Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Microvasc Res ; 122: 34-40, 2019 03.
Article in English | MEDLINE | ID: mdl-30439484

ABSTRACT

OBJECTIVE: Freshly isolated endothelial cells from both conduit arteries and microvasculature were used to test the hypothesis that eNOS protein content and nitric oxide production in coronary endothelial cells increases with vessel radius. METHODS: Porcine hearts were obtained from a local abattoir. Large and small arteries as well as arterioles were dissected free of myocardium and homogenized as whole vessels. Additionally, endothelial cells were isolated from both conduit arteries and left ventricular myocardium by tissue digestion with collagenase, followed by endothelial cell isolation using biotinylated-anti-CD31 and streptavidin-coated paramagnetic beads. Purity of isolated endothelial cells was confirmed by immunofluorescence and immunoblot. RESULTS: In whole vessel lysate, immunoblot analysis revealed that protein content for eNOS was greater in arterioles compared to small and large arteries. Nitric oxide metabolites (nitrite plus nitrate; NOx) levels measured from whole vessel lysate decreased as vessel size increased, with both arterioles and small arteries displaying significantly greater NOx content than conduit. Consistent with our hypothesis, both eNOS protein level and NOx were significantly greater in endothelial cells isolated from conduit arteries compared with those from coronary microvasculature. Furthermore, confocal microscopy revealed that eNOS protein was present in all conduit and microvascular endothelial cells, although eNOS staining was less intense in microvascular cells than those of conduit artery. CONCLUSIONS: These findings demonstrate increased eNOS protein and NOx content in endothelial cells of conduit arteries compared with the microcirculation and underscore the importance of comparing endothelial-specific molecules in freshly isolated endothelial cells, rather than whole lysate of different sized vessels.


Subject(s)
Arterioles/enzymology , Coronary Vessels/enzymology , Endothelial Cells/enzymology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Animals , Arterioles/cytology , Coronary Vessels/cytology , Nitrates/metabolism , Nitrites/metabolism , Sus scrofa
2.
J Lipid Res ; 59(4): 646-657, 2018 04.
Article in English | MEDLINE | ID: mdl-29414765

ABSTRACT

Phytocannabinoids, such as Δ9-tetrahydrocannabinol (THC), bind and activate cannabinoid (CB) receptors, thereby "piggy-backing" on the same pathway's endogenous endocannabinoids (ECs). The recent discovery that liver fatty acid binding protein-1 (FABP1) is the major cytosolic "chaperone" protein with high affinity for both Δ9-THC and ECs suggests that Δ9-THC may alter hepatic EC levels. Therefore, the impact of Δ9-THC or EC treatment on the levels of endogenous ECs, such as N-arachidonoylethanolamide (AEA) and 2-arachidonoylglycerol (2-AG), was examined in cultured primary mouse hepatocytes from WT and Fabp1 gene-ablated (LKO) mice. Δ9-THC alone or 2-AG alone significantly increased AEA and especially 2-AG levels in WT hepatocytes. LKO alone markedly increased AEA and 2-AG levels. However, LKO blocked/diminished the ability of Δ9-THC to further increase both AEA and 2-AG. In contrast, LKO potentiated the ability of exogenous 2-AG to increase the hepatocyte level of AEA and 2-AG. These and other data suggest that Δ9-THC increases hepatocyte EC levels, at least in part, by upregulating endogenous AEA and 2-AG levels. This may arise from Δ9-THC competing with AEA and 2-AG binding to FABP1, thereby decreasing targeting of bound AEA and 2-AG to the degradative enzymes, fatty acid amide hydrolase and monoacylglyceride lipase, to decrease hydrolysis within hepatocytes.


