Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Lipid Res ; 64(1): 100316, 2023 01.
Article in English | MEDLINE | ID: mdl-36410424

ABSTRACT

The large HDL particles generated by administration of cholesteryl ester transfer protein inhibitors (CETPi) remain poorly characterized, despite their potential importance in the routing of cholesterol to the liver for excretion, which is the last step of the reverse cholesterol transport. Thus, the effects of the CETPi dalcetrapib and anacetrapib on HDL particle composition were studied in rabbits and humans. The association of rabbit HDL to the LDL receptor (LDLr) in vitro was also evaluated. New Zealand White rabbits receiving atorvastatin were treated with dalcetrapib or anacetrapib. A subset of patients from the dal-PLAQUE-2 study treated with dalcetrapib or placebo were also studied. In rabbits, dalcetrapib and anacetrapib increased HDL-C by more than 58% (P < 0.01) and in turn raised large apo E-containing HDL by 66% (P < 0.001) and 59% (P < 0.01), respectively. Additionally, HDL from CETPi-treated rabbits competed with human LDL for binding to the LDLr on HepG2 cells more than control HDL (P < 0.01). In humans, dalcetrapib increased concentrations of large HDL particles (+69%, P < 0.001) and apo B-depleted plasma apo E (+24%, P < 0.001), leading to the formation of apo E-containing HDL (+47%, P < 0.001) devoid of apo A-I. Overall, in rabbits and humans, CETPi increased large apo E-containing HDL particle concentration, which can interact with hepatic LDLr. The catabolism of these particles may depend on an adequate level of LDLr to contribute to reverse cholesterol transport.


Subject(s)
Anticholesteremic Agents , Humans , Rabbits , Animals , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Cholesterol/metabolism , Apolipoproteins E/metabolism , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol, HDL
2.
Nat Commun ; 12(1): 4662, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34341345

ABSTRACT

Impaired cellular cholesterol efflux is a key factor in the progression of renal, cardiovascular, and autoimmune diseases. Here we describe a class of 5-arylnicotinamide compounds, identified through phenotypic drug discovery, that upregulate ABCA1-dependent cholesterol efflux by targeting Oxysterol Binding Protein Like 7 (OSBPL7). OSBPL7 was identified as the molecular target of these compounds through a chemical biology approach, employing a photoactivatable 5-arylnicotinamide derivative in a cellular cross-linking/immunoprecipitation assay. Further evaluation of two compounds (Cpd A and Cpd G) showed that they induced ABCA1 and cholesterol efflux from podocytes in vitro and normalized proteinuria and prevented renal function decline in mouse models of proteinuric kidney disease: Adriamycin-induced nephropathy and Alport Syndrome. In conclusion, we show that small molecule drugs targeting OSBPL7 reveal an alternative mechanism to upregulate ABCA1, and may represent a promising new therapeutic strategy for the treatment of renal diseases and other disorders of cellular cholesterol homeostasis.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Cholesterol/metabolism , Diabetic Nephropathies/metabolism , Organic Chemicals/pharmacology , Podocytes/metabolism , Proteinuria/metabolism , Receptors, Steroid/antagonists & inhibitors , ATP Binding Cassette Transporter 1/genetics , Animals , Biological Transport/drug effects , Cells, Cultured , Disease Models, Animal , HEK293 Cells , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Mice, 129 Strain , Mice, Knockout , Molecular Structure , Niacinamide/chemistry , Niacinamide/pharmacology , Organic Chemicals/chemical synthesis , Organic Chemicals/chemistry , Podocytes/cytology , RNA Interference , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , THP-1 Cells
3.
J Clin Invest ; 129(8): 3387-3400, 2019 07 22.
Article in English | MEDLINE | ID: mdl-31329164

ABSTRACT

Fibroblasts from patients with Tangier disease carrying ATP-binding cassette A1 (ABCA1) loss-of-function mutations are characterized by cardiolipin accumulation, a mitochondrial-specific phospholipid. Suppression of ABCA1 expression occurs in glomeruli from patients with diabetic kidney disease (DKD) and in human podocytes exposed to DKD sera collected prior to the development of DKD. We demonstrated that siRNA ABCA1 knockdown in podocytes led to reduced oxygen consumption capabilities associated with alterations in the oxidative phosphorylation (OXPHOS) complexes and with cardiolipin accumulation. Podocyte-specific deletion of Abca1 (Abca1fl/fl) rendered mice susceptible to DKD, and pharmacological induction of ABCA1 improved established DKD. This was not mediated by free cholesterol, as genetic deletion of sterol-o-acyltransferase-1 (SOAT1) in Abca1fl/fl mice was sufficient to cause free cholesterol accumulation but did not cause glomerular injury. Instead, cardiolipin mediates ABCA1-dependent susceptibility to podocyte injury, as inhibition of cardiolipin peroxidation with elamipretide improved DKD in vivo and prevented ABCA1-dependent podocyte injury in vitro and in vivo. Collectively, we describe a pathway definitively linking ABCA1 deficiency to cardiolipin-driven mitochondrial dysfunction. We demonstrated that this pathway is relevant to DKD and that ABCA1 inducers or inhibitors of cardiolipin peroxidation may each represent therapeutic strategies for the treatment of established DKD.


