP-x (DKO) individually or both together (TKO) was examined in female mice. Biliary bile acid levels were decreased in LKO, DKO, and TKO mice; however, hepatic bile acid concentration was decreased in LKO mice only. In contrast, biliary phospholipid level was decreased only in TKO mice, while biliary cholesterol levels were unaltered regardless of phenotype. The loss of either or both genes increased hepatic expression of the major bile acid synthetic enzymes (CYP7A1 and/or CYP27A1). Loss of L-FABP and/or SCP-2/SCP-x genes significantly altered the molecular composition of biliary bile acids, but not the proportion of conjugated/unconjugated bile acids or overall bile acid hydrophobicity index. These data suggested that L-FABP was more important in hepatic retention of bile acids, while SCP-2/SCP-x more broadly affected biliary bile acid and phospholipid levels.


Subject(s)
Biliary Tract/metabolism , Carrier Proteins/metabolism , Fatty Acid-Binding Proteins/metabolism , Liver/metabolism , Animals , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , Carrier Proteins/genetics , Cholesterol/metabolism , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phospholipids/metabolism
5.
Biochemistry ; 53(45): 7051-66, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25338003

ABSTRACT

Although perilipin 2 (Plin2) has been shown to bind lipids with high affinity, the Plin2 lipid binding site has yet to be defined. This is of interest since Plin2's affinity for lipids has been suggested to be important for lipid droplet biogenesis and intracellular triacylglycerol accumulation. To define these regions, mouse Plin2 and several deletion mutants expressed as recombinant proteins and in mammalian cells were assessed by molecular modeling, fluorescence binding, circular dichroic, and fluorescence resonance energy transfer techniques to identify the structural and functional requirements for lipid binding. Major findings of this study indicate (1) the N-terminal PAT domain does not bind cholesterol or stearic acid; (2) Plin2 residues 119-251, containing helix α4, the α-ß domain, and part of helix α6 form a Plin3-like cleft found to be important for highest affinity lipid binding; (3) both stearic acid and cholesterol interact favorably with the Plin2 cleft formed by conserved residues in helix α6 and adjacent strands, which is common to all the active lipid-binding constructs; and (4) discrete targeting of the Plin2 mutants to lipid droplets supports Plin2 containing two independent, nonoverlapping lipid droplet targeting domains in its central and C-terminal sequences. Thus, the current work reveals specific domains responsible for Plin2-lipid interactions that involves the protein's lipid binding and targeting functions.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/physiology , Animals , Binding Sites/physiology , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Mice , Perilipin-2 , Protein Structure, Secondary , Protein Structure, Tertiary
6.
Am J Physiol Gastrointest Liver Physiol ; 307(11): G1130-43, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25277800

ABSTRACT

On the basis of their abilities to bind bile acids and/or cholesterol, the physiological role(s) of liver fatty acid-binding protein (L-FABP) and sterol carrier protein (SCP) 2/SCP-x (SCP-2/SCP-x) gene products in biliary bile acid and cholesterol formation was examined in gene-ablated male mice. L-FABP (LKO) or L-FABP/SCP-2/SCP-x [triple-knockout (TKO)] ablation markedly decreased hepatic bile acid concentration, while SCP-2/SCP-x [double-knockout (DKO)] ablation alone had no effect. In contrast, LKO increased biliary bile acid, while DKO and TKO had no effect on biliary bile acid levels. LKO and DKO also altered biliary bile acid composition to increase bile acid hydrophobicity. Furthermore, LKO and TKO decreased hepatic uptake and biliary secretion of high-density lipoprotein (HDL)-derived 22-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-23,24-bisnor-5-cholen-3ß-ol (NBD-cholesterol), while DKO alone had no effect. Finally, LKO and, to a lesser extent, DKO decreased most indexes contributing to cholesterol solubility in biliary bile. These results suggest different, but complementary, roles for L-FABP and SCP-2/SCP-x in biliary bile acid and cholesterol formation. L-FABP appears to function more in hepatic retention of bile acids as well as hepatic uptake and biliary secretion of HDL-cholesterol. Conversely, SCP-2/SCP-x may function more in formation and biliary secretion of bile acid, with less impact on hepatic uptake or biliary secretion of HDL-cholesterol.