Subject(s)
Dronabinol/adverse effects , Endocannabinoids/metabolism , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Animals , Dronabinol/pharmacology , Fatty Acid-Binding Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Biochemistry ; 57(41): 6027-6042, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30232874

ABSTRACT

Although serum Δ9-tetrahydrocannabinol (Δ9-THC) undergoes rapid hepatic clearance and metabolism, almost nothing is known regarding the mechanism(s) whereby this highly lipophilic phytocannabinoid is transported for metabolism/excretion. A novel NBD-arachidonoylethanolamide (NBD-AEA) fluorescence displacement assay showed that liver fatty acid binding protein (FABP1), the major hepatic endocannabinoid (EC) binding protein, binds the first major metabolite of Δ9-THC (Δ9-THC-OH) as well as Δ9-THC itself. Circular dichroism (CD) confirmed that not only Δ9-THC and Δ9-THC-OH but also downstream metabolites Δ9-THC-COOH and Δ9-THC-CO-glucuronide directly interact with FABP1. Δ9-THC and metabolite interaction differentially altered the FABP1 secondary structure, increasing total α-helix (all), decreasing total ß-sheet (Δ9-THC-COOH, Δ9-THC-CO-glucuronide), increasing turns (Δ9-THC-OH, Δ9-THC-COOH, Δ9-THC-CO-glucuronide), and decreasing unordered structure (Δ9-THC, Δ9-THC-OH). Cultured primary hepatocytes from wild-type (WT) mice took up and converted Δ9-THC to the above metabolites. Fabp1 gene ablation (LKO) dramatically increased hepatocyte accumulation of Δ9-THC and even more so its primary metabolites Δ9-THC-OH and Δ9-THC-COOH. Concomitantly, rtPCR and Western blotting indicated that LKO significantly increased Δ9-THC's ability to regulate downstream nuclear receptor transcription of genes important in both EC ( Napepld > Daglb > Dagla, Naaa, Cnr1) and lipid ( Cpt1A > Fasn, FATP4) metabolism. Taken together, the data indicated that FABP1 may play important roles in Δ9-THC uptake and elimination as well as Δ9-THC induction of genes regulating hepatic EC levels and downstream targets in lipid metabolism.


Subject(s)
Dronabinol , Fatty Acid-Binding Proteins , Hepatocytes/metabolism , Lipid Metabolism/drug effects , Animals , Circular Dichroism , Dronabinol/pharmacokinetics , Dronabinol/pharmacology , Fatty Acid-Binding Proteins/chemistry , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Humans , Mice , Mice, Knockout , Protein Structure, Secondary
4.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(3): 323-338, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29307784

ABSTRACT

Although singly ablating Fabp1 or Scp2/Scpx genes may exacerbate the impact of high fat diet (HFD) on whole body phenotype and non-alcoholic fatty liver disease (NAFLD), concomitant upregulation of the non-ablated gene, preference for ad libitum fed HFD, and sex differences complicate interpretation. Therefore, these issues were addressed in male and female mice ablated in both genes (Fabp1/Scp2/Scpx null or TKO) and pair-fed HFD. Wild-type (WT) males gained more body weight as fat tissue mass (FTM) and exhibited higher hepatic lipid accumulation than WT females. The greater hepatic lipid accumulation in WT males was associated with higher hepatic expression of enzymes in glyceride synthesis, higher hepatic bile acids, and upregulation of transporters involved in hepatic reuptake of serum bile acids. While TKO had little effect on whole body phenotype and hepatic bile acid accumulation in either sex, TKO increased hepatic accumulation of lipids in both, specifically phospholipid and cholesteryl esters in males and females and free cholesterol in females. TKO-induced increases in glycerides were attributed not only to complete loss of FABP1, SCP2 and SCPx, but also in part to sex-dependent upregulation of hepatic lipogenic enzymes. These data with WT and TKO mice pair-fed HFD indicate that: i) Sex significantly impacted the ability of HFD to increase body weight, induce hepatic lipid accumulation and increase hepatic bile acids; and ii) TKO exacerbated the HFD ability to induce hepatic lipid accumulation, regardless of sex, but did not significantly alter whole body phenotype in either sex.


