Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Nature ; 584(7819): 87-92, 2020 08.
Article in English | MEDLINE | ID: mdl-32699412

ABSTRACT

The initial colonization of the Americas remains a highly debated topic1, and the exact timing of the first arrivals is unknown. The earliest archaeological record of Mexico-which holds a key geographical position in the Americas-is poorly known and understudied. Historically, the region has remained on the periphery of research focused on the first American populations2. However, recent investigations provide reliable evidence of a human presence in the northwest region of Mexico3,4, the Chiapas Highlands5, Central Mexico6 and the Caribbean coast7-9 during the Late Pleistocene and Early Holocene epochs. Here we present results of recent excavations at Chiquihuite Cave-a high-altitude site in central-northern Mexico-that corroborate previous findings in the Americas10-17of cultural evidence that dates to the Last Glacial Maximum (26,500-19,000 years ago)18, and which push back dates for human dispersal to the region possibly as early as 33,000-31,000 years ago. The site yielded about 1,900 stone artefacts within a 3-m-deep stratified sequence, revealing a previously unknown lithic industry that underwent only minor changes over millennia. More than 50 radiocarbon and luminescence dates provide chronological control, and genetic, palaeoenvironmental and chemical data document the changing environments in which the occupants lived. Our results provide new evidence for the antiquity of humans in the Americas, illustrate the cultural diversity of the earliest dispersal groups (which predate those of the Clovis culture) and open new directions of research.


Subject(s)
Human Migration/history , Ice Cover , Altitude , Archaeology , Bayes Theorem , Caves , Cultural Diversity , DNA, Ancient/analysis , History, Ancient , Humans , Mexico
2.
Curr Opin Lipidol ; 34(6): 278-286, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37732779

ABSTRACT

PURPOSE OF REVIEW: Several large studies have shown increased mortality due to all-causes and to atherosclerotic cardiovascular disease. In most clinical settings, plasma HDL-cholesterol is determined as a sum of free cholesterol and cholesteryl ester, two molecules with vastly different metabolic itineraries. We examine the evidence supporting the concept that the pathological effects of elevations of plasma HDL-cholesterol are due to high levels of the free cholesterol component of HDL-C. RECENT FINDINGS: In a small population of humans, a high plasma HDL-cholesterol is associated with increased mortality. Similar observations in the HDL-receptor deficient mouse (Scarb1 -/- ), a preclinical model of elevated HDL-C, suggests that the pathological component of HDL in these patients is an elevated plasma HDL-FC. SUMMARY: Collective consideration of the human and mouse data suggests that clinical trials, especially in the setting of high plasma HDL, should measure free cholesterol and cholesteryl esters and not just total cholesterol.


Subject(s)
Atherosclerosis , Hypercholesterolemia , Humans , Animals , Mice , Cholesterol, HDL , Cholesterol Esters/metabolism , Cholesterol , Atherosclerosis/genetics , Cholesterol Ester Transfer Proteins
3.
J Lipid Res ; 64(2): 100327, 2023 02.
Article in English | MEDLINE | ID: mdl-36596339

ABSTRACT

Human female infertility, 20% of which is idiopathic, is a public health problem for which better diagnostics and therapeutics are needed. A novel cause of infertility emerged from studies of female mice deficient in the HDL receptor gene (Scarb1). These mice are infertile and have high plasma HDL cholesterol (C) concentrations, due to elevated HDL-free cholesterol (FC), which transfers from HDL to all tissues. Previous studies have indicated that oral delivery of probucol, an HDL-lowering drug, to female Scarb1-/- mice reduces plasma HDL-C concentrations and rescues fertility. Additionally, serum opacity factor (SOF), a bacterial virulence factor, disrupts HDL structure, and bolus SOF injection into mice reduces plasma HDL-C concentrations. Here, we discovered that delivering SOF to female Scarb1-/- mice with an adeno-associated virus (AAVSOF) induces constitutive SOF expression, reduces HDL-FC concentrations, and rescues fertility while normalizing ovary morphology. Although AAVSOF did not alter ovary-FC content, the ovary-mol% FC correlated with plasma HDL-mol% FC in a fertility-dependent way. Therefore, reversing the abnormal plasma microenvironment of high plasma HDL-mol% FC in female Scarb1-/- mice rescues fertility. These data provide the rationale to search for similar mechanistic links between HDL-mol% FC and infertility and the rescue of fertility in women by reducing plasma HDL-mol% FC.