Subject(s)
ATP Binding Cassette Transporter 1/deficiency , Cardiolipins/metabolism , Diabetic Nephropathies/metabolism , Lipid Peroxidation , Mitochondria/metabolism , ATP Binding Cassette Transporter 1/metabolism , Animals , Cardiolipins/genetics , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Humans , Mice , Mitochondria/genetics , Mitochondria/pathology , Podocytes , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism
4.
Elife ; 82019 03 13.
Article in English | MEDLINE | ID: mdl-30864945

ABSTRACT

Age-related macular degeneration (AMD) is a progressive disease of the retinal pigment epithelium (RPE) and the retina leading to loss of central vision. Polymorphisms in genes involved in lipid metabolism, including the ATP-binding cassette transporter A1 (ABCA1), have been associated with AMD risk. However, the significance of retinal lipid handling for AMD pathogenesis remains elusive. Here, we study the contribution of lipid efflux in the RPE by generating a mouse model lacking ABCA1 and its partner ABCG1 specifically in this layer. Mutant mice show lipid accumulation in the RPE, reduced RPE and retinal function, retinal inflammation and RPE/photoreceptor degeneration. Data from human cell lines indicate that the ABCA1 AMD risk-conferring allele decreases ABCA1 expression, identifying the potential molecular cause that underlies the genetic risk for AMD. Our results highlight the essential homeostatic role for lipid efflux in the RPE and suggest a pathogenic contribution of reduced ABCA1 function to AMD.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Lipid Metabolism , Retinal Degeneration/physiopathology , Retinal Pigment Epithelium/physiopathology , ATP Binding Cassette Transporter 1/deficiency , ATP Binding Cassette Transporter, Subfamily G, Member 1/deficiency , Animals , Cell Line , Disease Models, Animal , Humans , Inflammation/pathology , Mice , Photoreceptor Cells/pathology
5.
Sci Rep ; 8(1): 5865, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29650998

ABSTRACT

Genome-wide association studies (GWAS) have identified numerous genetic variants in the human genome associated with diseases and traits. Nevertheless, for most loci the causative variant is still unknown. Expression quantitative trait loci (eQTL) in disease relevant tissues is an excellent approach to correlate genetic association with gene expression. While liver is the primary site of gene transcription for two pathways relevant to age-related macular degeneration (AMD), namely the complement system and cholesterol metabolism, we explored the contribution of AMD associated variants to modulate liver gene expression. We extracted publicly available data and computed the largest eQTL data set for liver tissue to date. Genotypes and expression data from all studies underwent rigorous quality control. Subsequently, Matrix eQTL was used to identify significant local eQTL. In total, liver samples from 588 individuals revealed 202,489 significant eQTL variants affecting 1,959 genes (Q-Value < 0.001). In addition, a further 101 independent eQTL signals were identified in 93 of the 1,959 eQTL genes. Importantly, our results independently reinforce the notion that high density lipoprotein metabolism plays a role in AMD pathogenesis. Taken together, our study generated a first comprehensive map reflecting the genetic regulatory landscape of gene expression in liver.


Subject(s)
Gene Expression Regulation/genetics , Genetic Predisposition to Disease , Macular Degeneration/genetics , Quantitative Trait Loci/genetics , Genome, Human/genetics , Genome-Wide Association Study , Genotype , Humans , Liver/metabolism , Macular Degeneration/pathology , Phenotype , Polymorphism, Single Nucleotide
6.
Exp Eye Res ; 165: 65-77, 2017 12.
Article in English | MEDLINE | ID: mdl-28943268