Subject(s)
Carrier Proteins/metabolism , Cholesterol/metabolism , Dietary Fats/adverse effects , Fatty Acid-Binding Proteins/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Phospholipids/metabolism , Animals , Carrier Proteins/genetics , Cholesterol/genetics , Dietary Fats/pharmacology , Fatty Acid-Binding Proteins/genetics , Female , Liver/pathology , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Phospholipids/genetics
5.
Arch Biochem Biophys ; 650: 93-102, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29763591

ABSTRACT

Dysregulation of the hepatic endocannabinoid (EC) system and high fat diet (HFD) are associated with non-alcoholic fatty liver disease. Liver cytosol contains high levels of two novel endocannabinoid binding proteins-liver fatty acid binding protein (FABP1) and sterol carrier protein-2 (SCP-2). While Fabp1 gene ablation significantly increases hepatic levels of arachidonic acid (ARA)-containing EC and sex-dependent response to pair-fed high fat diet (HFD), the presence of SCP-2 complicates interpretation. These issues were addressed by ablating Scp-2/Scp-x in Fabp1 null mice (TKO). In control-fed mice, TKO increased hepatic levels of arachidonoylethanolamide (AEA) in both sexes. HFD impacted hepatic EC levels by decreasing AEA in TKO females and decreasing 2-arachidonoyl glycerol (2-AG) in WT of both sexes. Only TKO males on HFD had increased hepatic 2-AG levels. Hepatic ARA levels were decreased in control-fed TKO of both sexes. Changes in hepatic AEA/2-AG levels were not associated with altered amounts of hepatic proteins involved in AEA/2-AG synthesis or degradation. These findings suggested that ablation of the Scp-2/Scp-x gene in Fabp1 null mice exacerbated hepatic EC accumulation and antagonized the impact of HFD on hepatic EC levels-suggesting both proteins play important roles in regulating the hepatic EC system.


Subject(s)
Carrier Proteins/genetics , Diet, High-Fat , Dietary Fats/metabolism , Endocannabinoids/metabolism , Fatty Acid-Binding Proteins/genetics , Liver/metabolism , Animals , Carrier Proteins/metabolism , Fatty Acid-Binding Proteins/metabolism , Female , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Biochemistry ; 56(38): 5147-5159, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28853554

ABSTRACT

Using recombinant human wild-type fatty acid binding protein 1 (WT FABP1 T94T) and a variant (FABP1 T94A) protein, fluorescence binding assays, and circular dichroism, it was shown for the first time that WT FABP1 and the T94A variant each have a single, relatively hydrophobic site for binding fluorescent NBD-labeled analogues of N-arachidonoylethanolamide and 2-arachidonoylglycerol with high affinity. Most native N-acylethanolamides (NAEs) but only one 2-monoacylglycerol [i.e., 2-arachidonoylglycerol (2-AG)] displaced WT FABP1-bound fluorescently labeled endocannabinoids (ECs). While the T94A variant did not differ in affinity for AEA and most other NAEs, it exhibited a modestly higher affinity for OEA, as well as a higher affinity for 2-AG. Binding of AEA and 2-AG altered WT FABP1's secondary structure more extensively than any other previously examined ligand did. The T94A variant without a ligand was more susceptible to temperature-induced unfolding. While the T94A variant was much less sensitive to ligand (i.e., AEA or 2-AG)-induced conformational change, nevertheless binding of AEA and 2-AG significantly stabilized the T94A structure to thermal unfolding. These data provide the first evidence that ECs not only bind to but also alter the secondary structure of the human FABP1, with the latter markedly impacted by the T94A substitution, a variant strongly associated with hepatic accumulation of lipids and non-alcoholic fatty liver disease (NAFLD). Importantly, NAFLD has been associated with elevated hepatic levels of ECs and FABP1.


Subject(s)
Endocannabinoids/metabolism , Fatty Acid-Binding Proteins/chemistry , Fatty Acid-Binding Proteins/metabolism , Animals , Arachidonic Acids/metabolism , Binding Sites , Circular Dichroism , Endocannabinoids/chemistry , Fatty Acid-Binding Proteins/genetics , Fluorescence , Glycerides/metabolism , Humans , Polyunsaturated Alkamides/metabolism , Protein Structure, Secondary , Rats , Temperature
7.
J Lipid Res ; 58(6): 1153-1165, 2017 06.
Article in English | MEDLINE | ID: mdl-28411199