Subject(s)
Cholesterol , Infertility , Animals , Female , Humans , Mice , Biological Availability , Cholesterol/metabolism , Cholesterol, HDL , Fertility , Scavenger Receptors, Class B/genetics
4.
J Lipid Res ; 64(11): 100456, 2023 11.
Article in English | MEDLINE | ID: mdl-37821077

ABSTRACT

Compared with WT mice, HDL receptor-deficient (Scarb1-/-) mice have higher plasma levels of free cholesterol (FC)-rich HDL and exhibit multiple pathologies associated with a high mol% FC in ovaries, platelets, and erythrocytes, which are reversed by lowering HDL. Bacterial serum opacity factor (SOF) catalyzes the opacification of plasma by targeting and quantitatively converting HDL to neo HDL (HDL remnant), a cholesterol ester-rich microemulsion, and lipid-free APOA1. SOF delivery with an adeno-associated virus (AAVSOF) constitutively lowers plasma HDL-FC and reverses female infertility in Scarb1-/- mice in an HDL-dependent way. We tested whether AAVSOF delivery to Scarb1-/- mice will normalize erythrocyte morphology in an HDL-FC-dependent way. We determined erythrocyte morphology and FC content (mol%) in three groups-WT, untreated Scarb1-/- (control), and Scarb1-/- mice receiving AAVSOF-and correlated these with their respective HDL-mol% FC. Plasma-, HDL-, and tissue-lipid compositions were also determined. Plasma- and HDL-mol% FC positively correlated across all groups. Among Scarb1-/- mice, AAVSOF treatment normalized reticulocyte number, erythrocyte morphology, and erythrocyte-mol% FC. Erythrocyte-mol% FC positively correlated with HDL-mol% FC and with both the number of reticulocytes and abnormal erythrocytes. AAVSOF treatment also reduced FC of extravascular tissues to a lesser extent. HDL-FC spontaneously transfers from plasma HDL to cell membranes. AAVSOF treatment lowers erythrocyte-FC and normalizes erythrocyte morphology and lipid composition by reducing HDL-mol% FC.


Subject(s)
Cholesterol , Peptide Hydrolases , Female , Mice , Animals , Cholesterol, HDL , Cholesterol Esters/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism
5.
Arterioscler Thromb Vasc Biol ; 41(10): e453-e467, 2021 10.
Article in English | MEDLINE | ID: mdl-34380332

ABSTRACT

Objective: Overall and atherosclerosis-associated mortality is elevated in humans with very high HDL (high-density lipoprotein) cholesterol concentrations. Mice with a deficiency of the HDL receptor, Scarb1 (scavenger receptor class B type 1), are a robust model of this phenotype and exhibit several additional pathologies. We hypothesized that the previously reported high plasma concentration of free cholesterol (FC)-rich HDL in Scarb1-/- mice produces a state of high HDL-FC bioavailability that increases whole-body FC and dysfunction in multiple tissue sites. Approach and Results: The higher mol% FC in Scarb1-/- versus WT (wild type) HDL (41.1 versus 16.0 mol%) affords greater FC bioavailability for transfer to multiple sites. Plasma clearance of autologous HDL-FC mass was faster in WT versus Scarb1-/- mice. FC influx from Scarb1-/- HDL to LDL (low-density lipoprotein) and J774 macrophages was greater ([almost equal to]4x) than that from WT HDL, whereas FC efflux capacity was similar. The higher mol% FC of ovaries, erythrocytes, heart, and macrophages of Scarb1-/- versus WT mice is associated with previously reported female infertility, impaired cell maturation, cardiac dysfunction, and atherosclerosis. The FC contents of other tissues were similar in the two genotypes, and these tissues were not associated with any overt pathology. In addition to the differences between WT versus Scarb1-/- mice, there were many sex-dependent differences in tissue-lipid composition and plasma FC clearance rates. Conclusions: Higher HDL-FC bioavailability among Scarb1-/- versus WT mice drives increased FC content of multiple cell sites and is a potential biomarker that is mechanistically linked to multiple pathologies.


Subject(s)
Atherosclerosis/metabolism , Cholesterol/metabolism , Macrophages/metabolism , Scavenger Receptors, Class B/deficiency , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Biological Availability , Cell Line , Cholesterol, HDL/metabolism , Cholesterol, LDL/metabolism , Female , Humans , Kinetics , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Plaque, Atherosclerotic , Scavenger Receptors, Class B/genetics , Sex Factors , Tissue Distribution
6.
Nanomedicine ; 33: 102361, 2021 04.
Article in English | MEDLINE | ID: mdl-33540069

ABSTRACT

Liposome-based nanoparticles (NPs) comprised mostly of phospholipids (PLs) have been developed to deliver diagnostic and therapeutic agents. Whereas reassembled plasma lipoproteins have been tested as NP carriers of hydrophobic molecules, they are unstable because the components can spontaneously transfer to other PL surfaces-cell membranes and lipoproteins-and can be degraded by plasma lipases. Here we review two strategies for NP stabilization. One is to use PLs that contain long acyl-chains: according to a quantitative thermodynamic model and in vivo tests, increasing the chain length of a PL reduces the spontaneous transfer rate and increases plasma lifetime. A second strategy is to substitute ether for ester bonds which makes the PLs lipase resistant. We conclude with recommendations of simple ex vivo and in vitro tests of NP stability that should be conducted before in vivo tests are begun.


