Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nutrients ; 15(12)2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37375658

ABSTRACT

Although vegan diets have been reported to be associated with a reduced risk of cardiovascular disease, it was not known whether this might be partly due to vegan diets' effects on plasma triglyceride metabolism. This study aimed to investigate if there are differences in the activity of lipoprotein lipase (LPL), an enzyme that functions at the vascular endothelium and is responsible for triglyceride breakdown, in sera obtained from vegans and omnivores. LPL activity was assessed using isothermal titration calorimetry, which allows measurements in undiluted serum samples, mimicking physiological conditions. Fasted sera from 31 healthy participants (12F 2M vegans, 11F 6M omnivores) were analyzed. The results indicated no significant differences in average LPL activity between the vegan and omnivore groups. Interestingly, despite similar triglyceride levels, there were considerable variations in LPL activity and total very-low-density lipoprotein triglyceride breakdowns between individuals within both groups. Biomarker analysis showed that vegans had lower total cholesterol and LDL-C levels compared to omnivores. These findings suggest that the lipid-related benefits of a vegan diet, in terms of atherogenic risk, may primarily stem from cholesterol reduction rather than affecting serum as a medium for LPL-mediated triglyceride breakdown. In healthy individuals, lipid-related changes in serum composition in response to a vegan diet are likely overshadowed by genetic or other lifestyle factors.


Subject(s)
Diet, Vegan , Vegans , Humans , Lipoprotein Lipase , Triglycerides , Lipolysis , Diet, Vegetarian , Diet
2.
PLoS One ; 18(4): e0283358, 2023.
Article in English | MEDLINE | ID: mdl-37043509

ABSTRACT

Lipoprotein lipase (LPL), a crucial enzyme in the intravascular hydrolysis of triglyceride-rich lipoproteins, is a potential drug target for the treatment of hypertriglyceridemia. The activity and stability of LPL are influenced by a complex ligand network. Previous studies performed in dilute solutions suggest that LPL can appear in various oligomeric states. However, it was not known how the physiological environment, that is blood plasma, affects the action of LPL. In the current study, we demonstrate that albumin, the major protein component in blood plasma, has a significant impact on LPL stability, oligomerization, and ligand interactions. The effects induced by albumin could not solely be reproduced by the macromolecular crowding effect. Stabilization, isothermal titration calorimetry, and surface plasmon resonance studies revealed that albumin binds to LPL with affinity sufficient to form a complex in both the interstitial space and the capillaries. Negative stain transmission electron microscopy and raster image correlation spectroscopy showed that albumin, like heparin, induced reversible oligomerization of LPL. However, the albumin induced oligomers were structurally different from heparin-induced filament-like LPL oligomers. An intriguing observation was that no oligomers of either type were formed in the simultaneous presence of albumin and heparin. Our data also suggested that the oligomer formation protected LPL from the inactivation by its physiological regulator angiopoietin-like protein 4. The concentration of LPL and its environment could influence whether LPL follows irreversible inactivation and aggregation or reversible LPL oligomer formation, which might affect interactions with various ligands and drugs. In conclusion, the interplay between albumin and heparin could provide a mechanism for ensuring the dissociation of heparan sulfate-bound LPL oligomers into active LPL upon secretion into the interstitial space.


Subject(s)
Heparin , Lipoprotein Lipase , Lipoprotein Lipase/metabolism , Heparin/pharmacology , Heparin/chemistry , Ligands , Triglycerides , Hydrolysis , Angiopoietin-Like Protein 4 , Albumins
3.
Cell Metab ; 34(11): 1719-1731.e5, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36220067

ABSTRACT

Recombinant human leptin (metreleptin) reduces hepatic lipid content in patients with lipodystrophy and overweight patients with non-alcoholic fatty liver disease and relative hypoleptinemia independent of its anorexic action. In rodents, leptin signaling in the brain increases very-low-density lipoprotein triglyceride (VLDL-TG) secretion and reduces hepatic lipid content via the vagus nerve. In this randomized, placebo-controlled crossover trial (EudraCT Nr. 2017-003014-22), we tested whether a comparable mechanism regulates hepatic lipid metabolism in humans. A single metreleptin injection stimulated hepatic VLDL-TG secretion (primary outcome) and reduced hepatic lipid content in fasted, lean men (n = 13, age range 20-38 years) but failed to do so in metabolically healthy liver transplant recipients (n = 9, age range 26-62 years) who represent a model for hepatic denervation. In an independent cohort of lean men (n = 10, age range 23-31 years), vagal stimulation by modified sham feeding replicated the effects of metreleptin on VLDL-TG secretion. Therefore, we propose that leptin has anti-steatotic properties that are independent of food intake by stimulating hepatic VLDL-TG export via a brain-vagus-liver axis.