ABSTRACT

Studies in vitro have suggested that both sterol carrier protein-2/sterol carrier protein-x (Scp-2/Scp-x) and liver fatty acid binding protein [Fabp1 (L-FABP)] gene products facilitate hepatic uptake and metabolism of lipotoxic dietary phytol. However, interpretation of physiological function in mice singly gene ablated in the Scp-2/Scp-x has been complicated by concomitant upregulation of FABP1. The work presented herein provides several novel insights: i) An 8-anilino-1-naphthalenesulfonic acid displacement assay showed that neither SCP-2 nor L-FABP bound phytol, but both had high affinity for its metabolite, phytanic acid; ii) GC-MS studies with phytol-fed WT and Fabp1/Scp-2/SCP-x gene ablated [triple KO (TKO)] mice showed that TKO exacerbated hepatic accumulation of phytol metabolites in vivo in females and less so in males. Concomitantly, dietary phytol increased hepatic levels of total long-chain fatty acids (LCFAs) in both male and female WT and TKO mice. Moreover, in both WT and TKO female mice, dietary phytol increased hepatic ratios of saturated/unsaturated and polyunsaturated/monounsaturated LCFAs, while decreasing the peroxidizability index. However, in male mice, dietary phytol selectively increased the saturated/unsaturated ratio only in TKO mice, while decreasing the peroxidizability index in both WT and TKO mice. These findings suggested that: 1) SCP-2 and FABP1 both facilitated phytol metabolism after its conversion to phytanic acid; and 2) SCP-2/SCP-x had a greater impact on hepatic phytol metabolism than FABP1.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Gene Knockout Techniques , Liver/metabolism , Phytol/metabolism , Animals , Endoplasmic Reticulum/metabolism , Female , Male , Mice , Peroxisomes/metabolism , Phytanic Acid/metabolism , Substrate Specificity
8.
J Lipid Res ; 58(11): 2114-2126, 2017 11.
Article in English | MEDLINE | ID: mdl-28972119

ABSTRACT

Upregulation of the hepatic endocannabinoid (EC) receptor [cannabinoid receptor-1 (CB1)] and arachidonoylethanolamide (AEA) is associated with nonalcoholic fatty liver disease (NAFLD). Male mice fed high-fat diet (HFD) ad libitum also exhibit NAFLD, increased hepatic AEA, and obesity. But, preference for HFD complicates interpretation and almost nothing is known about these effects in females. These issues were addressed by pair-feeding HFD. Similarly to ad libitum-fed HFD, pair-fed HFD also increased WT male and female mouse fat tissue mass (FTM), but preferentially at the expense of lean tissue mass. In contrast, pair-fed HFD did not elicit NAFLD in WT mice regardless of sex. Concomitantly, pair-fed HFD oppositely impacted hepatic AEA, 2-arachidonoyl glycerol, and/or CB1 in WT males versus females. In pair-fed HFD mice, liver FA binding protein-1 (Fabp1) gene ablation (LKO): i) exacerbated FTM in both sexes; ii) did not elicit liver neutral lipid accumulation in males and only slightly in females; iii) increased liver AEA in males, but decreased it in females; and iv) decreased CB1 only in males. Thus, pair-fed HFD selectively impacted hepatic ECs more in females, but did not elicit NAFLD in either sex. These effects were modified by LKO consistent with FABP1's ability to impact EC and FA metabolism.


Subject(s)
Diet, High-Fat/adverse effects , Endocannabinoids/metabolism , Fatty Acid-Binding Proteins/deficiency , Fatty Acid-Binding Proteins/genetics , Gene Knockout Techniques , Liver/drug effects , Liver/metabolism , Animals , Biological Transport/drug effects , Biological Transport/genetics , Biomarkers/blood , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytosol/drug effects , Cytosol/metabolism , Fatty Acids/metabolism , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Lipogenesis/drug effects , Lipogenesis/genetics , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Organ Size/genetics , Phenotype , Sterol Regulatory Element Binding Protein 1/genetics
9.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(3): 291-304, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27940000