Subject(s)
Delayed-Action Preparations/chemistry , Liposomes/chemistry , Nanoparticles/chemistry , Phospholipids/chemistry , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Drug Liberation , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Lipoproteins/blood , Lipoproteins/metabolism , Liposomes/blood , Nanomedicine , Structure-Activity Relationship , Thermodynamics
7.
Arterioscler Thromb Vasc Biol ; 39(12): 2457-2467, 2019 12.
Article in English | MEDLINE | ID: mdl-31597448

ABSTRACT

The HDL (high-density lipoprotein) Workshop was established in 2009 as a forum for candid discussions among academic basic scientists, clinical investigators, and industry researchers about the role of HDL in cardiovascular disease. This ninth HDL Workshop was held on May 16 to 17, 2019 in Boston, MA, and included outstanding oral presentations from established and emerging investigators. The Workshop featured 5 sessions with topics that tackled the role of HDL in the vasculature, its structural complexity, its role in health and disease states, and its interaction with the intestinal microbiome. The highlight of the program was awarding the Jack Oram Award to the distinguished professor emeritus G.S. Getz from the University of Chicago. The tenth HDL Workshop will be held on May 2020 in Chicago and will continue the focus on intellectually stimulating presentations by established and emerging investigators on novel roles of HDL in cardiovascular and noncardiovascular health and disease states.


Subject(s)
Biomedical Research/methods , Blood Vessels/metabolism , Cardiology , Cardiovascular Diseases/metabolism , Cholesterol, HDL/metabolism , Hypolipidemic Agents/therapeutic use , Societies, Medical , Animals , Cardiovascular Diseases/prevention & control , Congresses as Topic , Humans
8.
J Biol Chem ; 292(21): 8864-8873, 2017 05 26.
Article in English | MEDLINE | ID: mdl-28373285

ABSTRACT

Reverse cholesterol transport (transfer of macrophage-cholesterol in the subendothelial space of the arterial wall to the liver) is terminated by selective high density lipoprotein (HDL)-cholesteryl ester (CE) uptake, mediated by scavenger receptor class B, type 1 (SR-B1). We tested the validity of two models for this process: "gobbling," i.e. one-step transfer of all HDL-CE to the cell and "nibbling," multiple successive cycles of SR-B1-HDL association during which a few CEs transfer to the cell. Concurrently, we compared cellular uptake of apoAI with that of apoAII, which is more lipophilic than apoAI, using HDL-[3H]CE labeled with [125I]apoAI or [125I]apoAII. The studies were conducted in CHO-K1 and CHO-ldlA7 cells (LDLR-/-) with (CHO-SR-B1) and without SR-B1 overexpression and in human Huh7 hepatocytes. Relative to CE, both apoAI and apoAII were excluded from uptake by all cells. However, apoAII was more highly excluded from uptake (2-4×) than apoAI. To distinguish gobbling versus nibbling mechanisms, media from incubations of HDL with CHO-SR-B1 cells were analyzed by non-denaturing PAGE, size-exclusion chromatography, and the distribution of apoAI, apoAII, cholesterol, and phospholipid among HDL species as a function of incubation time. HDL size gradually decreased, i.e. nibbling, with the concurrent release of lipid-free apoAI; apoAII was retained in an HDL remnant. Our data support an SR-B1 nibbling mechanism that is similar to that of streptococcal serum opacity factor, which also selectively removes CE and releases apoAI, leaving an apoAII-rich remnant.


Subject(s)
Apolipoprotein A-II/metabolism , Cholesterol Esters/metabolism , Hepatocytes/metabolism , Lipoproteins, HDL/metabolism , Scavenger Receptors, Class B/metabolism , Animals , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Apolipoprotein A-II/genetics , CHO Cells , Cholesterol Esters/genetics , Cricetinae , Cricetulus , Humans , Lipoproteins, HDL/genetics , Scavenger Receptors, Class B/genetics
9.
Arterioscler Thromb Vasc Biol ; 37(12): 2260-2270, 2017 12.
Article in English | MEDLINE | ID: mdl-29074589