Subject(s)
Bile Acids and Salts/metabolism , Bile/metabolism , Carrier Proteins/physiology , Cholesterol, HDL/metabolism , Fatty Acid-Binding Proteins/physiology , Animals , Carrier Proteins/genetics , Cholesterol/metabolism , Fatty Acid-Binding Proteins/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phospholipids/metabolism
7.
Am J Physiol Gastrointest Liver Physiol ; 307(2): G164-76, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24875102

ABSTRACT

Although human liver fatty acid-binding protein (FABP1) T94A variant has been associated with nonalcoholic fatty liver disease and reduced ability of fenofibrate to lower serum triglycerides (TG) to target levels, molecular events leading to this phenotype are poorly understood. Cultured primary hepatocytes from female human subjects expressing the FABP1 T94A variant exhibited increased neutral lipid (TG, cholesteryl ester) accumulation associated with (1) upregulation of total FABP1, a key protein stimulating mitochondrial glycerol-3-phosphate acyltransferase (GPAM), the rate-limiting enzyme in lipogenesis; (2) increased mRNA expression of key enzymes in lipogenesis (GPAM, LPIN2) in heterozygotes; (3) decreased mRNA expression of microsomal triglyceride transfer protein; (4) increased secretion of ApoB100 but not TG; (5) decreased long-chain fatty acid (LCFA) ß-oxidation. TG accumulation was not due to any increase in LCFA uptake, de novo lipogenesis, or the alternate monoacylglycerol O-acyltransferase pathway in lipogenesis. Despite increased expression of total FABP1 mRNA and protein, fenofibrate-mediated FABP1 redistribution to nuclei and ligand-induced peroxisome proliferator-activated receptor (PPAR-α) transcription of LCFA ß-oxidative enzymes (carnitine palmitoyltransferase 1A, carnitine palmitoyltransferase 2, and acyl-coenzyme A oxidase 1, palmitoyl) were attenuated in FABP1 T94A hepatocytes. Although the phenotype of FABP1 T94A variant human hepatocytes exhibits some similarities to that of FABP1-null or PPAR-α-null hepatocytes and mice, expression of FABP1 T94A variant did not abolish or reduce ligand binding. Thus the FABP1 T94A variant represents an altered/reduced function mutation resulting in TG accumulation.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Fatty Acids/metabolism , Hepatocytes/metabolism , PPAR alpha/metabolism , Apolipoprotein B-100/metabolism , Cells, Cultured , Cholesterol Esters/metabolism , Fatty Acid-Binding Proteins/genetics , Female , Fenofibrate/pharmacology , Gene Expression Regulation, Enzymologic , Genetic Variation , Hepatocytes/drug effects , Heterozygote , Homozygote , Humans , Hypolipidemic Agents/pharmacology , Lipogenesis/genetics , Middle Aged , Oxidation-Reduction , PPAR alpha/agonists , Phenotype , Protein Binding , Protein Transport , RNA, Messenger/metabolism , Time Factors , Transcription, Genetic , Triglycerides/metabolism
8.
Biochemistry ; 52(51): 9347-57, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24299557

ABSTRACT

Although the human liver fatty acid binding protein (L-FABP) T94A variant arises from the most commonly occurring single-nucleotide polymorphism in the entire FABP family, there is a complete lack of understanding regarding the role of this polymorphism in human disease. It has been hypothesized that the T94A substitution results in the complete loss of ligand binding ability and function analogous to that seen with L-FABP gene ablation. This possibility was addressed using the recombinant human wild-type (WT) T94T and T94A variant L-FABP and cultured primary human hepatocytes. Nonconservative replacement of the medium-sized, polar, uncharged T residue with a smaller, nonpolar, aliphatic A residue at position 94 of the human L-FABP significantly increased the L-FABP α-helical structure content at the expense of ß-sheet content and concomitantly decreased the thermal stability. T94A did not alter the binding affinities for peroxisome proliferator-activated receptor α (PPARα) agonist ligands (phytanic acid, fenofibrate, and fenofibric acid). While T94A did not alter the impact of phytanic acid and only slightly altered that of fenofibrate on the human L-FABP secondary structure, the active metabolite fenofibric acid altered the T94A secondary structure much more than that of the WT T94T L-FABP. Finally, in cultured primary human hepatocytes, the T94A variant exhibited a significantly reduced extent of fibrate-mediated induction of PPARα-regulated proteins such as L-FABP, FATP5, and PPARα itself. Thus, while the T94A substitution did not alter the affinity of the human L-FABP for PPARα agonist ligands, it significantly altered the human L-FABP structure, stability, and conformational and functional response to fibrate.


Subject(s)
Fatty Acid-Binding Proteins/genetics , Fibric Acids/pharmacology , Hypolipidemic Agents/pharmacology , Liver/metabolism , Polymorphism, Single Nucleotide , Amino Acid Substitution , Animals , Binding Sites , Cells, Cultured , Fatty Acid Transport Proteins/agonists , Fatty Acid Transport Proteins/genetics , Fatty Acid Transport Proteins/metabolism , Fatty Acid-Binding Proteins/agonists , Fatty Acid-Binding Proteins/chemistry , Fatty Acid-Binding Proteins/metabolism , Fenofibrate/analogs & derivatives , Fenofibrate/metabolism , Fenofibrate/pharmacology , Fibric Acids/metabolism , Gene Expression Regulation/drug effects , Hot Temperature , Humans , Hypolipidemic Agents/metabolism , Ligands , Liver/cytology , Mice , PPAR alpha/agonists , PPAR alpha/genetics , PPAR alpha/metabolism , Phytanic Acid/metabolism , Phytanic Acid/pharmacology , Protein Stability , Protein Structure, Secondary , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
9.
Am J Physiol Cell Physiol ; 303(7): C728-42, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22744009

ABSTRACT

Despite increasing awareness of the health risks associated with excess lipid storage in cells and tissues, knowledge of events governing lipid exchange at the surface of lipid droplets remains unclear. To address this issue, fluorescence resonance energy transfer (FRET) was performed to examine live cell interactions of Plin2 with lipids involved in maintaining lipid droplet structure and function. FRET efficiencies (E) between CFP-labeled Plin2 and fluorescently labeled phosphatidylcholine, sphingomyelin, stearic acid, and cholesterol were quantitated on a pixel-by-pixel basis to generate FRET image maps that specified areas with high E (>60%) in lipid droplets. The mean E and the distance R between the probes indicated a high yield of energy transfer and demonstrated molecular distances on the order of 44-57 Å, in keeping with direct molecular contact. In contrast, FRET between CFP-Plin2 and Nile red was not detected, indicating that the CFP-Plin2/Nile red interaction was beyond FRET proximity (>100 Å). An examination of the effect of Plin2 on cellular metabolism revealed that triacylglycerol, fatty acid, and cholesteryl ester content increased while diacylglycerol remained constant in CFP-Plin2-overexpressing cells. Total phospholipids also increased, reflecting increased phosphatidylcholine and sphingomyelin. Consistent with these results, expression levels of enzymes involved in triacylglycerol, cholesteryl ester, and phospholipid synthesis were significantly upregulated in CFP-Plin2-expressing cells while those associated with lipolysis either decreased or were unaffected. Taken together, these data show for the first time that Plin2 interacts directly with lipids on the surface of lipid droplets and influences levels of key enzymes and lipids involved in maintaining lipid droplet structure and function.


Subject(s)
Fibroblasts/metabolism , Fluorescence Resonance Energy Transfer/methods , Lipid Metabolism/physiology , Membrane Proteins/metabolism , Animals , Cells, Cultured , Humans , Mice , Perilipin-2 , Protein Binding/physiology , Structure-Activity Relationship , Surface Properties
10.
J Lipid Res ; 53(3): 467-480, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22223861

ABSTRACT

Although lipid-rich microdomains of hepatocyte plasma membranes serve as the major scaffolding regions for cholesterol transport proteins important in cholesterol disposition, little is known regarding intracellular factors regulating cholesterol distribution therein. On the basis of its ability to bind cholesterol and alter hepatic cholesterol accumulation, the cytosolic liver type FA binding protein (L-FABP) was hypothesized to be a candidate protein regulating these microdomains. Compared with wild-type hepatocyte plasma membranes, L-FABP gene ablation significantly increased the proportion of cholesterol-rich microdomains. Lack of L-FABP selectively increased cholesterol, phospholipid (especially phosphatidylcholine), and branched-chain FA accumulation in the cholesterol-rich microdomains. These cholesterol-rich microdomains are important, owing to enrichment therein of significant amounts of key transport proteins involved in uptake of cholesterol [SR-B1, ABCA-1, P-glycoprotein (P-gp), sterol carrier binding protein (SCP-2)], FA transport protein (FATP), and glucose transporters 1 and 2 (GLUT1, GLUT2) insulin receptor. L-FABP gene ablation enhanced the concentration of SCP-2, SR-B1, FATP4, and GLUT1 in the cholesterol-poor microdomains, with functional implications in HDL-mediated uptake and efflux of cholesterol. Thus L-FABP gene ablation significantly impacted the proportion of cholesterol-rich versus -poor microdomains in the hepatocyte plasma membrane and altered the distribution of lipids and proteins involved in cholesterol uptake therein.


Subject(s)
Cell Membrane/metabolism , Fatty Acid-Binding Proteins/metabolism , Hepatocytes/metabolism , Liver/metabolism , Membrane Microdomains/metabolism , Animals , Blotting, Western , Cell Membrane/genetics , Cells, Cultured , Cholesterol/genetics , Cholesterol/metabolism , Fatty Acid Transport Proteins/genetics , Fatty Acid Transport Proteins/metabolism , Fatty Acid-Binding Proteins/genetics , Membrane Microdomains/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Confocal , Phospholipids/chemistry , Phospholipids/metabolism
11.
Am J Physiol Gastrointest Liver Physiol ; 302(8): G824-39, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22241858

ABSTRACT

A major gap in our knowledge of rapid hepatic HDL cholesterol clearance is the role of key intracellular factors that influence this process. Although the reverse cholesterol transport pathway targets HDL to the liver for net elimination of free cholesterol from the body, molecular details governing cholesterol uptake into hepatocytes are not completely understood. Therefore, the effects of sterol carrier protein (SCP)-2 and liver fatty acid-binding protein (L-FABP), high-affinity cholesterol-binding proteins present in hepatocyte cytosol, on HDL-mediated free cholesterol uptake were examined using gene-targeted mouse models, cultured primary hepatocytes, and 22-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)-amino]-23,24-bisnor-5-cholen-3ß-ol (NBD-cholesterol). While SCP-2 overexpression enhanced NBD-cholesterol uptake, counterintuitively, SCP-2/SCP-x gene ablation also 1) enhanced the rapid molecular phase of free sterol uptake detectable in <1 min and initial rate and maximal uptake of HDL free cholesterol and 2) differentially enhanced free cholesterol uptake mediated by the HDL3, rather than the HDL2, subfraction. The increased HDL free cholesterol uptake was not due to increased expression or distribution of the HDL receptor [scavenger receptor B1 (SRB1)], proteins regulating SRB1 [postsynaptic density protein (PSD-95)/Drosophila disk large tumor suppressor (dlg)/tight junction protein (ZO1) and 17-kDa membrane-associated protein], or other intracellular cholesterol trafficking proteins (steroidogenic acute response protein D, Niemann Pick C, and oxysterol-binding protein-related proteins). However, expression of L-FABP, the single most prevalent hepatic cytosolic protein that binds cholesterol, was upregulated twofold in SCP-2/SCP-x null hepatocytes. Double-immunogold electron microscopy detected L-FABP sufficiently close to SRB1 for direct interaction, similar to SCP-2. These data suggest a role for L-FABP in HDL cholesterol uptake, a finding confirmed with SCP-2/SCP-x/L-FABP null mice and hepatocytes. Taken together, these results suggest that L-FABP, particularly in the absence of SCP-2, plays a significant role in HDL-mediated cholesterol uptake in cultured primary hepatocytes.


Subject(s)
Carrier Proteins/metabolism , Cholesterol, HDL/metabolism , Cholesterol/metabolism , Fatty Acid-Binding Proteins/metabolism , Hepatocytes/metabolism , 4-Chloro-7-nitrobenzofurazan/analogs & derivatives , 4-Chloro-7-nitrobenzofurazan/pharmacology , Animals , Blotting, Western , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Separation , Cells, Cultured , Cholesterol/analogs & derivatives , Cholesterol/pharmacology , Fatty Acid-Binding Proteins/biosynthesis , Fatty Acid-Binding Proteins/genetics , Immunohistochemistry , Lipoproteins/biosynthesis , Lipoproteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Immunoelectron
12.
J Biol Chem ; 285(24): 18693-708, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20382741

ABSTRACT

Despite the considerable beneficial effects of n-3 and n-6 very long chain polyunsaturated fatty acids (VLC-PUFAs), very little is known about the factors that regulate their uptake and intracellular distribution in living cells. This issue was addressed in cells expressing liver-type fatty acid-binding protein (L-FABP) by real time multiphoton laser scanning microscopy of novel fluorescent VLC-PUFAs containing a conjugated tetraene fluorophore near the carboxyl group and natural methylene-interrupted n-3 or n-6 grouping. The fluorescent VLC-PUFAs mimicked many properties of their native nonfluorescent counterparts, including uptake, distribution, and metabolism in living cells. The unesterified fluorescent VLC-PUFAs distributed either equally in nuclei versus cytoplasm (22-carbon n-3 VLC-PUFA) or preferentially to cytoplasm (20-carbon n-3 and n-6 VLC-PUFAs). L-FABP bound fluorescent VLC-PUFA with affinity and specificity similar to their nonfluorescent natural counterparts. Regarding n-3 and n-6 VLC-PUFA, L-FABP expression enhanced uptake into the cell and cytoplasm, selectively altered the pattern of fluorescent n-6 and n-3 VLC-PUFA distribution in cytoplasm versus nuclei, and preferentially distributed fluorescent VLC-PUFA into nucleoplasm versus nuclear envelope, especially for the 22-carbon n-3 VLC-PUFA, correlating with its high binding by L-FABP. Multiphoton laser scanning microscopy data showed for the first time VLC-PUFA in nuclei of living cells and suggested a model, whereby L-FABP facilitated VLC-PUFA targeting to nuclei by enhancing VLC-PUFA uptake and distribution into the cytoplasm and nucleoplasm.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Fatty Acids, Unsaturated/chemistry , Fluorescent Dyes/chemistry , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Fatty Acids/metabolism , Fibroblasts/metabolism , Fluorescent Dyes/pharmacology , Kinetics , Liver/metabolism , Mice , Microscopy, Fluorescence/methods , Photons , Rats , Recombinant Proteins/metabolism
13.
Am J Physiol Endocrinol Metab ; 301(5): E991-E1003, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21846905

ABSTRACT

The significance of lipid droplets (LD) in lipid metabolism, cell signaling, and membrane trafficking is increasingly recognized, yet the role of the LD phospholipid monolayer in LD protein targeting and function remains unknown. To begin to address this issue, two populations of LD were isolated by ConA sepharose affinity chromatography: 1) functionally active LD enriched in perilipin, caveolin-1, and several lipolytic proteins, including ATGL and HSL; and 2) LD enriched in ADRP and TIP47 that contained little to no lipase activity. Coimmunoprecipitation experiments confirmed the close association of caveolin and perilipin and lack of interaction between caveolin and ADRP, in keeping with the separation observed with the ConA procedure. The phospholipid monolayer structure was evaluated to reveal that the perilipin-enriched LD exhibited increased rigidity (less fluidity), as shown by increased cholesterol/phospholipid, Sat/Unsat, and Sat/MUFA ratios. These results were confirmed by DPH-TMA, NBD-cholesterol, and NBD-sphingomyelin fluorescence polarization studies. By structure and organization, the perilipin-enriched LD most closely resembled the adipocyte PM. In contrast, the ADRP/TIP47-enriched LD contained a more fluid monolayer membrane, reflecting decreased polarizations and lipid order based on phospholipid fatty acid analysis. Taken together, results indicate that perilipin and associated lipolytic enzymes target areas in the phospholipid monolayer that are highly organized and rigid, similar in structure to localized areas of the PM where cholesterol and fatty acid uptake and efflux occur.


Subject(s)
Carrier Proteins/metabolism , Cytoplasmic Vesicles/metabolism , Membrane Fluidity , Phosphoproteins/metabolism , Adipocytes/chemistry , Adipocytes/metabolism , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane Structures/chemistry , Cell Membrane Structures/metabolism , Cells, Cultured , Chromatography, Affinity , Cytoplasmic Vesicles/chemistry , Lipid Metabolism/physiology , Male , Membrane Fluidity/physiology , Mice , Mice, Inbred C57BL , Perilipin-1 , Phospholipids/chemistry , Phospholipids/metabolism , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism
14.
Subcell Biochem ; 51: 279-318, 2010.
Article in English | MEDLINE | ID: mdl-20213548

ABSTRACT

While the existence of membrane lateral microdomains has been known for over 30 years, interest in these structures accelerated in the past decade due to the discovery that cholesterol-rich microdomains serve important biological functions. It is increasingly appreciated that cholesterol-rich microdomains in the plasma membranes of eukaryotic cells represent an organizing nexus for multiple cellular proteins involved in transmembrane nutrient uptake (cholesterol, fatty acid, glucose, etc.), cell-signaling, immune recognition, pathogen entry, and many other roles. Despite these advances, however, relatively little is known regarding the organization of cholesterol itself in these plasma membrane microdomains. Although a variety of non-sterol markers indicate the presence of microdomains in the plasma membranes of living cells, none of these studies have demonstrated that cholesterol is enriched in these microdomains in living cells. Further, the role of cholesterol-rich membrane microdomains as targets for intracellular cholesterol trafficking proteins such as sterol carrier protein-2 (SCP-2) that facilitate cholesterol uptake and transcellular transport for targeting storage (cholesterol esters) or efflux is only beginning to be understood. Herein, we summarize the background as well as recent progress in this field that has advanced our understanding of these issues.


Subject(s)
Carrier Proteins/physiology , Caveolins/metabolism , Cholesterol/metabolism , Membrane Microdomains/metabolism , Animals , Bile/metabolism , Carrier Proteins/genetics , Cell Membrane/physiology , Cholesterol/analogs & derivatives , Fatty Acid-Binding Proteins/physiology , Fluorescence Resonance Energy Transfer , G(M1) Ganglioside/metabolism , Humans , Liver/metabolism , Membrane Lipids/metabolism , Membrane Proteins/metabolism , Models, Biological , Phosphoproteins/physiology , Vesicular Transport Proteins/physiology
15.
J Lipid Res ; 51(5): 1157-72, 2010 May.
Article in English | MEDLINE | ID: mdl-20008119

ABSTRACT

While plasma membrane cholesterol-rich microdomains play a role in cholesterol trafficking, little is known about the appearance and dynamics of cholesterol through these domains in living cells. The fluorescent cholesterol analog 6-dansyl-cholestanol (DChol), its biochemical fractionation, and confocal imaging of L-cell fibroblasts contributed the following new insights: i) fluorescence properties of DChol were sensitive to microenvironment polarity and mobility; (ii) DChol taken up by L-cell fibroblasts was distributed similarly as cholesterol and preferentially into cholesterol-rich vs. -poor microdomains resolved by affinity chromatography of purified plasma membranes; iii) DChol reported similar polarity (dielectric constant near 18) but higher mobility near phospholipid polar head group region for cholesterol in purified cholesterol-rich versus -poor microdomains; and iv) real-time confocal imaging, quantitative colocalization analysis, and fluorescence resonance energy transfer with cholesterol-rich and -poor microdomain markers confirmed that DChol preferentially localized in plasma membrane cholesterol-rich microdomains of living cells. Thus, DChol sensed a unique, relatively more mobile microenvironment for cholesterol in plasma membrane cholesterol-rich microdomains, consistent with the known, more rapid exchange dynamics of cholesterol from cholesterol-rich than -poor microdomains.


Subject(s)
Cell Membrane/metabolism , Cholestanols/metabolism , Cholesterol/metabolism , Fluorescent Dyes/metabolism , Animals , Biological Transport , Biomarkers/metabolism , Buffers , Cell Survival , Chromatography, Affinity , Fluorescence Resonance Energy Transfer , L Cells , Membrane Microdomains/metabolism , Mice , Sterols/chemistry , Sterols/metabolism , Time Factors , Unilamellar Liposomes/metabolism , Water/metabolism
16.
Am J Physiol Gastrointest Liver Physiol ; 299(1): G244-54, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20395534

ABSTRACT

Although HDL-mediated cholesterol transport to the liver is well studied, cholesterol efflux from hepatocytes back to HDL is less well understood. Real-time imaging of efflux of 22-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)-amino)-23,24-bisnor-5-cholen-3beta-ol (NBD-cholesterol), which is poorly esterified, and [(3)H]cholesterol, which is extensively esterified, from cultured primary hepatocytes of wild-type and sterol carrier protein-2 (SCP-2) gene-ablated mice showed that 1) NBD-cholesterol efflux was affected by the type of lipoprotein acceptor, i.e., HDL3 over HDL2; 2) NBD-cholesterol efflux was rapid (detected in 1-2 min) and resolved into fast [half time (t((1/2))) = 2.4 min, 6% of total] and slow (t((1/2)) = 26.5 min, 94% of total) pools, consistent with protein- and vesicle-mediated cholesterol transfer, respectively; 3) SCP-2 gene ablation increased efflux of NBD-cholesterol, as well as [(3)H]cholesterol, albeit less so due to competition by esterification of [(3)H]cholesterol, but not NBD-cholesterol; and 4) SCP-2 gene ablation increased initial rate (2.3-fold) and size (9.7-fold) of rapid effluxing sterol, suggesting an increased contribution of molecular cholesterol transfer. In addition, colocalization, double-immunolabeling fluorescence resonance energy transfer, and electron microscopy, as well as cross-linking coimmunoprecipitation, indicated that SCP-2 directly interacted with the HDL receptor, scavenger receptor class B type 1 (SRB1), in hepatocytes. Other membrane proteins in cholesterol efflux [SRB1 and ATP-binding cassettes (ABC) A-1, ABCG-1, ABCG-5, and ABCG-8] and several soluble/vesicle-associated proteins facilitating intracellular cholesterol trafficking (StARDs, NPCs, ORPs) were not upregulated. However, loss of SCP-2 elicited twofold upregulation of liver fatty acid-binding protein (L-FABP), a protein with lower affinity for cholesterol but higher cytosolic concentration than SCP-2. Ablation of SCP-2 and L-FABP decreased HDL-mediated NBD-cholesterol efflux. These results indicate that SCP-2 expression plays a significant role in HDL-mediated cholesterol efflux by regulating the size of rapid vs. slow cholesterol efflux pools and/or eliciting concomitant upregulation of L-FABP in cultured primary hepatocytes.


Subject(s)
4-Chloro-7-nitrobenzofurazan/analogs & derivatives , Carrier Proteins/metabolism , Cholesterol/analogs & derivatives , Hepatocytes/metabolism , Lipoproteins, HDL3/metabolism , 4-Chloro-7-nitrobenzofurazan/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Biological Transport , Carrier Proteins/genetics , Cell Culture Techniques , Cells, Cultured , Cholesterol/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Fluorescence Resonance Energy Transfer , Gene Knockout Techniques , Immunoprecipitation , Kinetics , Lipoproteins, HDL2/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Phosphoproteins/metabolism , Protein Binding , Scavenger Receptors, Class B/metabolism , Transport Vesicles/metabolism
17.
J Lipid Res ; 50(8): 1663-75, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19289416

ABSTRACT

Although studies with liver type fatty acid binding protein (L-FABP) gene ablated mice demonstrate a physiological role for L-FABP in hepatic fatty acid metabolism, little is known about the mechanisms whereby L-FABP elicits these effects. Studies indicate that L-FABP may function to shuttle lipids to the nucleus, thereby increasing the availability of ligands of nuclear receptors, such as peroxisome proliferator-activated receptor-alpha (PPARalpha). The data herein suggest that such mechanisms involve direct interaction of L-FABP with PPARalpha. L-FABP was shown to directly interact with PPARalpha in vitro through co-immunoprecipitation (co-IP) of pure proteins, altered circular dichroic (CD) spectra, and altered fluorescence spectra. In vitro fluorescence resonance energy transfer (FRET) between Cy3-labeled PPARalpha and Cy5-labeled L-FABP proteins showed that these proteins bound with high affinity (Kd approximately 156 nM) and in close proximity (intermolecular distance of 52A). This interaction was further substantiated by co-IP of both proteins from liver homogenates of wild-type mice. Moreover, double immunogold electron microscopy and FRET confocal microscopy of cultured primary hepatocytes showed that L-FABP was in close proximity to PPARalpha (intermolecular distance 40-49A) in vivo. Taken together, these studies were consistent with L-FABP regulating PPARalpha transcriptional activity in hepatocytes through direct interaction with PPARalpha. Our in vitro and imaging experiments demonstrate high affinity, structural molecular interaction of L-FABP with PPARalpha and suggest a functional role for L-FABP interaction with PPARalpha in long chain fatty acid (LCFA) metabolism.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Hepatocytes/metabolism , PPAR alpha/metabolism , Animals , Cell Compartmentation , Cell Nucleus/metabolism , Cells, Cultured , Fatty Acid-Binding Proteins/chemistry , Fatty Acids/metabolism , Hepatocytes/ultrastructure , Ligands , Male , Mice , Mice, Knockout , PPAR alpha/chemistry , Palmitic Acid/metabolism , Protein Binding , Protein Conformation , Protein Structure, Quaternary , Recombinant Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism
18.
J Lipid Res ; 50(7): 1429-47, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19289417

ABSTRACT

Although in vitro studies suggest a role for sterol carrier protein-2 (SCP-2) in cholesterol trafficking and metabolism, the physiological significance of these observations remains unclear. This issue was addressed by examining the response of mice overexpressing physiologically relevant levels of SCP-2 to a cholesterol-rich diet. While neither SCP-2 overexpression nor cholesterol-rich diet altered food consumption, increased weight gain, hepatic lipid, and bile acid accumulation were observed in wild-type mice fed the cholesterol-rich diet. SCP-2 overexpression further exacerbated hepatic lipid accumulation in cholesterol-fed females (cholesterol/cholesteryl esters) and males (cholesterol/cholesteryl esters and triacyglycerol). Primarily in female mice, hepatic cholesterol accumulation induced by SCP-2 overexpression was associated with increased levels of LDL-receptor, HDL-receptor scavenger receptor-B1 (SR-B1) (as well as PDZK1 and/or membrane-associated protein 17 kDa), SCP-2, liver fatty acid binding protein (L-FABP), and 3alpha-hydroxysteroid dehydrogenase, without alteration of other proteins involved in cholesterol uptake (caveolin), esterification (ACAT2), efflux (ATP binding cassette A-1 receptor, ABCG5/8, and apolipoprotein A1), or oxidation/transport of bile salts (cholesterol 7alpha-hydroxylase, sterol 27alpha-hydroxylase, Na(+)/taurocholate cotransporter, Oatp1a1, and Oatp1a4). The effects of SCP-2 overexpression and cholesterol-rich diet was downregulation of proteins involved in cholesterol transport (L-FABP and SR-B1), cholesterol synthesis (related to sterol regulatory element binding protein 2 and HMG-CoA reductase), and bile acid oxidation/transport (via Oapt1a1, Oatp1a4, and SCP-x). Levels of serum and hepatic bile acids were decreased in cholesterol-fed SCP-2 overexpression mice, especially in females, while the total bile acid pool was minimally affected. Taken together, these findings support an important role for SCP-2 in hepatic cholesterol homeostasis.


Subject(s)
Carrier Proteins/metabolism , Cholesterol, Dietary , Cholesterol , Liver/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Bile Acids and Salts/blood , Bile Acids and Salts/metabolism , Body Weight , Carrier Proteins/genetics , Caveolin 1/genetics , Caveolin 1/metabolism , Cholestanetriol 26-Monooxygenase/genetics , Cholestanetriol 26-Monooxygenase/metabolism , Cholesterol/administration & dosage , Cholesterol/metabolism , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Female , Humans , Lipid Metabolism , Lipids/chemistry , Liver/anatomy & histology , Male , Mice , Mice, Transgenic , Organ Size , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Phenotype , Phospholipids/blood , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Tissue Distribution
19.
Am J Physiol Gastrointest Liver Physiol ; 297(6): G1053-65, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19815623

ABSTRACT

Although the function of liver fatty acid binding protein in hepatic fatty acid metabolism has been extensively studied, its potential role in hepatic cholesterol homeostasis is less clear. Although hepatic cholesterol accumulation was initially reported in L-FABP-null female mice, that study was performed with early N2 backcross generation mice. To resolve whether the hepatic cholesterol phenotype in these L-FABP(-/-) mice was attributable to genetic inhomogeneity, these L-FABP(-/-) mice were further backcrossed to C57Bl/6 mice up to the N10 (99.9% homogeneity) generation. Hepatic total cholesterol accumulation was observed in female, but not male, L-FABP(-/-) mice at all (N2, N4, N6, N10) backcross generations examined. The greater total cholesterol was due to increased hepatic levels of both unesterified (free) cholesterol and esterified cholesterol. Altered hepatic cholesterol accumulation correlated directly with L-FABP's ability to bind cholesterol with high affinity as shown by direct L-FABP binding of fluorescent cholesterol analogs (NBD-cholesterol, dansyl-cholesterol), a photoactivatable cholesterol analog [free cholesterol benzophenone (FCBP)], and free cholesterol (circular dichroism, isothermal titration microcalorimetry). One mole of fluorescent sterol was bound per mole of L-FABP. This was confirmed by photo-cross-linking studies with the photoactivatable cholesterol analog FCBP and by isothermal titration calorimetry with free cholesterol, which showed that L-FABP bound only one sterol molecule per L-FABP molecule. In contrast, the hepatic phenotype of male, but not female, L-FABP(-/-) mice was characterized by decreased hepatic triacylglycerol levels at all backcross generations examined. Taken together, these data support the hypothesis that L-FABP plays a role in physiological regulation of not only hepatic fatty acid metabolism, but also that of hepatic cholesterol.


Subject(s)
Cholesterol/metabolism , Fatty Acid-Binding Proteins/deficiency , Liver/metabolism , 4-Chloro-7-nitrobenzofurazan/analogs & derivatives , 4-Chloro-7-nitrobenzofurazan/metabolism , Animals , Binding Sites , Binding, Competitive , Calorimetry , Carrier Proteins/metabolism , Cholesterol/analogs & derivatives , Cholesterol/blood , Cholesterol Esters/metabolism , Circular Dichroism , Crosses, Genetic , Fatty Acid-Binding Proteins/chemistry , Fatty Acid-Binding Proteins/genetics , Fatty Acids/metabolism , Female , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phospholipids/metabolism , Protein Conformation , Sex Factors , Spectrometry, Fluorescence , Spectrometry, Mass, Electrospray Ionization , Triglycerides/metabolism
20.
Arch Biochem Biophys ; 485(2): 160-73, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19285478

ABSTRACT

The effect of liver type fatty acid binding protein (L-FABP) gene ablation on the uptake and distribution of long chain fatty acids (LCFA) to the nucleus by real-time laser scanning confocal imaging and peroxisome proliferator-activated receptor-alpha (PPARalpha) activity was examined in cultured primary hepatocytes from livers wild-type L-FABP+/+ and gene ablated L-FABP-/- mice. Cultured primary hepatocytes from livers of L-FABP-/- mice exhibited: (i) reduced oxidation of palmitic acid, a common dietary long chain fatty acid (LCFA); (ii) reduced expression of fatty acid oxidative enzymes-proteins transcriptionally regulated by PPARalpha; (iii) reduced palmitic acid-induced PPARalpha co-immunoprecipitation with coactivator SRC-1 concomitant with increased PPARalpha co-immunoprecipitation with coinhibitor N-CoR; (iv) reduced palmitic acid-induced PPARalpha. Diminished PPARalpha activation in L-FABP null hepatocytes was associated with lower uptake of common dietary LCFA (palmitic acid as well as its fluorescent derivative BODIPY FL C(16)), reduced level of total unesterified LCFA, and real-time redistribution of BODIPY FL C(16) from the central nucleoplasm to the nuclear envelope. Taken together, these studies support the hypothesis that L-FABP may facilitate ligand (LCFA)-activated PPARalpha transcriptional activity at least in part by increasing total LCFA ligand available to PPARalpha for inducing PPARalpha-mediated transcription of proteins involved in LCFA metabolism.


Subject(s)
Fatty Acid-Binding Proteins/physiology , Hepatocytes/metabolism , PPAR alpha/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Fatty Acid-Binding Proteins/genetics , Immunoprecipitation , Ligands , Male , Mice , Mice, Knockout , Microscopy, Confocal
SELECTION OF CITATIONS
SEARCH DETAIL