ABSTRACT

In vitro studies suggest that liver fatty acid binding protein (L-FABP) and sterol carrier protein-2/sterol carrier protein-x (SCP2/SCPx) gene products facilitate uptake and metabolism and detoxification of dietary-derived phytol in mammals. However, concomitant upregulation of L-FABP in SCP2/SCPx null mice complicates interpretation of their physiological phenotype. Therefore, the impact of ablating both the L-FABP gene and SCP2/SCPx gene (L-FABP/SCP2/SCPx null or TKO) was examined in phytol-fed female wild-type (WT) and TKO mice. TKO increased hepatic total lipid accumulation, primarily phospholipid, by mechanisms involving increased hepatic levels of proteins in the phospholipid synthetic pathway. Concomitantly, TKO reduced expression of proteins in targeting fatty acids towards the triacylglycerol synthetic pathway. Increased hepatic lipid accumulation was not associated with any concomitant upregulation of membrane fatty acid transport/translocase proteins involved in fatty acid uptake (FATP2, FATP4, FATP5 or GOT) or cytosolic proteins involved in fatty acid intracellular targeting (ACBP). In addition, TKO exacerbated dietary phytol-induced whole body weight loss, especially lean tissue mass. Since individually ablating SCPx or SCP2/SCPx elicited concomitant upregulation of L-FABP, these findings with TKO mice help to resolve the contributions of SCP2/SCPx gene ablation on dietary phytol-induced whole body and hepatic lipid phenotype independent of concomitant upregulation of L-FABP.


Subject(s)
Carrier Proteins/metabolism , Fatty Acid-Binding Proteins/metabolism , Lipid Metabolism/drug effects , Phytol/administration & dosage , Animals , Diet/methods , Fatty Acids/metabolism , Female , Lipids/physiology , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phospholipids/metabolism , Triglycerides/metabolism , Up-Regulation/drug effects
10.
Arch Biochem Biophys ; 635: 17-26, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29051070

ABSTRACT

While prior studies focusing on male mice suggest a role for sterol carrier protein-2/sterol carrier protein-x (SCP-2/SCP-x; DKO) on hepatic phytol metabolism, its role in females is unresolved. This issue was addressed using female and male wild-type (WT) and DKO mice fed a phytoestrogen-free diet without or with 0.5% phytol. GC/MS showed that hepatic: i) phytol was absent and its branched-chain fatty acid (BCFA) metabolites were barely detectable in WT control-fed mice; ii) accumulation of phytol as well as its peroxisomal metabolite BCFAs (phytanic acid ¼ pristanic and 2,3-pristenic acids) was increased by dietary phytol in WT females, but only slightly in WT males; iii) accumulation of phytol and BCFA was further increased by DKO in phytol-fed females, but much more markedly in males. Livers of phytol-fed WT female mice as well as phytol-fed DKO female and male mice also accumulated increased proportion of saturated straight-chain fatty acids (LCFA) at the expense of unsaturated LCFA. Liver phytol accumulation was not due to increased SCP-2 binding/transport of phytol since SCP-2 bound phytanic acid, but not its precursor phytol. Thus, the loss of Scp-2/Scp-x contributed to a sex-dependent hepatic accumulation of dietary phytol and BCFA.


Subject(s)
Carrier Proteins/metabolism , Lipid Metabolism/physiology , Liver/metabolism , Phytanic Acid/analogs & derivatives , Phytol/pharmacokinetics , Administration, Oral , Animals , Carrier Proteins/genetics , Female , Gene Silencing , Male , Metabolic Clearance Rate , Mice , Mice, Inbred C57BL , Mice, Knockout , Phytanic Acid/metabolism , Phytol/administration & dosage , Sex Factors
11.
Biochemistry ; 55(37): 5243-55, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27552286