ABSTRACT

OBJECTIVE: Reverse cholesterol transport comprises cholesterol efflux from ABCA1-expressing macrophages to apolipoprotein (apo) AI, giving nascent high-density lipoprotein (nHDL), esterification of nHDL-free cholesterol (FC), selective hepatic extraction of HDL lipids, and hepatic conversion of HDL cholesterol to bile salts, which are excreted. We tested this model by identifying the fates of nHDL-[3H]FC, [14C] phospholipid (PL), and [125I]apo AI in serum in vitro and in vivo. APPROACH AND RESULTS: During in vitro incubation of human serum, nHDL-[3H]FC and [14C]PL rapidly transfer to HDL and low-density lipoproteins (t1/2=2-7 minutes), whereas nHDL-[125I]apo AI transfers solely to HDL (t1/2<10 minutes) and to the lipid-free form (t1/2>480 minutes). After injection into mice, nHDL-[3H]FC and [14C]PL rapidly transfer to liver (t1/2=≈2-3 minutes), whereas apo AI clears with t1/2=≈460 minutes. The plasma nHDL-[3H]FC esterification rate is slow (0.46%/h) compared with hepatic uptake. PL transfer protein enhances nHDL-[14C]PL but not nHDL-[3H]FC transfer to cultured Huh7 hepatocytes. CONCLUSIONS: nHDL-FC, PL, and apo AI enter different pathways in vivo. Most nHDL-[3H]FC and [14C]PL are rapidly extracted by the liver via SR-B1 (scavenger receptor class B member 1) and spontaneous transfer; hepatic PL uptake is promoted by PL transfer protein. nHDL-[125I]apo AI transfers to HDL and to the lipid-free form that can be recycled to nHDL formation. Cholesterol esterification by lecithin:cholesterol acyltransferase is a minor process in nHDL metabolism. These findings could guide the design of therapies that better mobilize peripheral tissue-FC to hepatic disposal.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Apolipoprotein A-I/blood , Cholesterol, HDL/blood , High-Density Lipoproteins, Pre-beta/blood , ATP Binding Cassette Transporter 1/genetics , Animals , Biomarkers/blood , Cell Line , Cholesterol Esters/blood , Chromatography, Gel , Half-Life , Hepatocytes/metabolism , Humans , Kinetics , Liver/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Models, Biological , Particle Size , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Phospholipids/blood , Transfection
10.
Biochim Biophys Acta ; 1861(11): 1787-1795, 2016 11.
Article in English | MEDLINE | ID: mdl-27594697

ABSTRACT

Although human plasma high density lipoproteins (HDL) concentrations negatively correlate with atherosclerotic cardiovascular disease, underlying mechanisms are unknown. Thus, there is continued interest in HDL structure and functionality. Numerous plasma factors disrupt HDL structure while inducing the release of lipid free apolipoprotein (apo) AI. Given that HDL is an unstable particle residing in a kinetic trap, we tested whether HDL could be stabilized by acylation with acetyl and hexanoyl anhydrides, giving AcHDL and HexHDL respectively. Lysine analysis with fluorescamine showed that AcHDL and HexHDL respectively contained 11 acetyl and 19 hexanoyl groups. Tests with biological and physicochemical perturbants showed that HexHDL was more stable than HDL to perturbant-induced lipid free apo AI formation. Like the reaction of streptococcal serum opacity factor against HDL, the interaction of HDL with its receptor, scavenger receptor class B member 1 (SR-B1), removes CE from HDL. Thus, we tested and validated the hypothesis that selective uptake of HexHDL-[3H]CE by Chinese Hamster Ovary cells expressing SR-B1 is less than that of HDL-[3H]CE; thus, selective SR-B1 uptake of HDL-CE depends on HDL instability. However, in mice, plasma clearance, hepatic uptake and sterol secretion into bile were faster from HexHDL-[3H]CE than from HDL-[3H]CE. Collectively, our data show that acylation increases HDL stability and that the reaction of plasma factors with HDL and SR-B1-mediated uptake are reduced by increased HDL stability. In vivo data suggest that HexHDL promotes charge-dependent reverse cholesterol transport, by a mechanism that increases hepatic sterol uptake via non SR-B1 receptors, thereby increasing bile acid output.


Subject(s)
Lipoproteins, HDL/blood , Lysine/metabolism , Acylation , Animals , Bile/metabolism , CHO Cells , Cholesterol Esters/metabolism , Chromatography, Gel , Cricetinae , Cricetulus , Gallbladder/metabolism , Humans , Kinetics , Liver/metabolism , Mice, Inbred C57BL , Molecular Weight , Peptide Hydrolases , Phospholipid Transfer Proteins/metabolism , Protein Stability
11.
Biochim Biophys Acta ; 1861(3): 196-204, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26709142

ABSTRACT

Plasma high density lipoprotein-cholesterol (HDL-C) concentrations negatively correlate with atherosclerotic cardiovascular disease. HDL is thought to have several atheroprotective functions, which are likely distinct from the epidemiological inverse relationship between HDL-C levels and risk. Specifically, strategies that reduce HDL-C while promoting reverse cholesterol transport (RCT) may have therapeutic value. The major product of the serum opacity factor (SOF) reaction versus HDL is a cholesteryl ester (CE)-rich microemulsion (CERM), which contains apo E and the CE of ~400,000 HDL particles. Huh7 hepatocytes take up CE faster when delivered as CERM than as HDL, in part via the LDL-receptor (LDLR). Here we compared the final RCT step, hepatic uptake and subsequent intracellular processing to cholesterol and bile salts for radiolabeled HDL-, CERM- and LDL-CE by Huh7 cells and in vivo in C57BL/6J mice. In Huh7 cells, uptake from LDL was greater than from CERM (2-4X) and HDL (5-10X). Halftimes for [(14)C]CE hydrolysis were 3.0±0.2, 4.4±0.6 and 5.4±0.7h respectively for HDL, CERM and LDL-CE. The fraction of sterols secreted as bile acids was ~50% by 8h for all three particles. HDL, CERM and LDL-CE metabolism in mice showed efficient plasma clearance of CERM-CE, liver uptake and metabolism, and secretion as bile acids into the gall bladder. This work supports the therapeutic potential of the SOF reaction, which diverts HDL-CE to the LDLR, thereby increasing hepatic CE uptake, and sterol disposal as bile acids.


Subject(s)
Anticholesteremic Agents/pharmacology , Bile Acids and Salts/metabolism , Cholesterol Esters/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Peptide Hydrolases/pharmacology , Animals , Apolipoproteins E/metabolism , Cell Line, Tumor , Cholesterol, HDL/metabolism , Cholesterol, LDL/metabolism , Gene Expression Regulation , Humans , Hydrolysis , Kinetics , Lipid Metabolism/genetics , Mice , Mice, Inbred C57BL
12.
Biochemistry ; 55(41): 5845-5853, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27662183

ABSTRACT

Injection of streptococcal serum opacity factor (SOF) into mice reduces the plasma cholesterol level by ∼40%. In vitro, SOF converts high-density lipoproteins (HDLs) into multiple products, including a small HDL, neo HDL. In vitro, neo HDL accounts for ∼60% of the protein mass of the SOF reaction products; in vivo, the accumulated mass of neo HDL is <1% of that observed in vitro. To identify the underlying cause of this difference, we determined the fate of neo HDL in plasma in vitro and in vivo. Following incubation with HDL, neo HDL-PC rapidly transfers to HDL, giving a small remnant, which fuses with HDL. An increased level of SR-B1 expression in Huh7 hepatoma cells and a reduced level of LDLR expression in CHO cells had little effect on neo HDL-[3H]CE uptake. Thus, the dominant receptors for neo HDL uptake are not LDLR or SR-B1. The in vivo metabolic fates of neo HDL-[3H]CE and HDL-[3H]CE were different. Thirty minutes after the injection of neo HDL-[3H]CE and HDL-[3H]CE into mice, plasma [3H]CE counts were 40 and 53%, respectively, of injected counts, with 10 times more [3H]CE appearing in the livers of neo HDL-[3H]CE-injected than in those of HDL-[3H]CE-injected mice. These data support a model of neo HDL-[3H]CE clearance by two parallel pathways. At early post-neo HDL-[3H]CE injection times, some neo HDL is directly removed by the liver; the remainder transfers its PC to HDL, leaving a remnant that fuses with HDL, which is also hepatically removed more slowly. Given that SR-B1 and SOF both remove CE from HDL, this novel mechanism may also underlie the metabolism of remnants released by hepatocytes following selective SR-B1-mediated uptake of HDL-CE.


Subject(s)
Lipoproteins, HDL/biosynthesis , Liver/metabolism , Peptide Hydrolases/metabolism , Streptococcus/metabolism , Animals , Cell Line , Cricetinae , Humans , Lipoproteins, HDL/metabolism , Mice , Mice, Inbred C57BL
13.
Biochemistry ; 54(14): 2295-302, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25790332

ABSTRACT

The reaction of Streptococcal serum opacity factor (SOF) against plasma high-density lipoproteins (HDL) produces a large cholesteryl ester-rich microemulsion (CERM), a smaller neo HDL that is apolipoprotein (apo) AI-poor, and lipid-free apo AI. SOF is active versus both human and mouse plasma HDL. In vivo injection of SOF into mice reduces plasma cholesterol ∼40% in 3 h while forming the same products observed in vitro, but at different ratios. Previous studies supported the hypothesis that labile apo AI is required for the SOF reaction vs HDL. Here we further tested that hypothesis by studies of SOF against HDL from apo AI-null mice. When injected into apo AI-null mice, SOF reduced plasma cholesterol ∼35% in 3 h. The reaction of SOF vs apo AI-null HDL in vitro produced a CERM and neo HDL, but no lipid-free apo. Moreover, according to the rate of CERM formation, the extent and rate of the SOF reaction versus apo AI-null mouse HDL were less than that against wild-type (WT) mouse HDL. Chaotropic perturbation studies using guanidine hydrochloride showed that apo AI-null HDL was more stable than WT HDL. Human apo AI added to apo AI-null HDL was quantitatively incorporated, giving reconstituted HDL. Both SOF and guanidine hydrochloride displaced apo AI from the reconstituted HDL. These results support the conclusion that apo AI-null HDL is more stable than WT HDL because it lacks apo AI, a labile protein that is readily displaced by physicochemical and biochemical perturbations. Thus, apo AI-null HDL is less SOF-reactive than WT HDL. The properties of apo AI-null HDL can be partially restored to those of WT HDL by the spontaneous incorporation of human apo AI. It remains to be determined what other HDL functions are affected by apo AI deletion.


Subject(s)
Apolipoprotein A-I/chemistry , Lipoproteins, HDL/chemistry , Peptide Hydrolases/chemistry , Animals , Apolipoprotein A-I/genetics , Guanidine/chemistry , Humans , Lipoproteins, HDL/blood , Mice, Knockout , Peptide Hydrolases/metabolism
14.
Arterioscler Thromb Vasc Biol ; 31(8): 1834-41, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21597008

ABSTRACT

OBJECTIVE: Recombinant streptococcal serum opacity factor (rSOF) mediates the in vitro disassembly of human plasma high-density lipoprotein (HDL) into lipid-free apolipoprotein (apo) A-I, a neo-HDL that is cholesterol poor, and a cholesteryl ester-rich microemulsion (CERM) containing apoE. Given the occurrence of apoE on the CERM, we tested the hypothesis that rSOF injection into mice would reduce total plasma cholesterol clearance via apoE-dependent hepatic low-density lipoprotein receptors (LDLR). METHODS AND RESULTS: rSOF (4 µg) injection into wild-type C57BL/6J mice formed neo-HDL, CERM, and lipid-free apoA-I, as observed in vitro, and reduced plasma total cholesterol (-43%, t(1/2)=44±18 minutes) whereas control saline injections had a negligible effect. Similar experiments with apoE(-/-) and LDLR(-/-) mice reduced plasma total cholesterol ≈0% and 20%, respectively. rSOF was potent; injection of 0.18 µg of rSOF produced 50% of maximum reduction of plasma cholesterol 3 hours postinjection, corresponding to a ≈0.5-mg human dose. Most cholesterol was cleared hepatically (>99%), with rSOF treatment increasing clearance by 65%. CONCLUSIONS: rSOF injection into mice formed a CERM that was cleared via hepatic LDLR that recognize apoE. This reaction could provide an alternative mechanism for reverse cholesterol transport.


Subject(s)
Apolipoproteins E/metabolism , Cholesterol, HDL/blood , Peptide Hydrolases/administration & dosage , Receptors, LDL/metabolism , Animals , Apolipoprotein A-I/blood , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Bacterial Proteins/administration & dosage , Cholesterol Esters/blood , Humans , Liver/drug effects , Liver/metabolism , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Recombinant Proteins/administration & dosage , Tissue Distribution
15.
Nat Rev Cardiol ; 18(10): 712-723, 2021 10.
Article in English | MEDLINE | ID: mdl-33833449

ABSTRACT

Plasma HDL-cholesterol concentrations correlate negatively with the risk of atherosclerotic cardiovascular disease (ASCVD). According to a widely cited model, HDL elicits its atheroprotective effect through its role in reverse cholesterol transport, which comprises the efflux of cholesterol from macrophages to early forms of HDL, followed by the conversion of free cholesterol (FCh) contained in HDL into cholesteryl esters, which are hepatically extracted from the plasma by HDL receptors and transferred to the bile for intestinal excretion. Given that increasing plasma HDL-cholesterol levels by genetic approaches does not reduce the risk of ASCVD, the focus of research has shifted to HDL function, especially in the context of macrophage cholesterol efflux. In support of the reverse cholesterol transport model, several large studies have revealed an inverse correlation between macrophage cholesterol efflux to plasma HDL and ASCVD. However, other studies have cast doubt on the underlying reverse cholesterol transport mechanism: in mice and humans, the FCh contained in HDL is rapidly cleared from the plasma (within minutes), independently of esterification and HDL holoparticle uptake by the liver. Moreover, the reversibility of FCh transfer between macrophages and HDL has implicated the reverse process - that is, the transfer of FCh from HDL to macrophages - in the aetiology of increased ASCVD under conditions of very high plasma HDL-FCh concentrations.


Subject(s)
Atherosclerosis , Cholesterol , Lipoproteins, HDL , Animals , Atherosclerosis/epidemiology , Biological Transport , Cholesterol/metabolism , Humans , Lipoproteins, HDL/blood , Mice , Risk Assessment
16.
Biochemistry ; 49(50): 10656-65, 2010 Dec 21.
Article in English | MEDLINE | ID: mdl-21073165

ABSTRACT

It is expected that the attendant structural heterogeneity of human high-density lipoprotein (HDL) complexes is a determinant of its varied metabolic functions. To determine the structural heterogeneity of HDL, we determined major apolipoprotein stoichiometry profiles in human HDL. First, HDL was separated into two main populations, with and without apolipoprotein (apo) A-II, LpA-I and LpA-I/A-II, respectively. Each main population was further separated into six individual subfractions using size exclusion chromatography (SEC). Protein proximity profiles (PPPs) of major apolipoproteins in each individual subfraction was determined by optimally cross-linking apolipoproteins within individual particles with bis(sulfosuccinimidyl) suberate (BS(3)), a bifunctional cross-linker, followed by molecular mass determination by MALDI-MS. The PPPs of LpA-I subfractions indicated that the number of apoA-I molecules increased from two to three to four with an increase in the LpA-I particle size. On the other hand, the entire population of LpA-I/A-II demonstrated the presence of only two proximal apoA-I molecules per particle, while the number of apoA-II molecules varied from one dimeric apoA-II to two and then to three. For most of the PPPs described above, an additional population that contained a single molecule of apoC-III in addition to apoA-I and/or apoA-II was detected. Upon composition analyses of individual subpopulations, LpA-I/A-II exhibited comparable proportions for total protein (∼58%), phospholipids (∼21%), total cholesterol (∼16%), triglycerides (∼5%), and free cholesterol (∼4%) across subfractions. LpA-I components, on the other hand, showed significant variability. This novel information about HDL subfractions will form a basis for an improved understanding of particle-specific functions of HDL.


Subject(s)
Lipoproteins, HDL/chemistry , Apolipoprotein A-I/blood , Apolipoprotein A-I/chemistry , Apolipoprotein A-II/blood , Apolipoprotein A-II/chemistry , Chromatography, Gel , Electrophoresis , Humans , Lipoproteins, HDL/blood , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
17.
Biochemistry ; 49(45): 9866-73, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-20879789

ABSTRACT

Serum opacity factor (SOF), a virulence determinant of Streptococcus pyogenes, converts plasma high-density lipoproteins (HDL) to three distinct species: lipid-free apolipoprotein (apo) A-I, neo HDL, a small discoidal HDL-like particle, and a large cholesteryl ester-rich microemulsion (CERM) that contains the cholesterol esters (CE) of up to ∼400000 HDL particles and apo E as its major protein. Similar SOF reaction products are obtained with HDL, total plasma lipoproteins, and whole plasma. We hypothesized that hepatic uptake of CERM-CE via multiple apo E-dependent receptors would be faster than that of HDL-CE. We tested our hypothesis using human hepatoma cells and lipoprotein receptor-specific Chinese hamster ovary (CHO) cells. The uptake of [(3)H]CE by HepG2 and Huh7 cells from HDL after SOF treatment, which transfers >90% of HDL-CE to CERM, was 2.4 and 4.5 times faster, respectively, than from control HDL. CERM-[(3)H]CE uptake was inhibited by LDL and HDL, suggestive of uptake by both the LDL receptor (LDL-R) and scavenger receptor class B type I (SR-BI). Studies in CHO cells specifically expressing LDL-R and SR-BI confirmed CERM-[(3)H]CE uptake by both receptors. RAP and heparin inhibit CERM-[(3)H]CE but not HDL-[(3)H]CE uptake, thereby implicating LRP-1 and cell surface proteoglycans in this process. These data demonstrate that SOF treatment of HDL increases the rate of CE uptake via multiple hepatic apo E receptors. In so doing, SOF might increase the level of hepatic disposal of plasma cholesterol in a way that is therapeutically useful.


Subject(s)
Cholesterol, HDL/metabolism , Hepatocytes/metabolism , Peptide Hydrolases/pharmacology , Streptococcus pyogenes/metabolism , Animals , Boron Compounds/metabolism , CHO Cells/metabolism , Cell Culture Techniques , Cholesterol Esters/metabolism , Cholesterol, HDL/drug effects , Cricetinae , Cricetulus , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Kinetics , Liver/metabolism , Microscopy, Confocal , Streptococcus pyogenes/pathogenicity , Virulence
18.
Lipids ; 55(4): 299-307, 2020 07.
Article in English | MEDLINE | ID: mdl-32255209

ABSTRACT

Moderate alcohol consumption is associated with increased plasma high-density lipoprotein (HDL)-cholesterol concentrations and reduced risk for cardiovascular disease. Plasma cholesteryl ester transfer activity (CETA) mediates the exchange of HDL-cholesteryl ester (CE) for the triacylglycerol (TAG) of very-low-density lipoproteins. We compared the effects of oral challenges of Alcohol, saturated fat (SAT), and (Alcohol + SAT) on plasma CETA, cholesterol, nonesterified fatty acids (NEFA), and TAG among normo-triglyceridemic (NTG) and mildly hypertriglyceridemic (HTG) volunteers having a range of plasma TAG concentrations. The major changes were (1) CETA increased more after ingestion of SAT and (Alcohol + SAT) in the HTG group versus the NTG group; (2) after all three challenges, elevation of plasma TAG concentration persisted longer in the HTG versus NTG group. Plasma cholesterol was not affected by the three dietary challenges, while Alcohol increased NEFA more in the HTG group than the NTG group. Plasma TAG best predicted plasma CETA, suggesting that intestinally derived lipoproteins are acceptors of HDL-CE. Unexpectedly, ingestion of (Alcohol + SAT) reduced the strength of the correlation between plasma TAG and CETA, that is the effects of (SAT and Alcohol) on plasma CETA are not synergistic nor additive but rather mutually suppressive. The alcohol-mediated inhibition of CE-transfer to chylomicrons maintains a higher plasma HDL-cholesterol concentration, which is athero-protective, although the suppressive metabolite underlying this correlation could be acetate, the terminal alcohol metabolite, other factors, including CETA inhibitors, are also likely important.


Subject(s)
Alcohols/administration & dosage , Cholesterol Ester Transfer Proteins/blood , Cholesterol/blood , Fatty Acids/administration & dosage , Hypertriglyceridemia/blood , Triglycerides/blood , Adult , Aged , Alcohols/pharmacology , Fatty Acids/pharmacology , Fatty Acids, Nonesterified/blood , Female , Humans , Male , Middle Aged , Young Adult
19.
Biomolecules ; 10(7)2020 07 01.
Article in English | MEDLINE | ID: mdl-32630283

ABSTRACT

Ingestion of alcohol is associated with numerous changes in human energy metabolism, especially that of plasma lipids and lipoproteins. Regular moderate alcohol consumption is associated with reduced atherosclerotic cardiovascular disease (ASCVD), an effect that has been attributed to the concurrent elevations of plasma high-density lipoprotein-cholesterol (HDL-C) concentrations. More recent evidence has accrued against the hypothesis that raising plasma HDL concentrations prevents ASCVD so that other metabolic processes associated with alcohol consumption have been considered. This review explored the roles of other metabolites induced by alcohol consumption-triglyceride-rich lipoproteins, non-esterified free fatty acids, and acetate, the terminal alcohol metabolite in athero-protection: Current evidence suggests that acetate has a key role in athero-protection but additional studies are needed.


Subject(s)
Alcohol Drinking/blood , Atherosclerosis/prevention & control , Lipoproteins, HDL/blood , Acetates/blood , Atherosclerosis/blood , Energy Metabolism/drug effects , Humans
20.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1865(12): 158794, 2020 12.
Article in English | MEDLINE | ID: mdl-32810603

ABSTRACT

OBJECTIVE: Apolipoprotein A1 (APOA1) is essential to reverse cholesterol transport, a physiologically important process that protects against atherosclerotic cardiovascular disease. APOA1 is a 28 kDa protein comprising multiple lipid-binding amphiphatic helices initialized by proline residues, which are conserved across multiple species. We tested the hypothesis that the evolutionarily conserved residues are essential to high density lipoprotein (HDL) function. APPROACH: We used biophysical and physiological assays of the function of APOA1P➔A variants, i.e., rHDL formation via dimyristoylphosphatidylcholine (DMPC) microsolubilization, activation of lecithin: cholesterol acyltransferase, cholesterol efflux from human monocyte-derived macrophages (THP-1) to each variant, and comparison of the size and composition of HDL from APOA1-/- mice receiving adeno-associated virus delivery of each human variant. RESULTS: Differences in microsolubilization were profound and showed that conserved prolines, especially those in the C-terminus of APOA1, are essential to efficient rHDL formation. In contrast, P➔A substitutions produced small changes (-25 to +25%) in rates of cholesterol efflux and no differences in the rates of LCAT activation. The HDL particles formed following ectopic expression of each variant in APOA1-/- mice were smaller and more heterogeneous than those from control animals. CONCLUSION: Studies of DMPC microsolubilization show that proline residues are essential to the optimal interaction of APOA1 with membranes, the initial step in cholesterol efflux and HDL production. In contrast, P➔A substitutions modestly reduce the cholesterol efflux capacity of APOA1, have no effect on LCAT activation, but according to the profound reduction in the size of HDL formed in vivo, P➔A substitutions alter HDL biogenesis, thereby implicating other cellular and in vivo processes as determinants of HDL metabolism and function.


Subject(s)
Apolipoprotein A-I/metabolism , Lipoproteins, HDL/metabolism , Amino Acid Sequence , Animals , Apolipoprotein A-I/chemistry , Cells, Cultured , Cholesterol/metabolism , Conserved Sequence , Humans , Mice , Models, Molecular
SELECTION OF CITATIONS
SEARCH DETAIL