ABSTRACT

Genetic studies have linked age-related macular degeneration (AMD) to genes involved in high-density lipoprotein (HDL) metabolism, including ATP-binding cassette transporter A1 (ABCA1). The retinal pigment epithelium (RPE) handles large amounts of lipids, among others cholesterol, partially derived from internalized photoreceptor outer segments (OS) and lipids physiologically accumulate in the aging eye. To analyze the potential function of ABCA1 in the eye, we measured cholesterol efflux, the first step of HDL generation, in RPE cells. We show the expression of selected genes related to HDL metabolism in mouse and human eyecups as well as in ARPE-19 and human primary RPE cells. Immunofluorescence staining revealed localization of ABCA1 on both sides of polarized RPE cells. This was functionally confirmed by directional efflux to apolipoprotein AI (ApoA-I) of 3H-labeled cholesterol given to the cells via serum or via OS. ABCA1 expression and activity was modulated using a liver-X-receptor (LXR) agonist and an ABCA1 neutralizing antibody, demonstrating that the efflux was ABCA1-dependent. We concluded that the ABCA1-mediated lipid efflux pathway, and hence HDL biosynthesis, is functional in RPE cells towards both the basal (choroidal) and apical (subretinal) space. Impaired activity of the pathway might cause age-related perturbations of lipid homeostasis in the outer retina and thus may contribute to disease development and/or progression.


Subject(s)
Cholesterol/metabolism , Retinal Photoreceptor Cell Outer Segment/metabolism , Retinal Pigment Epithelium/metabolism , ATP Binding Cassette Transporter 1/metabolism , Animals , Blotting, Western , Cells, Cultured , Epithelial Cells/metabolism , Humans , Mice , Polymerase Chain Reaction , Swine
7.
J Lipid Res ; 58(7): 1282-1291, 2017 07.
Article in English | MEDLINE | ID: mdl-28515138

ABSTRACT

Inhibition of cholesteryl ester transfer protein (CETP) increases HDL cholesterol (HDL-C) levels. However, the circulating CETP level varies and the impact of its inhibition in species with high CETP levels on HDL structure and function remains poorly characterized. This study investigated the effects of dalcetrapib and anacetrapib, the two CETP inhibitors (CETPis) currently being tested in large clinical outcome trials, on HDL particle subclass distribution and cholesterol efflux capacity of serum in rabbits and monkeys. New Zealand White rabbits and vervet monkeys received dalcetrapib and anacetrapib. In rabbits, CETPis increased HDL-C, raised small and large α-migrating HDL, and increased ABCA1-induced cholesterol efflux. In vervet monkeys, although anacetrapib produced similar results, dalcetrapib caused opposite effects because the LDL-C level was increased by 42% and HDL-C decreased by 48% (P < 0.01). The levels of α- and preß-HDL were reduced by 16% (P < 0.001) and 69% (P < 0.01), resulting in a decrease of the serum cholesterol efflux capacity. CETPis modulate the plasma levels of mature and small HDL in vivo and consequently the cholesterol efflux capacity. The opposite effects of dalcetrapib in different species indicate that its impact on HDL metabolism could vary greatly according to the metabolic environment.


Subject(s)
Cholesterol, HDL/chemistry , Cholesterol, HDL/metabolism , Oxazolidinones/pharmacology , Sulfhydryl Compounds/pharmacology , Amides , Animals , Apolipoprotein A-I/metabolism , Biological Transport/drug effects , Chlorocebus aethiops , Cholesterol Ester Transfer Proteins/metabolism , Esters , Hep G2 Cells , Humans , Male , Rabbits , Species Specificity
8.
J Lipid Res ; 58(7): 1325-1337, 2017 07.
Article in English | MEDLINE | ID: mdl-28442497

ABSTRACT

We developed an in silico mathematical model of retinal cholesterol (Ch) dynamics (RCD) to quantify the physiological rate of Ch turnover in the rod outer segment (ROS), the lipoprotein transport mechanisms by which Ch enters and leaves the outer retina, and the rates of drusen growth and macrophage-mediated clearance in dry age-related macular degeneration. Based on existing experimental data and mechanistic hypotheses, we estimated the Ch turnover rate in the ROS to be 1-6 pg/mm2/min, dependent on the rate of Ch recycling in the outer retina, and found comparable rates for LDL receptor-mediated endocytosis of Ch by the retinal pigment epithelium (RPE), ABCA1-mediated Ch transport from the RPE to the outer retina, ABCA1-mediated Ch efflux from the RPE to the choroid, and the secretion of 70 nm ApoB-Ch particles from the RPE. The drusen growth rate is predicted to increase from 0.7 to 4.2 µm/year in proportion to the flux of ApoB-Ch particles. The rapid regression of drusen may be explained by macrophage-mediated clearance if the macrophage density reaches ∼3,500 cells/mm2 The RCD model quantifies retinal Ch dynamics and suggests that retinal Ch turnover and recycling, ApoB-Ch particle efflux, and macrophage-mediated clearance may explain the dynamics of drusen growth and regression.


Subject(s)
Cholesterol/metabolism , Computer Simulation , Macular Degeneration/metabolism , Retina/metabolism , Biological Transport , Humans , Macular Degeneration/physiopathology , Retinal Pigment Epithelium/metabolism , Rod Cell Outer Segment/metabolism
9.
J Struct Biol ; 194(2): 191-8, 2016 May.
Article in English | MEDLINE | ID: mdl-26876146

ABSTRACT

The cholesteryl ester transfer protein (CETP) enables the transfer of cholesteryl ester (CE) from high-density lipoproteins (HDL) to low-density lipoproteins (LDL) in the plasma compartment. CETP inhibition raises plasma levels of HDL cholesterol; a ternary tunnel complex with CETP bridging HDL and LDL was suggested as a mechanism. Here, we test whether the inhibition of CETP tunnel complex formation is a promising approach to suppress CE transfer from HDL to LDL, for potential treatment of cardio-vascular disease (CVD). Three monoclonal antibodies against different epitopes of CETP are assayed for their potential to interfere with CE transfer between HDL and/or LDL. Surprisingly, antibodies that target the tips of the elongated CETP molecule, interaction sites sterically required to form the suggested transfer complexes, do not interfere with CETP activity, but an antibody binding to the central region does. We show that CETP interacts with HDL, but not with LDL. Our findings demonstrate that a ternary tunnel complex is not the mechanistic prerequisite to transfer CE among lipoproteins.


Subject(s)
Cholesterol Ester Transfer Proteins/metabolism , Cholesterol Esters/metabolism , Epitopes/chemistry , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , Biological Transport , Cell Line , Cholesterol Ester Transfer Proteins/genetics , Cholesterol Ester Transfer Proteins/ultrastructure , Epitopes/ultrastructure , Gene Expression , Humans , Lipoproteins, HDL/ultrastructure , Lipoproteins, LDL/ultrastructure , Microscopy, Electron, Transmission , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure
10.
Biochim Biophys Acta ; 1831(11): 1644-50, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23872476

ABSTRACT

Cholesteryl ester transfer protein (CETP), a key regulator of high-density lipoprotein (HDL) metabolism, induces HDL remodeling by transferring lipids between apolipoprotein B-containing lipoproteins and HDL, and/or by promoting lipid transfer between HDL subparticles. In this study, we investigated the mechanism as to how CETP induces the generation of lipid-poor particles (pre-ß-HDL) from HDL, which increases ATP-binding cassette transporter 1-mediated cholesterol efflux. This CETP-dependent HDL remodeling is enhanced by the CETP modulator dalcetrapib both in plasma and isolated HDL. The interaction of dalcetrapib with cysteine 13 of CETP is required, since this effect was abolished when using mutant CETP in which cysteine 13 was substituted for a serine residue. Other thiol-containing compounds were identified as CETP modulators interacting with cysteine 13 of CETP. In order to mimic dalcetrapib-bound CETP, mutant CETP proteins were prepared by replacing cysteine 13 with the bulky amino acid tyrosine or tryptophan. The resultant mutants showed virtually no CETP-dependent lipid transfer activity but demonstrated preserved CETP-dependent pre-ß-HDL generation. Overall, these data demonstrate that the two functions of CETP i.e., cholesteryl ester transfer and HDL remodeling can be uncoupled by interaction of thiol-containing compounds with cysteine 13 of CETP or by introducing large amino acid residues in place of cysteine 13.


Subject(s)
Cholesterol Ester Transfer Proteins/chemistry , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol/metabolism , Cysteine/chemistry , Lipoproteins, HDL/metabolism , Biological Transport/genetics , Biological Transport/physiology , Cell Line , Cholesterol Ester Transfer Proteins/genetics , Cysteine/genetics , Humans , Plasma , Structure-Activity Relationship
11.
Atherosclerosis ; 229(1): 94-101, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23725986