ABSTRACT

Endocannabinoids (ECs) and cannabinoids are very lipophilic molecules requiring the presence of cytosolic binding proteins that chaperone these molecules to intracellular targets. While three different fatty acid binding proteins (FABP3, -5, and -7) serve this function in brain, relatively little is known about how such hydrophobic ECs and cannabinoids are transported within the liver. The most prominent hepatic FABP, liver fatty acid binding protein (FABP1 or L-FABP), has high affinity for arachidonic acid (ARA) and ARA-CoA, suggesting that FABP1 may also bind ARA-derived ECs (AEA and 2-AG). Indeed, FABP1 bound ECs with high affinity as shown by displacement of FABP1-bound fluorescent ligands and by quenching of FABP1 intrinsic tyrosine fluorescence. FABP1 also had high affinity for most non-ARA-containing ECs, FABP1 inhibitors, EC uptake/hydrolysis inhibitors, and phytocannabinoids and less so for synthetic cannabinoid receptor (CBR) agonists and antagonists. The physiological impact was examined with liver from wild-type (WT) versus FABP1 gene-ablated (LKO) male mice. As shown by liquid chromatography and mass spectrometry, FABP1 gene ablation significantly increased hepatic levels of AEA, 2-AG, and 2-OG. These increases were not due to increased protein levels of EC synthetic enzymes (NAPEPLD and DAGL) or a decreased level of EC degradative enzyme (FAAH) but correlated with complete loss of FABP1, a decreased level of SCP2 (8-fold less prevalent than FABP1, but also binds ECs), and a decreased level of degradative enzymes (NAAA and MAGL). These data indicated that FABP1 not only is the most prominent endocannabinoid and cannabinoid binding protein but also impacts hepatic endocannabinoid levels.


Subject(s)
Endocannabinoids/metabolism , Fatty Acid-Binding Proteins/metabolism , Receptors, Cannabinoid/metabolism , Animals , Female , Fluorescent Dyes , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL
12.
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
13.
Am J Physiol Gastrointest Liver Physiol ; 309(5): G387-99, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26113298

ABSTRACT

While a high-cholesterol diet induces hepatic steatosis, the role of intracellular sterol carrier protein-2/sterol carrier protein-x (SCP-2/SCP-x) proteins is unknown. We hypothesized that ablating SCP-2/SCP-x [double knockout (DKO)] would impact hepatic lipids (cholesterol and cholesteryl ester), especially in high-cholesterol-fed mice. DKO did not alter food consumption, and body weight (BW) gain decreased especially in females, concomitant with hepatic steatosis in females and less so in males. DKO-induced steatosis in control-fed wild-type (WT) mice was associated with 1) loss of SCP-2; 2) upregulation of liver fatty acid binding protein (L-FABP); 3) increased mRNA and/or protein levels of sterol regulatory element binding proteins (SREBP1 and SREBP2) as well as increased expression of target genes of cholesterol synthesis (Hmgcs1 and Hmgcr) and fatty acid synthesis (Acc1 and Fas); and 4) cholesteryl ester accumulation was also associated with increased acyl-CoA cholesterol acyltransferase-2 (ACAT2) in males. DKO exacerbated the high-cholesterol diet-induced hepatic cholesterol and glyceride accumulation, without further increasing SREBP1, SREBP2, or target genes. This exacerbation was associated both with loss of SCP-2 and concomitant downregulation of Ceh/Hsl, apolipoprotein B (ApoB), MTP, and/or L-FABP protein expression. DKO diminished the ability to secrete excess cholesterol into bile and oxidize cholesterol to bile acid for biliary excretion, especially in females. This suggested that SCP-2/SCP-x affects cholesterol transport to particular intracellular compartments, with ablation resulting in less to the endoplasmic reticulum for SREBP regulation, making more available for cholesteryl ester synthesis, for cholesteryl-ester storage in lipid droplets, and for bile salt synthesis and/or secretion. These alterations are significant findings, since they affect key processes in regulation of sterol metabolism.


Subject(s)
Carrier Proteins/metabolism , Cholesterol, Dietary/pharmacology , Lipid Metabolism , Liver/metabolism , Acetyltransferases/genetics , Acetyltransferases/metabolism , Animals , Apolipoprotein B-100 , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Carrier Proteins/genetics , Cholesterol, Dietary/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Female , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , fas Receptor/genetics , fas Receptor/metabolism , Sterol O-Acyltransferase 2
14.
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
15.
Biochim Biophys Acta ; 1831(8): 1412-25, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23747828