Subject(s)
Leptin , Non-alcoholic Fatty Liver Disease , Male , Humans , Young Adult , Adult , Leptin/pharmacology , Leptin/metabolism , Lipoproteins, VLDL/metabolism , Triglycerides/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Vagus Nerve/metabolism
4.
Biochem Biophys Res Commun ; 534: 457-460, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33276951

ABSTRACT

Spike and nucleocapsid proteins of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2-SP, SARS-CoV-2-NP) are the main immunogenic targets for antibodies. We herein demonstrate that the glycosylation of SARS-CoV-2-NP masks some of its antibody epitopes. In many cases, this can lead to false-negative serological tests. Deglycosylation of SARS-CoV-2-NP significantly increased the number of positive tests. The glycosylation pattern analysis of this protein revealed that the putative N-linked glycosylation sites, at the amino acid positions 48 and 270, co-located with two of the main immunodominant B cell epitopes.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Epitopes/immunology , Recombinant Proteins/immunology , Spike Glycoprotein, Coronavirus/immunology , Animals , CHO Cells , Cricetinae , Cricetulus , Epitopes/genetics , Epitopes/metabolism , Escherichia coli/genetics , Glycosylation , Humans , Recombinant Proteins/metabolism , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism
5.
Article in English | MEDLINE | ID: mdl-31676442

ABSTRACT

Pancreatic lipase (PNLIP) is a digestive enzyme that is a potential drug target for the treatment of obesity. A better understanding of its regulation mechanisms would facilitate the development of new therapeutics. Recent studies indicate that intestinal lipolysis by PNLIP is reduced by Angiopoietin-like protein 4 (ANGPTL4), whose N-terminal domain (nANGPTL4) is a known inactivator of lipoprotein lipase (LPL) in blood circulation and adipocytes. To elucidate the mechanism of PNLIP inhibition by ANGPTL4, we developed a novel approach, using isothermal titration calorimetry (ITC). The obtained results were compared with those of well-described inhibitors of PNLIP - ε-polylysine (EPL), (-)-epigallocatechin-3-gallate (EGCG) and tetrahydrolipstatin. We demonstrate that ITC allows to investigate PNLIP inhibition mechanisms in complex substrate emulsions and that the ITC-based assay is highly sensitive - the lowest concentration for quantification of PNLIP is 1.5 pM. Combining ITC with surface plasmon resonance and fluorescence measurements, we present evidence that ANGPTL4 is a lipid-binding protein that influences PNLIP activity through interactions with components of substrate emulsions (bile salts, phospholipids and triglycerides), and this promotes the aggregation of triglyceride emulsions similarly to the PNLIP inhibitors EPL and EGCG. In the absence of substrate emulsion, unlike in the case of LPL, ANGPTL4 did not induce the inactivation of PNLIP. Our data also prove that due to various interactions with components of substrate systems, the effect of a PNLIP inhibitor depends on whether its effect is measured in a complex substrate emulsion or in a simple substrate system.


Subject(s)
Angiopoietin-Like Protein 4/pharmacology , Anti-Obesity Agents/pharmacology , Calorimetry , Enzyme Assays/methods , Lipase/antagonists & inhibitors , Angiopoietin-Like Protein 4/therapeutic use , Anti-Obesity Agents/therapeutic use , Catechin/analogs & derivatives , Catechin/pharmacology , Drug Evaluation, Preclinical/methods , Humans , Lipase/genetics , Lipase/metabolism , Obesity/drug therapy , Obesity/metabolism , Orlistat/pharmacology , Polylysine/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
6.
Biochem Biophys Res Commun ; 519(1): 67-72, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31477272

ABSTRACT

Elevated plasma triglyceride (TG) levels are associated with higher risk of atherosclerotic cardiovascular disease. One way to reduce plasma TG is to increase the activity of lipoprotein lipase (LPL), the rate limiting enzyme in plasma TG metabolism. An apolipoprotein (apo) C-II mimetic peptide (18A-CII-a) has been recently developed that stimulated LPL activity in vitro and decreased plasma TG concentration in animal models for hypertriglyceridemia. Since this peptide can serve as a new therapeutic approach for treatment of hypertriglyceridemia, we investigated how 18A-CII-a peptide influences LPL activity in human plasma. We used recently described isothermal titration calorimetry based approach to assess the peptide, which enables the analysis in nearly undiluted human plasma. The 18A-CII-a peptide was 3.5-fold more efficient in stimulating LPL activity than full-length apoC-II in plasma sample from normolipidemic individual. Furthermore, 18A-CII-a also increased LPL activity in hypertriglyceridemic plasma samples. Unlike apoC-II, high concentrations of the 18A-CII-a peptide did not inhibit LPL activity. The increase in LPL activity after addition of 18A-CII-a or apoC-II to plasma was due to the increase of the amount of available substrate for LPL. Measurements with isolated lipoproteins revealed that the relative activation effects of 18A-CII-a and apoC-II on LPL activity were greater in smaller size lipoprotein fractions, such as remnant lipoproteins, low-density lipoproteins and high-density lipoproteins. In summary, this report describes a novel mechanism of action for stimulation of LPL activity by apoC-II mimetic peptides.


Subject(s)
Apolipoprotein C-II/metabolism , Calorimetry/methods , Lipoprotein Lipase/blood , Peptides/metabolism , Animals , Cattle , Fatty Acids/metabolism , Humans , Hydrolysis , Substrate Specificity
7.
Chem Sci ; 8(3): 2184-2190, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28694954

ABSTRACT

A novel eight-membered macrocycle of the hemicucurbit[n]uril family, chiral (all-R)-cyclohexanohemicucurbit[8]uril (cycHC[8]) ‡The name cyclohexylhemicucurbituril, previously used for these macrocycles, is changed in accordance with the IUPAC nomenclature for fused cycles, as the cyclohexane substituents are fused with the parent hemicucurbituril. binds anions in a purely protic solvent with remarkable selectivity. The cycHC[8] portals open and close to fully encapsulate anions in a 1 : 1 ratio, resembling a molecular Pac-Man™. Comprehensive gas, solution and solid phase studies prove that the binding is governed by the size, shape and charge distribution of the bound anion. Gas phase studies show an order of SbF6- ≈ PF6- > ReO4- > ClO4- > SCN- > BF4- > HSO4- > CF3SO3- for anion complexation strength. An extensive crystallographic study reveals the preferred orientations of the anions within the octahedral cavity of cycHC[8] and highlights the importance of the size- and shape-matching between the anion and the receptor cavity. The solution studies show the strongest binding of the ideally fitting SbF6- anion, with an association constant of 2.5 × 105 M-1 in pure methanol. The symmetric, receptor cavity-matching charge distribution of the anions results in drastically stronger binding than in the case of anions with asymmetric charge distribution. Isothermal titration calorimetry (ITC) reveals the complexation to be exothermic and enthalpy-driven. The DFT calculations and VT-NMR studies confirmed that the complexation proceeds through a pre-complex formation while the exchange of methanol solvent with the anion is the rate-limiting step. The octameric cycHC[8] offers a unique example of template-controlled design of an electroneutral host for binding large anions in a competitive polar solvent.

8.
J Lipid Res ; 58(1): 279-288, 2017 01.
Article in English | MEDLINE | ID: mdl-27845686

ABSTRACT

LPL hydrolyzes triglycerides in plasma lipoproteins. Due to the complex regulation mechanism, it has been difficult to mimic the physiological conditions under which LPL acts in vitro. We demonstrate that isothermal titration calorimetry (ITC), using human plasma as substrate, overcomes several limitations of previously used techniques. The high sensitivity of ITC allows continuous recording of the heat released during hydrolysis. Both initial rates and kinetics for complete hydrolysis of plasma lipids can be studied. The heat rate was shown to correspond to the release of fatty acids and was linearly related to the amount of added enzyme, either purified LPL or postheparin plasma. Addition of apoC-III reduced the initial rate of hydrolysis by LPL, but the inhibition became less prominent with time when the lipoproteins were triglyceride poor. Addition of angiopoietin-like protein (ANGPTL)3 or ANGPTL4 caused reduction of the activity of LPL via a two-step mechanism. We conclude that ITC can be used for quantitative measurements of LPL activity and interactions under in vivo-like conditions, for comparisons of the properties of plasma samples from patients and control subjects as substrates for LPL, as well as for testing of drug candidates developed with the aim to affect the LPL system.


Subject(s)
Calorimetry , Fatty Acids/blood , Lipolysis/drug effects , Lipoprotein Lipase/blood , Adult , Angiopoietin-Like Protein 3 , Angiopoietin-Like Protein 4 , Angiopoietin-like Proteins , Angiopoietins/administration & dosage , Animals , Apolipoprotein C-III/administration & dosage , Cattle , Female , Healthy Volunteers , Humans , Hydrolysis , Kinetics , Lipoproteins, VLDL/blood , Male , Triglycerides/blood
9.
J Biol Chem ; 290(22): 13919-34, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25873395

ABSTRACT

GPIHBP1 is an endothelial membrane protein that transports lipoprotein lipase (LPL) from the subendothelial space to the luminal side of the capillary endothelium. Here, we provide evidence that two regions of GPIHBP1, the acidic N-terminal domain and the central Ly6 domain, interact with LPL as two distinct binding sites. This conclusion is based on comparative binding studies performed with a peptide corresponding to the N-terminal domain of GPIHBP1, the Ly6 domain of GPIHBP1, wild type GPIHBP1, and the Ly6 domain mutant GPIHBP1 Q114P. Although LPL and the N-terminal domain formed a tight but short lived complex, characterized by fast on- and off-rates, the complex between LPL and the Ly6 domain formed more slowly and persisted for a longer time. Unlike the interaction of LPL with the Ly6 domain, the interaction of LPL with the N-terminal domain was significantly weakened by salt. The Q114P mutant bound LPL similarly to the N-terminal domain of GPIHBP1. Heparin dissociated LPL from the N-terminal domain, and partially from wild type GPIHBP1, but was unable to elute the enzyme from the Ly6 domain. When LPL was in complex with the acidic peptide corresponding to the N-terminal domain of GPIHBP1, the enzyme retained its affinity for the Ly6 domain. Furthermore, LPL that was bound to the N-terminal domain interacted with lipoproteins, whereas LPL bound to the Ly6 domain did not. In summary, our data suggest that the two domains of GPIHBP1 interact independently with LPL and that the functionality of LPL depends on its localization on GPIHBP1.


Subject(s)
Glycosylphosphatidylinositols/chemistry , Lipoprotein Lipase/chemistry , Lipoproteins/chemistry , Receptors, Lipoprotein/chemistry , Animals , Anisotropy , Binding Sites , Cattle , Cross-Linking Reagents/chemistry , Endothelium, Vascular/metabolism , Epitopes/chemistry , Fluorescent Dyes/chemistry , Heparin/chemistry , Humans , Hydrogen-Ion Concentration , Mass Spectrometry , Mice , Mutation , Peptides/chemistry , Protein Binding , Protein Interaction Mapping , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Surface Plasmon Resonance
10.
Biochem Biophys Res Commun ; 450(2): 1063-9, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24984153

ABSTRACT

Patients at increased cardiovascular risk commonly display high levels of plasma triglycerides (TGs), elevated LDL cholesterol, small dense LDL particles and low levels of HDL-cholesterol. Many remain at high risk even after successful statin therapy, presumably because TG levels remain high. Lipoprotein lipase (LPL) maintains TG homeostasis in blood by hydrolysis of TG-rich lipoproteins. Efficient clearance of TGs is accompanied by increased levels of HDL-cholesterol and decreased levels of small dense LDL. Given the central role of LPL in lipid metabolism we sought to find small molecules that could increase LPL activity and serve as starting points for drug development efforts against cardiovascular disease. Using a small molecule screening approach we have identified small molecules that can protect LPL from inactivation by the controller protein angiopoietin-like protein 4 during incubations in vitro. One of the selected compounds, 50F10, was directly shown to preserve the active homodimer structure of LPL, as demonstrated by heparin-Sepharose chromatography. On injection to hypertriglyceridemic apolipoprotein A-V deficient mice the compound ameliorated the postprandial response after an olive oil gavage. This is a potential lead compound for the development of drugs that could reduce the residual risk associated with elevated plasma TGs in dyslipidemia.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , Hypolipidemic Agents/pharmacology , Lipoprotein Lipase/metabolism , Triglycerides/blood , Angiopoietin-Like Protein 4 , Angiopoietins/metabolism , Animals , Apolipoprotein A-V , Apolipoproteins/genetics , Enzyme Stability , Heterocyclic Compounds, 4 or More Rings/chemistry , Hypertriglyceridemia/blood , Hypertriglyceridemia/drug therapy , Lipoprotein Lipase/chemistry , Mice, Inbred C57BL , Mice, Knockout , Postprandial Period , Protein Binding , Protein Multimerization , Pyridines/chemistry , Pyridines/pharmacology , Small Molecule Libraries , Structure-Activity Relationship
11.
J Biol Chem ; 288(47): 33997-34008, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24121499

ABSTRACT

Apolipoproteins (apo) C-I and C-III are known to inhibit lipoprotein lipase (LPL) activity, but the molecular mechanisms for this remain obscure. We present evidence that either apoC-I or apoC-III, when bound to triglyceride-rich lipoproteins, prevent binding of LPL to the lipid/water interface. This results in decreased lipolytic activity of the enzyme. Site-directed mutagenesis revealed that hydrophobic amino acid residues centrally located in the apoC-III molecule are critical for attachment to lipid emulsion particles and consequently inhibition of LPL activity. Triglyceride-rich lipoproteins stabilize LPL and protect the enzyme from inactivating factors such as angiopoietin-like protein 4 (angptl4). The addition of either apoC-I or apoC-III to triglyceride-rich particles severely diminished their protective effect on LPL and rendered the enzyme more susceptible to inactivation by angptl4. These observations were seen using chylomicrons as well as the synthetic lipid emulsion Intralipid. In the presence of the LPL activator protein apoC-II, more of apoC-I or apoC-III was needed for displacement of LPL from the lipid/water interface. In conclusion, we show that apoC-I and apoC-III inhibit lipolysis by displacing LPL from lipid emulsion particles. We also propose a role for these apolipoproteins in the irreversible inactivation of LPL by factors such as angptl4.


Subject(s)
Apolipoprotein C-III/chemistry , Apolipoprotein C-I/chemistry , Lipoprotein Lipase/chemistry , Triglycerides/chemistry , Angiopoietin-Like Protein 4 , Angiopoietins/chemistry , Angiopoietins/genetics , Angiopoietins/metabolism , Animals , Apolipoprotein C-I/genetics , Apolipoprotein C-I/metabolism , Apolipoprotein C-III/genetics , Apolipoprotein C-III/metabolism , Cattle , Emulsions , Humans , Lipolysis/physiology , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Mutagenesis, Site-Directed , Triglycerides/genetics , Triglycerides/metabolism
12.
J Lipid Res ; 54(3): 649-661, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23307945

ABSTRACT

During the diagnosis of three unrelated patients with severe hypertriglyceridemia, three APOA5 mutations [p.(Ser232_Leu235)del, p.Leu253Pro, and p.Asp332ValfsX4] were found without evidence of concomitant LPL, APOC2, or GPIHBP1 mutations. The molecular mechanisms by which APOA5 mutations result in severe hypertriglyceridemia remain poorly understood, and the functional impairment/s induced by these specific mutations was not obvious. Therefore, we performed a thorough structural and functional analysis that included follow-up of patients and their closest relatives, measurement of apoA-V serum concentrations, and sequencing of the APOA5 gene in 200 nonhyperlipidemic controls. Further, we cloned, overexpressed, and purified both wild-type and mutant apoA-V variants and characterized their capacity to activate LPL. The interactions of recombinant wild-type and mutated apoA-V variants with liposomes of different composition, heparin, LRP1, sortilin, and SorLA/LR11 were also analyzed. Finally, to explore the possible structural consequences of these mutations, we developed a three-dimensional model of full-length, lipid-free human apoA-V. A complex, wide array of impairments was found in each of the three mutants, suggesting that the specific residues affected are critical structural determinants for apoA-V function in lipoprotein metabolism and, therefore, that these APOA5 mutations are a direct cause of hypertriglyceridemia.


Subject(s)
Apolipoproteins A/chemistry , Apolipoproteins A/metabolism , Hypertriglyceridemia/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Apolipoprotein A-V , Apolipoproteins A/genetics , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Female , Humans , Liposomes/chemistry , Liposomes/metabolism , Male , Middle Aged , Mutagenesis, Site-Directed , Mutation , Young Adult
13.
J Biol Chem ; 287(35): 29739-52, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22773878

ABSTRACT

Angiopoietin-like protein 4 (Angptl4), a potent regulator of plasma triglyceride metabolism, binds to lipoprotein lipase (LPL) through its N-terminal coiled-coil domain (ccd-Angptl4) inducing dissociation of the dimeric enzyme to inactive monomers. In this study, we demonstrate that fatty acids reduce the inactivation of LPL by Angptl4. This was the case both with ccd-Angptl4 and full-length Angptl4, and the effect was seen in human plasma or in the presence of albumin. The effect decreased in the sequence oleic acid > palmitic acid > myristic acid > linoleic acid > linolenic acid. Surface plasmon resonance, isothermal titration calorimetry, fluorescence, and chromatography measurements revealed that fatty acids bind with high affinity to ccd-Angptl4. The interactions were characterized by fast association and slow dissociation rates, indicating formation of stable complexes. The highest affinity for ccd-Angptl4 was detected for oleic acid with a subnanomolar equilibrium dissociation constant (K(d)). The K(d) values for palmitic and myristic acid were in the nanomolar range. Linoleic and linolenic acid bound with much lower affinity. On binding of fatty acids, ccd-Angptl4 underwent conformational changes resulting in a decreased helical content, weakened structural stability, dissociation of oligomers, and altered fluorescence properties of the Trp-38 residue that is located close to the putative LPL-binding region. Based on these results, we propose that fatty acids play an important role in modulating the effects of Angptl4.


Subject(s)
Angiopoietins/metabolism , Fatty Acids/metabolism , Lipoprotein Lipase/metabolism , Protein Multimerization , Angiopoietin-Like Protein 4 , Angiopoietins/chemistry , Angiopoietins/genetics , Animals , Binding Sites , Cattle , Fatty Acids/chemistry , Humans , Lipoprotein Lipase/chemistry , Lipoprotein Lipase/genetics , Mice , Plasma/chemistry , Plasma/metabolism , Protein Stability , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serum Albumin/chemistry , Serum Albumin/genetics , Serum Albumin/metabolism , Substrate Specificity , Surface Plasmon Resonance
14.
Biochim Biophys Acta ; 1821(10): 1370-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22732211

ABSTRACT

Lipoprotein lipase (LPL) is important for clearance of triacylglycerols (TG) from plasma both as an enzyme and as a bridging factor between lipoproteins and receptors for endocytosis. The amount of LPL at the luminal side of the capillary endothelium determines to what extent lipids are taken up. Mechanisms to control both the activity of LPL and its transport to the endothelial sites are regulated, but poorly understood. Angiopoietin-like proteins (ANGPTLs) 3 and 4 are potential control proteins for LPL, but plasma concentrations of ANGPTLs do not correlate with plasma TG levels. We investigated the effects of recombinant human N-terminal (NT) ANGPTLs3 and 4 on LPL-mediated bridging of TG-rich lipoproteins to primary mouse hepatocytes and found that the NT-ANGPTLs, in concentrations sufficient to cause inactivation of LPL in vitro, were unable to prevent LPL-mediated lipoprotein uptake. We therefore investigated the effects of lipoproteins (chylomicrons, VLDL and LDL) on the inactivation of LPL in vitro by NT-ANGPTLs3 and 4 and found that LPL activity was protected by TG-rich lipoproteins. In vivo, postprandial TG protected LPL from inactivation by recombinant NT-ANGPTL4 injected to mice. We conclude that lipoprotein-bound LPL is stabilized against inactivation by ANGPTLs. The levels of ANGPTLs found in blood may not be sufficient to overcome this stabilization. Therefore it is likely that the prime site of action of ANGPTLs on LPL is in subendothelial compartments where TG-rich lipoprotein concentration is lower than in blood. This could explain why the plasma levels of TG and ANGPTLs do not correlate.


Subject(s)
Angiopoietins/pharmacology , Lipoprotein Lipase/metabolism , Lipoproteins/physiology , Triglycerides/physiology , Angiopoietin-Like Protein 3 , Angiopoietin-Like Protein 4 , Angiopoietin-like Proteins , Animals , Chylomicrons/physiology , Enzyme Activation , Hepatocytes/metabolism , Humans , Lipoproteins, LDL/physiology , Lipoproteins, VLDL/physiology , Mice
15.
Biochemistry ; 51(15): 3310-20, 2012 Apr 17.
Article in English | MEDLINE | ID: mdl-22448896

ABSTRACT

Activation of some lipoxygenases (LOX) is found to be related to the selective membrane binding upon cell stimulation. In this study, a systematic analysis of the effect of the lipid composition on the membrane binding efficiency, Ca(2+) affinity, and enzymatic activity of 11R-LOX was performed. The analysis of the membrane targeting by fluorometric and surface plasmon resonance measurements in the absence of Ca(2+) showed an exclusive binding of 11R-LOX to the anionic phospholipids (phosphatidylinositol < phosphatidylglycerol ≈ phosphatidylserine) containing model membranes. The presence of Ca(2+) enhanced the rate of interaction and influenced its mode. The modulation of the activity of 11R-LOX indicated that (i) Ca(2+) binding is a prerequisite for productive membrane association, (ii) the reaction of 11R-LOX with arachidonic acid coincided with and was driven by its Ca(2+)-mediated membrane association, and (iii) phosphatidylethanolamine and anionic phospholipids had a synergistic effect on the Ca(2+) affinity, in line with a target-activated messenger affinity mechanism [Corbin, J. A., et al. (2007) Biochemistry 46, 4322-4336]. According to the mechanism proposed in this report, 11R-LOX can bind to the membranes in two different modes and the efficiency of productive membrane binding is determined by a concerted association of Ca(2+) and lipid headgroups.


Subject(s)
Lipoxygenase/chemistry , Phospholipids/chemistry , Binding Sites , Calcium/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Kinetics , Lipoxygenase/metabolism , Phospholipids/metabolism , Spectrometry, Fluorescence , Surface Plasmon Resonance
16.
Nat Med ; 17(1): 105-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21151139

ABSTRACT

Adenovirus type 37 (Ad37) is a leading cause of epidemic keratoconjunctivitis (EKC), a severe and highly contagious ocular disease. Whereas most other adenoviruses infect cells by engaging CD46 or the coxsackie and adenovirus receptor (CAR), Ad37 binds previously unknown sialic acid-containing cell surface molecules. By glycan array screening, we show here that the receptor-recognizing knob domain of the Ad37 fiber protein specifically binds a branched hexasaccharide that is present in the GD1a ganglioside and that features two terminal sialic acids. Soluble GD1a glycan and GD1a-binding antibodies efficiently prevented Ad37 virions from binding and infecting corneal cells. Unexpectedly, the receptor is constituted by one or more glycoproteins containing the GD1a glycan motif rather than the ganglioside itself, as shown by binding, infection and flow cytometry experiments. Molecular modeling, nuclear magnetic resonance and X-ray crystallography reveal that the two terminal sialic acids dock into two of three previously established sialic acid-binding sites in the trimeric Ad37 knob. Surface plasmon resonance analysis shows that the knob-GD1a glycan interaction has high affinity. Our findings therefore form a basis for the design and development of sialic acid-containing antiviral drugs for topical treatment of EKC.


Subject(s)
Adenoviridae Infections/epidemiology , G(M1) Ganglioside/analogs & derivatives , Keratoconjunctivitis/virology , Receptors, Virus/physiology , Antiviral Agents/therapeutic use , Binding Sites , Cell Membrane/virology , Crystallography, X-Ray , Epithelium, Corneal/virology , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/immunology , G(M1) Ganglioside/metabolism , G(M1) Ganglioside/physiology , Humans , Keratoconjunctivitis/drug therapy , Keratoconjunctivitis/epidemiology , Keratoconjunctivitis/immunology , Models, Molecular , Protein Binding , Sialic Acids/metabolism , Sialic Acids/therapeutic use , Surface Plasmon Resonance
17.
J Biol Chem ; 285(10): 7484-92, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20042600

ABSTRACT

Apolipoprotein CII (apoCII) is a necessary activator for lipoprotein lipase (LPL). We had identified four residues (Tyr-63, Ile-66, Asp-69, and Gln-70), presumably contained in an alpha-helix, as a potential binding site for LPL. We have now used structure prediction, mutagenesis, and functional assays to explore the functional role of the secondary structure in this part of apoCII. First, mutants were generated by replacements with proline residues to disturb the helical structure. Activation by mutant G65P was reduced by 30%, whereas mutant S54P retained activation ability. Mutants V71P and L72P should be located outside the LPL-binding site, but V71P was totally inactive, whereas activation by L72P was reduced by 65%. Insertion of alanine after Tyr-63, changing the position of the putative LPL-binding site in relation to the hydrophobic face of the alpha-helix, also severely impeded the activation ability, and a double mutant (Y63A/I66A) was completely inactive. Next, to investigate the importance of conserved hydrophobic residues in the C-terminal end of apoCII, Phe-67, Val-71, Leu-72, and Leu-75 were exchanged for polar residues. Only F67S showed dramatic loss of function. Finally, fragment 39-62, previously claimed to activate LPL, was found to be completely inactive. Our data support the view that the helical structure close to the C-terminal end of apoCII is important for activation of LPL, probably by placing residues 63, 66, 69, and 70 in an optimal steric position. The structural requirements for the hydrophobic face on the back side of this helix and further out toward the C terminus were less stringent.


Subject(s)
Apolipoprotein C-II , Lipoprotein Lipase/metabolism , Mutagenesis, Site-Directed , Protein Structure, Secondary , Amino Acid Sequence , Animals , Apolipoprotein C-II/chemistry , Apolipoprotein C-II/genetics , Apolipoprotein C-II/metabolism , Cattle , Chylomicrons/metabolism , Circular Dichroism , Enzyme Activation , Humans , Lipoprotein Lipase/genetics , Liposomes/metabolism , Models, Molecular , Molecular Sequence Data
18.
Biochemistry ; 46(12): 3896-904, 2007 Mar 27.
Article in English | MEDLINE | ID: mdl-17326667

ABSTRACT

Apolipoprotein A-V is a potent modulator of plasma triacylglycerol levels. To investigate the molecular basis for this phenomenon we explored the ability of apolipoprotein A-V, in most experiments complexed to disks of dimyristoylphosphatidylcholine, to interact with two members of the low density lipoprotein receptor family, the low density lipoprotein receptor-related protein and the mosaic type-1 receptor, SorLA. Experiments using surface plasmon resonance showed specific binding of both free and lipid-bound apolipoprotein A-V to both receptors. The binding was calcium dependent and was inhibited by the receptor associated protein, a known ligand for members of the low density lipoprotein receptor family. Preincubation with heparin decreased the receptor binding of apolipoprotein A-V, indicating that overlap exists between the recognition sites for these receptors and for heparin. A double mutant, apolipoprotein A-V (Arg210Glu/Lys211Gln), showed decreased binding to heparin and decreased ability to bind the low density lipoprotein receptor-related protein. Association of apolipoprotein A-V with the low density lipoprotein receptor-related protein or SorLA resulted in enhanced binding of human chylomicrons to receptor-covered sensor chips. Our results indicate that apolipoprotein A-V may influence plasma lipid homeostasis by enhancing receptor-mediated endocytosis of triacylglycerol-rich lipoproteins.


Subject(s)
Apolipoproteins A/chemistry , Calcium/chemistry , Heparin/chemistry , LDL-Receptor Related Proteins/chemistry , Membrane Transport Proteins/chemistry , Amino Acid Substitution , Apolipoprotein A-V , Apolipoproteins A/genetics , Apolipoproteins A/metabolism , Calcium/metabolism , Endocytosis/physiology , Heparin/metabolism , Homeostasis/genetics , Humans , LDL-Receptor Related Proteins/metabolism , Lipid Metabolism/physiology , Membrane Transport Proteins/metabolism , Plasma/chemistry , Plasma/metabolism , Protein Binding/genetics , Surface Plasmon Resonance
19.
Proc Natl Acad Sci U S A ; 103(46): 17450-5, 2006 Nov 14.
Article in English | MEDLINE | ID: mdl-17088546

ABSTRACT

Lipoprotein lipase (LPL) has a central role in lipoprotein metabolism to maintain normal lipoprotein levels in blood and, through tissue specific regulation of its activity, to determine when and in what tissues triglycerides are unloaded. Recent data indicate that angiopoietin-like protein (Angptl)-4 inhibits LPL and retards lipoprotein catabolism. We demonstrate here that the N-terminal coiled-coil domain of Angptl-4 binds transiently to LPL and that the interaction results in conversion of the enzyme from catalytically active dimers to inactive, but still folded, monomers with decreased affinity for heparin. Inactivation occurred with less than equimolar ratios of Angptl-4 to LPL, was strongly temperature-dependent, and did not consume the Angptl-4. Furthermore, we show that Angptl-4 mRNA in rat adipose tissue turns over rapidly and that changes in the Angptl-4 mRNA abundance are inversely correlated to LPL activity, both during the fed-to-fasted and fasted-to-fed transitions. We conclude that Angptl-4 is a fasting-induced controller of LPL in adipose tissue, acting extracellularly on the native conformation in an unusual fashion, like an unfolding molecular chaperone.


Subject(s)
Adipose Tissue/enzymology , Angiopoietins/metabolism , Blood Proteins/metabolism , Lipoprotein Lipase/metabolism , Angiopoietin-Like Protein 4 , Angiopoietins/chemistry , Angiopoietins/genetics , Animals , Blood Proteins/chemistry , Blood Proteins/genetics , Blood Proteins/isolation & purification , Chromatography, Affinity , Dimerization , Enzyme Activation , Gene Expression , Heparin/analogs & derivatives , Heparin/metabolism , Lipoprotein Lipase/chemistry , Mice , Models, Molecular , Protein Binding , Protein Folding , Protein Structure, Quaternary , RNA, Messenger/genetics , Rats , Surface Plasmon Resonance
20.
J Biol Chem ; 281(33): 23405-13, 2006 Aug 18.
Article in English | MEDLINE | ID: mdl-16782967

ABSTRACT

Lipoprotein lipase (LPL), which is an important enzyme in lipid metabolism, binds to heparan sulfate (HS) proteoglycans. This interaction is crucial for several aspects of LPL function, such as intracellular/extracellular transport and high capacity attachment to cell surfaces. Retention of LPL on the capillary walls, and elsewhere, via HS chains is most likely affected by the quality and quantity of HS present. Earlier studies have demonstrated that LPL interacts with highly sulfated HS and heparin oligosaccharides. Since such structures are relatively rare in endothelial HS, we have re-addressed the question of physiological ligand structures for LPL by affinity purification of end-labeled oligosaccharides originating from heparin and HS on immobilized LPL. By a combination of chemical modification and fragmentation of the bound material we identified that the bound fraction contained modestly sulfated oligosaccharides with an average sulfation of one O-sulfate per disaccharide unit and tolerates N-acetylated glucosamine residues. Therefore LPL, containing several clusters of positive charges on each subunit, may constitute an ideal structure for a protein that needs to bind with reasonable affinity to a variety of modestly sulfated sequences of the type that is abundant in HS chains.


Subject(s)
Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Lipoprotein Lipase/metabolism , Acetylation , Animals , Binding, Competitive , Carbohydrate Sequence , Cattle , Endothelium, Vascular/chemistry , Endothelium, Vascular/metabolism , Glucosamine/metabolism , Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism , Heparin/metabolism , Heparitin Sulfate/isolation & purification , Hydrolysis , Lipoprotein Lipase/chemistry , Oligosaccharides/metabolism , Protein Binding , Protein Structure, Tertiary , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...