ABSTRACT

OBJECTIVE: Promoting reverse cholesterol transport (RCT) is a major atheroprotective property of HDL. The present study explored the effect of stimulating the first step of RCT (cholesterol efflux from macrophages) alone or in combination with stimulating the last step of RCT (fecal sterol excretion). METHODS AND RESULTS: Reconstituted HDL (rHDL) was injected into wild-type mice either with or without administration of the cholesterol absorption inhibitor ezetimibe or the bile acid sequestrant cholestyramine. Single dose administration of rHDL (100 mg apoA-I/kg) resulted in an early (4 h) increase in plasma free cholesterol levels (p < 0.001), without affecting hepatic cholesterol levels or fecal mass sterol excretion. rHDL injection also increased [(3)H]cholesterol appearance in plasma at an early time-point (4 h) after intraperitoneal administration of [(3)H]cholesterol-labeled mouse macrophage foam cells and fecal radioactivity excretion indicating completed RCT was increased by 26% (p < 0.05). Ezetimibe treatment inhibited intestinal cholesterol absorption by 74% (p < 0.01), but also the bile acid sequestrant cholestyramine decreased cholesterol absorption significantly (24%, p < 0.01). Consequently, ezetimibe increased RCT 2.1-fold (p < 0.001) primarily within fecal neutral sterols, while cholestyramine increased RCT by 3.6-fold (p < 0.001), primarily within bile acids (p < 0.001), but also within neutral sterols (p < 0.001). However, no additive effects of both intestinal sterol uptake inhibitors were observed on top of rHDL administration. CONCLUSION: These data demonstrate that increasing the first step of RCT by rHDL administration results in transient cholesterol mobilization from macrophages to plasma. This effect is not further enhanced by stimulating the last step of RCT, fecal sterol excretion.


Subject(s)
Azetidines/pharmacology , Cholesterol, HDL/metabolism , Cholesterol, HDL/pharmacology , Cholestyramine Resin/pharmacology , Animals , Anticholesteremic Agents/pharmacology , Apolipoprotein A-I/metabolism , Bile Acids and Salts/metabolism , Biological Transport/drug effects , Biological Transport/physiology , Cholesterol, HDL/blood , Drug Synergism , Ezetimibe , Feces , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Liver/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Sterols/metabolism , Triglycerides/blood
12.
Circ Res ; 113(2): 167-75, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23676183

ABSTRACT

RATIONALE: High-density lipoprotein cholesterol elevation via cholesteryl ester transfer protein (CETP) inhibition represents a novel therapy for atherosclerosis, which also may have relevance for type 2 diabetes mellitus. OBJECTIVE: The current study assessed the effects of a CETP inhibitor on postprandial insulin, ex vivo insulin secretion, and cholesterol efflux from pancreatic ß-cells. METHODS AND RESULTS: Healthy participants received a daily dose of CETP inhibitor (n=10) or placebo (n=15) for 14 days in a randomized double-blind study. Insulin secretion and cholesterol efflux from MIN6N8 ß-cells were determined after incubation with treated plasma. CETP inhibition increased plasma high-density lipoprotein cholesterol, apolipoprotein AI, and postprandial insulin. MIN6N8 ß-cells incubated with plasma from CETP inhibitor-treated individuals (compared with placebo) exhibited an increase in both glucose-stimulated insulin secretion and cholesterol efflux over the 14-day treatment period. CONCLUSIONS: CETP inhibition increased postprandial insulin and promoted ex vivo ß-cell glucose-stimulated insulin secretion, potentially via enhanced ß-cell cholesterol efflux.


Subject(s)
Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cholesterol Ester Transfer Proteins/blood , Cholesterol, HDL/blood , Insulin/metabolism , Amides , Animals , Cell Line , Double-Blind Method , Esters , Fasting/blood , Humans , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Rats , Sulfhydryl Compounds/pharmacology , Treatment Outcome
13.
J Lipid Res ; 52(12): 2323-2331, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21971713

ABSTRACT

The composition of lipoproteins and the association of proteins with various particles are of much interest in the context of cardiovascular disease. Here, we describe a technique for the multidimensional analysis of lipoproteins and their associated apolipoproteins. Plasma is separated by size exclusion chromatography (SEC), and fractions are analyzed by reverse-phase arrays. SEC fractions are spotted on nitrocellulose slides and incubated with different antibodies against individual apolipoproteins or antibodies against various apolipoproteins. In this way, tens of analytes can be measured simultaneously in 100 µl of plasma from a single SEC separation. This methodology is particularly suited to simultaneous analysis of multiple proteins that may change their distribution to lipoproteins or alter their conformation, depending on factors that influence circulating lipoprotein size or composition. We observed changes in the distribution of exchangeable apolipoproteins following addition of recombinant apolipoproteins or interaction with exogenous compounds. While the cholesteryl ester transfer protein (CETP)-dependent formation of pre-ß-HDL was inhibited by the CETP inhibitors torcetrapib and anacetrapib, it was not reduced by the CETP modulator dalcetrapib. This finding was elucidated using this technique.


Subject(s)
Blood Chemical Analysis/methods , Chromatography, Gel/methods , Lipoproteins/blood , Lipoproteins/isolation & purification , Protein Array Analysis/methods , Antibody Specificity , Artifacts , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol Ester Transfer Proteins/pharmacology , High-Density Lipoproteins, Pre-beta/blood , High-Density Lipoproteins, Pre-beta/metabolism , Humans , Lipoproteins/immunology , Lipoproteins/metabolism , Quinolines/pharmacology , Reproducibility of Results
14.
J Lipid Res ; 51(12): 3443-54, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20861162

ABSTRACT

The mechanism by which cholesteryl ester transfer protein (CETP) activity affects HDL metabolism was investigated using agents that selectively target CETP (dalcetrapib, torcetrapib, anacetrapib). In contrast with torcetrapib and anacetrapib, dalcetrapib requires cysteine 13 to decrease CETP activity, measured as transfer of cholesteryl ester (CE) from HDL to LDL, and does not affect transfer of CE from HDL3 to HDL2. Only dalcetrapib induced a conformational change in CETP, when added to human plasma in vitro, also observed in vivo and correlated with CETP activity. CETP-induced pre-ß-HDL formation in vitro in human plasma was unchanged by dalcetrapib ≤3 µM and increased at 10 µM. A dose-dependent inhibition of pre-ß-HDL formation by torcetrapib and anacetrapib (0.1 to 10 µM) suggested that dalcetrapib modulates CETP activity. In hamsters injected with [³H]cholesterol-labeled autologous macrophages, and given dalcetrapib (100 mg twice daily), torcetrapib [30 mg once daily (QD)], or anacetrapib (30 mg QD), only dalcetrapib significantly increased fecal elimination of both [³H]neutral sterols and [³H]bile acids, whereas all compounds increased plasma HDL-[³H]cholesterol. These data suggest that modulation of CETP activity by dalcetrapib does not inhibit CETP-induced pre-ß-HDL formation, which may be required to increase reverse cholesterol transport.


Subject(s)
Anticholesteremic Agents/pharmacology , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol/metabolism , High-Density Lipoproteins, Pre-beta/metabolism , Amides , Animals , Bile Acids and Salts/metabolism , Binding Sites , Biological Transport/drug effects , Cholesterol/blood , Cholesterol Ester Transfer Proteins/blood , Cricetinae , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Esters , High-Density Lipoproteins, Pre-beta/blood , Humans , Oxazolidinones/pharmacology , Quinolines/pharmacology , Sulfhydryl Compounds/pharmacology
15.
Toxicol In Vitro ; 24(3): 745-50, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20074633

ABSTRACT

Dalcetrapib (RO4607381/JTT-705), an agent that targets cholesteryl ester transfer protein, is in development for prevention of cardiovascular events. In vitro studies were performed to identify receptors that mediate an off-target effect of dalcetrapib observed in preclinical models: increased lipid uptake into the lamina propria of the small intestine and into mesenteric lymph node macrophages. Uptake of oxidized low-density lipoprotein (LDL) cholesterol or dalcetrapib-treated chylomicrons was quantitated by triglyceride assay or fluorescent labeling in primary macrophages and the cell lines CHO, J774A.1 (mouse macrophages) and THP-1 (human macrophages). Quantitative reverse-transcriptase polymerase chain reaction and immunoblotting measured candidate receptor expression. Lectin-like oxidized LDL receptor (LOX-1) and scavenger receptor type AI (SR-AI) were excluded as candidate receptors based on lack of association between their expression and uptake of dalcetrapib-treated lipids. In J774A.1 cells, uptake of dalcetrapib-treated chylomicrons was increased by LPS and associated with expression of MAcrophage Receptor with COllagenous domain (MARCO). MARCO was expressed at very low levels in human macrophages and was not inducible by LPS. The MARCO receptor may account for the variable species susceptibility towards dalcetrapib-mediated chylomicron uptake by macrophages.


Subject(s)
Anticholesteremic Agents/pharmacology , Lipid Metabolism/drug effects , Macrophages/metabolism , Receptors, Immunologic/physiology , Sulfhydryl Compounds/pharmacology , Amides , Amino Acid Oxidoreductases/metabolism , Animals , Blotting, Western , Cell Line , Cholesterol, VLDL/metabolism , Chylomicrons/metabolism , CpG Islands , Esters , Humans , Interleukin-12/biosynthesis , Interleukin-4/biosynthesis , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Oxidation-Reduction , RNA/biosynthesis , RNA/isolation & purification , Rats , Rats, Sprague-Dawley , Th1 Cells/drug effects
16.
Bioorg Med Chem Lett ; 19(6): 1654-7, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19231176

ABSTRACT

A series of tetrahydro-cyclopenta[b]indoles modulating the activity of the liver-X-receptor (LXR) were derived from a high throughput screening hit. The potency and selectivity for LXRbeta versus LXRalpha was improved. One compound, administered to wild-type mice modestly increased plasma HDL-cholesterol with no change in plasma triglycerides (TG) and reduced effects on liver TG content compared to T0901317. This novel series of LXR agonists shows promise to improve therapeutic efficacy with reduced potential to increase TG.


Subject(s)
Chemistry, Pharmaceutical/methods , DNA-Binding Proteins/chemistry , Indoles/chemical synthesis , Receptors, Cytoplasmic and Nuclear/chemistry , Animals , Cholesterol, HDL/metabolism , Drug Design , Hydrocarbons, Fluorinated/pharmacology , Indoles/pharmacology , Inhibitory Concentration 50 , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Models, Chemical , Orphan Nuclear Receptors , Sulfonamides/pharmacology , Transcriptional Activation , Triglycerides/metabolism
17.
J Biol Chem ; 284(10): 6093-100, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19136670

ABSTRACT

Endothelial lipase (EL) is a negative regulator of high density lipoprotein (HDL) cholesterol plasma levels, and scavenger receptor BI (SR-BI) is involved in remodeling of HDL. The present study investigates the requirement of SR-BI for the effects of EL-mediated phospholipid hydrolysis on HDL metabolism in vivo. In vitro, selective uptake from EL-modified HDL was 129% higher than selective uptake from control HDL in SR-BI-overexpressing cells (p=0.01). In vivo overexpression of human EL by means of recombinant adenovirus decreased HDL plasma levels significantly (p<0.01). Fast protein liquid chromatography analysis and agarose gel electrophoresis revealed that EL expression resulted in the generation of small pre-beta HDL particles in wild-type mice, whereas in SR-BI-/- mice small HDL were preferentially removed. In kinetic experiments the fractional catabolic rate (FCR) of HDL cholesteryl ester increased by 110% (p<0.001), and the FCR of HDL apolipoproteins increased by 64% (p<0.001) in response to EL overexpression in wild-type mice. In SR-BI-/- mice a similar increase in the HDL apolipoprotein FCR occurred (p<0.001); however, there was no further increase in HDL cholesteryl ester catabolism. The apparent whole body selective uptake was increased 3-fold by EL in wild-type mice (p<0.001), whereas there was no selective uptake in SR-BI knock-out mice. EL overexpression increased hepatic selective uptake as well as holoparticle uptake (each p<0.01) in wild-type mice, whereas in SR-BI knock-out mice only holoparticle uptake increased (p<0.01). Our results indicate that SR-BI-mediated selective uptake of HDL cholesteryl ester is essential for the remodeling of large alpha-migrating HDL particles by EL.


Subject(s)
Cholesterol Esters/metabolism , Lipase/metabolism , Lipoproteins, HDL/metabolism , Liver/metabolism , Scavenger Receptors, Class B/metabolism , Animals , Cell Line, Tumor , Cholesterol Esters/genetics , Humans , Kinetics , Lipase/genetics , Lipoproteins, HDL/genetics , Mice , Mice, Knockout , Scavenger Receptors, Class B/genetics
18.
Cardiovasc Res ; 77(4): 732-9, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18056760

ABSTRACT

AIMS: Cholesteryl ester transfer protein (CETP) has a well-established role in lipoprotein metabolism, but the effect of its overexpression or inhibition on the efficiency of reverse cholesterol transport (RCT) is unclear. METHODS AND RESULTS: Neither overexpression of CETP nor treatment with CETP inhibitor Torcetrapib of RAW 264.7 macrophages or HepG2 hepatocytes affected cholesterol efflux in vitro. Overexpression of CETP or treatment with Torcetrapib, respectively, stimulated or inhibited HDL cholesteryl ester uptake by HepG2 but not by RAW 264.7 cells. When RAW 264.7 cells transfected with CETP or ATP binding cassette transporter A1 (ABCA1) were injected intraperitoneally into mice, cholesterol egress from macrophages was elevated for ABCA1- but not for CETP-transfected macrophages. Systemic expression of CETP in mice by adenoviral infection stimulated egress of cholesterol to plasma and liver without affecting HDL levels. Treatment with Torcetrapib did not affect appearance of macrophage cholesterol in plasma and liver, but inhibited its excretion into feces. Treatment of hamsters with Torcetrapib led to elevation of HDL cholesterol, an increase in the capacity of plasma to support cholesterol efflux, and increased egress of cholesterol from macrophages to plasma and feces in vivo. CONCLUSION: Both increased (mice study) and decreased (hamster study) CETP activity could result in enhanced RCT.


Subject(s)
Anticholesteremic Agents/pharmacology , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol/metabolism , Macrophages/drug effects , Macrophages/metabolism , Quinolines/pharmacology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Cell Line , Cholesterol/blood , Cholesterol Ester Transfer Proteins/genetics , Cholesterol Esters/metabolism , Cholesterol, HDL/metabolism , Cholesterol, LDL/metabolism , Cholesterol, VLDL/metabolism , Cricetinae , Dose-Response Relationship, Drug , Feces/chemistry , Humans , Liver/drug effects , Liver/metabolism , Macrophages/transplantation , Mice , Time Factors , Transfection , Triglycerides/metabolism , Up-Regulation
19.
J Lipid Res ; 46(12): 2681-91, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16150825

ABSTRACT

The cellular and molecular mechanisms responsible for lipoprotein [a] (Lp[a]) catabolism are unknown. We examined the plasma clearance of Lp[a] and LDL in mice using lipoproteins isolated from human plasma coupled to radiolabeled tyramine cellobiose. Lipoproteins were injected into wild-type, LDL receptor-deficient (Ldlr-/-), and apolipoprotein E-deficient (Apoe-/-) mice. The fractional catabolic rate of LDL was greatly slowed in Ldlr-/- mice and greatly accelerated in Apoe-/- mice compared with wild-type mice. In contrast, the plasma clearance of Lp[a] in Ldlr-/- mice was similar to that in wild-type mice and was only slightly accelerated in Apoe-/- mice. Hepatic uptake of Lp[a] in wild-type mice was 34.6% of the injected dose over a 24 h period. The kidney accounted for only a small fraction of tissue uptake (1.3%). To test whether apolipoprotein [a] (apo[a]) mediates the clearance of Lp[a] from plasma, we coinjected excess apo[a] with labeled Lp[a]. Apo[a] acted as a potent inhibitor of Lp[a] plasma clearance. Asialofetuin, a ligand of the asialoglycoprotein receptor, did not inhibit Lp[a] clearance. In summary, the liver is the major organ accounting for the clearance of Lp[a] in mice, with the LDL receptor and apolipoprotein E having no major roles. Our studies indicate that apo[a] is the primary ligand that mediates Lp[a] uptake and plasma clearance.


Subject(s)
Apolipoproteins A/metabolism , Lipoprotein(a)/blood , Liver/metabolism , Animals , Apolipoproteins A/blood , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Asialoglycoproteins/blood , Cholesterol, LDL/metabolism , Fetuins , Lipoprotein(a)/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, LDL/metabolism , alpha-Fetoproteins
20.
Arterioscler Thromb Vasc Biol ; 25(3): 560-5, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15637307

ABSTRACT

OBJECTIVE: We addressed the role of the low-density lipoprotein (LDL) receptor in determining clearance rates and production rate (PR) of apolipoprotein B (apoB) in humans. METHODS AND RESULTS: Kinetic studies using endogenous labeling of apoB with deuterated leucine were performed in 7 genetically defined patients with homozygous familial hypercholesterolemia (FH) and compared with 4 controls. The fractional catabolic rates (FCR) and PRs for apoB were determined by multicompartmental modeling. The FCRs of very-low-density lipoprotein 1 (VLDL1), VLDL2, intermediate-density lipoprotein (IDL), and LDL apoB were lower in FH than in controls, with the LDL apoB FCR being significantly lower (0.148+/-0.049 versus 0.499+/-0.099 pools x d(-1); P=0.008). Whereas receptor-defective FH patients had a total apoB PR similar to controls, receptor-null FH patients had a significantly greater total apoB PR than controls (35.97+/-10.51 versus 21.32+/-4.21 mg x kg(-1) x d(-1), respectively; P=0.02). CONCLUSIONS: This first study of apoB metabolism in homozygous FH using endogenous labeling with stable isotopes demonstrates that the LDL receptor contributes significantly to the clearance of LDL from plasma but plays a lesser role in the clearance of larger apoB-containing lipoproteins. Furthermore, these data also indicate that absence of a LDL receptor in humans substantially influences the apoB PR in vivo.


Subject(s)
Apolipoproteins B/blood , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Adolescent , Adult , Apolipoprotein B-100 , Apolipoproteins B/biosynthesis , Cholesterol, LDL/blood , Cholesterol, VLDL/blood , Female , Homozygote , Humans , Leucine/pharmacokinetics , Male , Middle Aged , Receptors, LDL/deficiency , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...