ABSTRACT

Although liver fatty acid binding protein (L-FABP) binds fibrates and PPARα in vitro and enhances fibrate induction of PPARα in transformed cells, the functional significance of these findings is unclear, especially in normal hepatocytes. Studies with cultured primary mouse hepatocytes show that: 1) At physiological (6mM) glucose, fibrates (bezafibrate, fenofibrate) only weakly activated PPARα transcription of genes in LCFA ß-oxidation; 2) High (11-20mM) glucose, but not maltose (osmotic control), significantly potentiated fibrate-induction of mRNA of these and other PPARα target genes to increase LCFA ß-oxidation. These effects were associated with fibrate-mediated redistribution of L-FABP into nuclei-an effect prolonged by high glucose-but not with increased de novo fatty acid synthesis from glucose; 3) Potentiation of bezafibrate action by high glucose required an intact L-FABP/PPARα signaling pathway as shown with L-FABP null, PPARα null, PPARα inhibitor-treated WT, or PPARα-specific fenofibrate-treated WT hepatocytes. High glucose alone in the absence of fibrate was ineffective. Thus, high glucose potentiation of PPARα occurred through FABP/PPARα rather than indirectly through other PPARs or glucose induced signaling pathways. These data indicated L-FABP's importance in fibrate-induction of hepatic PPARα LCFA ß-oxidative genes, especially in the context of high glucose levels.


Subject(s)
Bezafibrate/pharmacology , Fatty Acid-Binding Proteins/metabolism , Fenofibrate/pharmacology , Glucose/pharmacology , Hepatocytes/metabolism , Hypolipidemic Agents/pharmacology , PPAR alpha/metabolism , Sweetening Agents/pharmacology , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acids/genetics , Fatty Acids/metabolism , Hepatocytes/cytology , Mice , Mice, Mutant Strains , Oxidation-Reduction/drug effects , PPAR alpha/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
16.
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
17.
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
18.
Am J Physiol Gastrointest Liver Physiol ; 304(3): G241-56, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23238934

ABSTRACT

Liver fatty acid binding protein (L-FABP) is the major soluble protein that binds very-long-chain n-3 polyunsaturated fatty acids (n-3 PUFAs) in hepatocytes. However, nothing is known about L-FABP's role in n-3 PUFA-mediated peroxisome proliferator activated receptor-α (PPARα) transcription of proteins involved in long-chain fatty acid (LCFA) ß-oxidation. This issue was addressed in cultured primary hepatocytes from wild-type, L-FABP-null, and PPARα-null mice with these major findings: 1) PUFA-mediated increase in the expression of PPARα-regulated LCFA ß-oxidative enzymes, LCFA/LCFA-CoA binding proteins (L-FABP, ACBP), and PPARα itself was L-FABP dependent; 2) PPARα transcription, robustly potentiated by high glucose but not maltose, a sugar not taken up, correlated with higher protein levels of these LCFA ß-oxidative enzymes and with increased LCFA ß-oxidation; and 3) high glucose altered the potency of n-3 relative to n-6 PUFA. This was not due to a direct effect of glucose on PPARα transcriptional activity nor indirectly through de novo fatty acid synthesis from glucose. Synergism was also not due to glucose impacting other signaling pathways, since it was observed only in hepatocytes expressing both L-FABP and PPARα. Ablation of L-FABP or PPARα as well as treatment with MK886 (PPARα inhibitor) abolished/reduced PUFA-mediated PPARα transcription of these genes, especially at high glucose. Finally, the PUFA-enhanced L-FABP distribution into nuclei with high glucose augmentation of the L-FABP/PPARα interaction reveals not only the importance of L-FABP for PUFA induction of PPARα target genes in fatty acid ß-oxidation but also the significance of a high glucose enhancement effect in diabetes.


Subject(s)
Fatty Acid-Binding Proteins/pharmacology , Fatty Acids, Unsaturated/pharmacology , Glucose/pharmacology , PPAR alpha/biosynthesis , Acyl-CoA Oxidase/metabolism , Animals , Blotting, Western , Carnitine O-Palmitoyltransferase/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Fatty Acids/biosynthesis , Fatty Acids/metabolism , Glucose/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Maltose/pharmacology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Osmolar Concentration , PPAR alpha/genetics , Real-Time Polymerase Chain Reaction , Stearic Acids/metabolism , Transcription, Genetic/drug effects
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL