Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 204
Filter
1.
J Zhejiang Univ Sci B ; 22(12): 1034-1044, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34904415

ABSTRACT

Apolipoprotein A-I (ApoA-I), the main protein component of high-density lipoprotein (HDL), plays a pivotal role in reverse cholesterol transport (RCT). Previous studies indicated a reduction of serum ApoA-I levels in various types of cancer, suggesting ApoA-I as a potential cancer biomarker. Herein, ectopically overexpressed ApoA-I in MDA-MB-231 breast cancer cells was observed to have antitumor effects, inhibiting cell proliferation and migration. Subsequent studies on the mechanism of expression regulation revealed that estradiol (E2)/estrogen receptor α (ERα) signaling activates ApoA-I gene transcription in breast cancer cells. Mechanistically, our ChIP-seq data showed that ERα directly binds to the estrogen response element (ERE) site within the ApoA-I gene and establishes an acetylation of histone 3 lysine 27 (H3K27ac)|-enriched chromatin microenvironment. Conversely, Fulvestrant (ICI 182780) treatment blocked ERα binding to ERE within the ApoA-I gene and downregulated the H3K27ac level on the ApoA-I gene. Treatment with p300 inhibitor also significantly decreased the ApoA-I messenger RNA (mRNA) level in MCF7 cells. Furthermore, the analysis of data from The Cancer Genome Atlas (TCGA) revealed a positive correlation between ERα and ApoA-I expression in breast cancer tissues. Taken together, our study not only revealed the antitumor potential of ApoA-I at the cellular level, but also found that ERα promotes the transcription of ApoA-I gene through direct genomic effects, and p300 may act as a co-activator of ERα in this process.


Subject(s)
Apolipoprotein A-I/genetics , Breast Neoplasms/metabolism , Chromatin/metabolism , Estrogen Receptor alpha/physiology , Genes, Tumor Suppressor/physiology , Tumor Microenvironment , Acetylation , Apolipoprotein A-I/physiology , Breast Neoplasms/genetics , Cell Line, Tumor , E1A-Associated p300 Protein/physiology , Exons , Female , Histones/metabolism , Humans
3.
Yakugaku Zasshi ; 140(2): 153-157, 2020.
Article in Japanese | MEDLINE | ID: mdl-32009037

ABSTRACT

Atherosclerosis is a vascular disease responsible for acute heart attacks and stroke, which are leading causes of death not only in industrialized countries but also worldwide, and the number of patients afflicted by this disease has been increasing in Japan. High-density lipoprotein (HDL) is the plasma lipoprotein that carries what is often called your "good cholesterol" through the blood. This good cholesterol moniker is associated with HDL because higher circulating levels of this lipoprotein are associated with a well-known reduction in the risk of arteriosclerosis. Moreover, many protective mechanisms by which HDL could reduce atherosclerosis are described, including reverse cholesterol transport, along with anti-oxidant, anti-inflammatory and anti-thrombosis activities. However, HDL-modulating therapies to lower cardiovascular risk are not yet available. It has recently been proposed that apolipoprotein A-I (apoA-I) binding protein (AIBP) enhances HDL function by accelerating lipid release from cells and reducing associated inflammatory processes. In this context, our research is focused on the function of HDL-related proteins, such as proteins that regulate HDL production (ATP-binding cassette transporters), and HDL-binding proteins. We expect that these studies could eventually help in the development of HDL-related prognostic and therapeutic strategies to reduce the burden of cardiovascular disease in the future.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Atherosclerosis , Cholesterol, HDL/metabolism , Drug Development , Apolipoprotein A-I/physiology , Atherosclerosis/prevention & control , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Humans , Protein Binding
4.
Lipids Health Dis ; 18(1): 223, 2019 Dec 16.
Article in English | MEDLINE | ID: mdl-31842884

ABSTRACT

Obesity is the most common nutritional disorder worldwide and is associated with dyslipidemia and atherosclerotic cardiovascular disease. The hallmark of dyslipidemia in obesity is low high density lipoprotein (HDL) cholesterol (HDL-C) levels. Moreover, the quality of HDL is also changed in the obese setting. However, there are still some disputes on the explanations for this phenomenon. There is increasing evidence that adipose tissue, as an energy storage tissue, participates in several metabolism activities, such as hormone secretion and cholesterol efflux. It can influence overall reverse cholesterol transport and plasma HDL-C level. In obesity individuals, the changes in morphology and function of adipose tissue affect plasma HDL-C levels and HDL function, thus, adipose tissue should be the main target for the treatment of HDL metabolism in obesity. In this review, we will summarize the cross-talk between adipocytes and HDL related to cardiovascular disease and focus on the new insights of the potential mechanism underlying obesity and HDL dysfunction.


Subject(s)
Adipocytes/physiology , Adipose Tissue/physiopathology , Atherosclerosis/etiology , Dyslipidemias/etiology , Lipoproteins, HDL/physiology , Obesity/complications , Animals , Apolipoprotein A-I/physiology , Atherosclerosis/physiopathology , Atherosclerosis/prevention & control , Cholesterol, HDL/blood , Dyslipidemias/physiopathology , Female , Humans , Lipolysis , Male
5.
Biochimie ; 165: 210-218, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31401190

ABSTRACT

Dysfunction in lipid metabolism may result in a decrease in hepatic autophagy, which contributes to the pathogenesis of non-alcoholic steatohepatitis. ATP-binding cassette transporter A1 transports free cholesterol and phospholipids to apolipoprotein A-I (apoA-I) to form nascent high-density lipoprotein particles. Results from previous studies showed that the overexpression of apoA-I significantly reduced levels of hepatic lipids and endoplasmic reticulum stress by modifying lipid transport. Here, we investigated the effects of apoA-I overexpression on hepatic autophagy in cultured hepatocytes and mice. The overexpression of apoA-I in HepG2 cells resulted in an increase in the levels of autophagy as well as the phosphorylation of AMP-activated protein kinase α (AMPKα) and ULK1 and a decrease in the phosphorylation of mammalian target of rapamycin (mTOR). An AMPK inhibitor and siRNA eliminated this apoA-I effect. Consistently, apoA-I transgenic mice showed increased autophagy and AMPKα phosphorylation. These results suggest that apoA-I overexpression alleviates steatohepatitis by increasing hepatic autophagy through the AMPK-mTOR-ULK1 pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apolipoprotein A-I/physiology , Autophagy-Related Protein-1 Homolog/metabolism , Fatty Liver/metabolism , Hepatocytes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Autophagy , Hep G2 Cells , Hepatocytes/cytology , Humans , Lipid Metabolism , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
6.
Gene ; 684: 76-81, 2019 Feb 05.
Article in English | MEDLINE | ID: mdl-30367981

ABSTRACT

BACKGROUND AND PURPOSES: Stroke is a leading cause of death and serious disability worldwide. Now, evidences indicate that dyslipidemia may play an important role in stroke. APOA1 and APOA5 involve in lipid metabolism. In this study, we investigated the association of APOA1 rs670 and APOA5 rs662799 with different stroke subtypes in the Han Chinese population of Taiwan. METHODS: A total of 1751 participants, including 459 control subjects, 606 large artery atherosclerosis (LAA), 339 small vessel occlusion (SVO), and 347 hypertensive intracranial hemorrhage (HICH), were enrolled. The presence of rs670 and rs662799 was analyzed through polymerase chain react ion and matrix-assisted laser desorption/ionization-time-of-flight-mass spectrometry. RESULTS: Notably, the frequency of the rs662799 C allele was significantly lower in the SVO patients than in the controls (24.36% vs. 29.74%, P = 0.024). The frequencies of heterozygote TC [odd ratio (OR) = 0.732, 95% confidence interval (CI) = 0.544-0.984, P = 0.038] and TC + CC (OR = 0.719, 95% CI = 0.542-0.953, P = 0.022) genotypes were significantly lower in the SVO patients than in the controls. In addition, triglyceride levels in individuals carrying the rs662799 TC + CC genotype were significantly higher than in those carrying the TT genotype, especially in older age, female, and body mass index (BMI) ≥ 25 groups. On the contrary, the low-density lipoprotein-cholesterol (LDL-C) was significantly lower in rs662799 TC + CC genotype than TT genotype. The BMI was significantly lower in subjects with rs662799 TC + CC genotype than those with TT genotype, especially in older age and female. High-density lipoprotein-cholesterol (HDL-C) levels were higher in individuals carrying the rs670 GG genotype than in those carrying the AG + AA genotype, especially in BMI < 25 group. Logistic regression analysis showed that the rs662799 C allele (TC + CC) was an independent protective factor for SVO after adjustment for conventional risk factors (OR = 0.709, 95% CI = 0.526-0.956; P = 0.024). CONCLUSION: GG genotype of rs670 is correlated with high serum HDL-C levels, whereas TC + CC genotype of rs662799 is associated with high serum triglyceride and low LDL and BMI levels. In addition, the rs662799 C allele (TC + CC) is an independent protective factor for SVO in the Han Chinese population in Taiwan.


Subject(s)
Apolipoprotein A-I/genetics , Apolipoprotein A-V/genetics , Stroke/genetics , Aged , Alleles , Apolipoprotein A-I/physiology , Apolipoprotein A-V/metabolism , Apolipoproteins A/genetics , Asian People/genetics , Case-Control Studies , Ethnicity/genetics , Female , Gene Frequency/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Risk Factors , Stroke/classification , Taiwan
7.
Int J Mol Sci ; 19(7)2018 07 06.
Article in English | MEDLINE | ID: mdl-29986413

ABSTRACT

High-density lipoprotein (HDL) particles have experienced a turbulent decade of falling from grace with widespread demotion from the most-sought-after therapeutic target to reverse cardiovascular disease (CVD), to mere biomarker status. HDL is slowly emerging from these dark times due to the HDL flux hypothesis wherein measures of HDL cholesterol efflux capacity (CEC) are better predictors of reduced CVD risk than static HDL-cholesterol (HDL-C) levels. HDL particles are emulsions of metabolites, lipids, protein, and microRNA (miR) built on the backbone of Apolipoprotein A1 (ApoA1) that are growing in their complexity due to the higher sensitivity of the respective "omic" technologies. Our understanding of particle composition has increased dramatically within this era and has exposed how our understanding of these particles to date has been oversimplified. Elucidation of the HDL proteome coupled with the identification of specific miRs on HDL have highlighted the "hormonal" characteristics of HDL in that it carries and delivers messages systemically. HDL can dock to most peripheral cells via its receptors, including SR-B1, ABCA1, and ABCG1, which may be a critical step for facilitating HDL-to-cell communication. The composition of HDL particles is, in turn, altered in numerous disease states including diabetes, auto-immune disease, and CVD. The consequence of changes in composition, however, on subsequent biological activities of HDL is currently poorly understood and this is an important avenue for the field to explore in the future. Improving HDL particle quality as opposed to HDL quantity may, in turn, prove a more beneficial investment to reduce CVD risk.


Subject(s)
Cardiovascular Diseases/metabolism , Cholesterol, HDL/metabolism , Inflammation/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoprotein A-I/physiology , Biomarkers , Cholesterol, HDL/chemistry , Humans , Metabolome , MicroRNAs , Models, Animal , Proteome , Risk Factors
8.
Arch Gynecol Obstet ; 298(2): 405-413, 2018 08.
Article in English | MEDLINE | ID: mdl-29938347

ABSTRACT

PURPOSE: To evaluate (a) the properties of high-density lipoproteins (HDL)/cholesterol, which include apolipoprotein A-1 (ApoA1) and paraoxonase1 (PON1), both are negative predictors of cardiovascular risk and (b) HDL function, among women with preeclampsia (PE). PE is a multi-system disorder, characterized by onset of hypertension and proteinuria or other end-organ dysfunction in the second half of pregnancy. Preeclampsia is associated with increased risk for later cardiovascular disease. The inverse association between HDL, cholesterol levels and the risk of developing atherosclerotic cardiovascular disease is well-established. METHODS: Twenty-five pregnant women [19 with PE and 6 with normal pregnancy (NP)] were recruited during admission for delivery. HDL was isolated from blood samples. PON1 activity and HDL were analyzed. An in vitro model of endothelial cells was used to evaluate the effect of HDL on the transcription response of vascular cell adhesion molecule-1 (VCAM-1) and endothelial nitric oxide synthase (eNOS) mRNA expression. RESULTS: PON1 activity (units/ml serum) was lower in the PE group compared to normal pregnancy (NP) (6.51 ± 0.73 vs. 9.98 ± 0.54; P = 0.015). Increased ApoA1 was released from PE-HDL as compared to NP-HDL (3.54 ± 0.72 vs. 0.89 ± 0.35; P = 0.01). PE-HDL exhibited increased VCAM-1 mRNA expression and decreased eNOS mRNA expression on TNF-α stimulated endothelial cells as compared to NP-HDL. CONCLUSIONS: HDL from women with PE reduced PON1 activity and increased ApoA1 release from HDL particles. This process was associated with increased HDL diameter, suggesting impaired HDL anti-oxidant activity. These changes might contribute to higher long-term cardiovascular risks among women with PE.


Subject(s)
Apolipoprotein A-I/metabolism , Aryldialkylphosphatase/metabolism , Lipoproteins, HDL/physiology , Pre-Eclampsia/metabolism , Adult , Apolipoprotein A-I/physiology , Aryldialkylphosphatase/physiology , Case-Control Studies , Cholesterol/metabolism , Female , Gene Expression Regulation , Humans , Hypertension/metabolism , Lipoproteins, HDL/blood , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Pre-Eclampsia/blood , Pre-Eclampsia/physiopathology , Pregnancy , Proteinuria/metabolism , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
9.
Biomed Khim ; 64(6): 463-471, 2018 Nov.
Article in Russian | MEDLINE | ID: mdl-30632974

ABSTRACT

Disturbance in lipid metabolism can be both a cause and a consequence of the development of diabetes mellitus (DM). One of the most informative indicator of lipid metabolism is the ratio of atherogenic and antiatherogenic fractions of lipoproteins and their protein components. The review summarizes literature data and own results indicating the important role of high-density lipoprotein (HDL) and their main protein component, apolipoprotein A-I (apoA-I), in the pathogenesis of type 2 DM. On the one hand, HDL are involved in the regulation of insulin secretion by b-cells and insulin-independent absorption of glucose. On the other hand, insulin resistance and hyperglycemia lead to a decrease in HDL levels and cause modification of their protein component. In addition, HDL, possessing anti-inflammatory and mitogenic properties, provide anti-diabetic protection through systemic mechanisms. Thus, maintaining a high concentration of HDL and apoA-I in blood plasma and preventing their modification are important issues in the context of prevention and treatment of diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Lipoproteins, HDL/physiology , Apolipoprotein A-I/physiology , Humans , Insulin
10.
J Biol Chem ; 292(49): 20313-20327, 2017 12 08.
Article in English | MEDLINE | ID: mdl-29030428

ABSTRACT

Lecithin:cholesterol acyltransferase (LCAT) plays a key role in reverse cholesterol transport by transferring an acyl group from phosphatidylcholine to cholesterol, promoting the maturation of high-density lipoproteins (HDL) from discoidal to spherical particles. LCAT is activated through an unknown mechanism by apolipoprotein A-I (apoA-I) and other mimetic peptides that form a belt around HDL. Here, we report the crystal structure of LCAT with an extended lid that blocks access to the active site, consistent with an inactive conformation. Residues Thr-123 and Phe-382 in the catalytic domain form a latch-like interaction with hydrophobic residues in the lid. Because these residues are mutated in genetic disease, lid displacement was hypothesized to be an important feature of apoA-I activation. Functional studies of site-directed mutants revealed that loss of latch interactions or the entire lid enhanced activity against soluble ester substrates, and hydrogen-deuterium exchange (HDX) mass spectrometry revealed that the LCAT lid is extremely dynamic in solution. Upon addition of a covalent inhibitor that mimics one of the reaction intermediates, there is an overall decrease in HDX in the lid and adjacent regions of the protein, consistent with ordering. These data suggest a model wherein the active site of LCAT is shielded from soluble substrates by a dynamic lid until it interacts with HDL to allow transesterification to proceed.


Subject(s)
Apolipoprotein A-I/physiology , Phosphatidylcholine-Sterol O-Acyltransferase/chemistry , Catalytic Domain , Crystallography, X-Ray , Deuterium Exchange Measurement , Enzyme Activation , Humans , Lipoproteins, HDL/metabolism , Mutagenesis, Site-Directed , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Protein Conformation
11.
Neurobiol Aging ; 60: 116-128, 2017 12.
Article in English | MEDLINE | ID: mdl-28941727

ABSTRACT

Beyond the crucial role of apolipoprotein A-I (ApoA-I) on peripheral cholesterol metabolism, this apolipoprotein has also been implicated in beta amyloid (Aß)-related neuropathologies. ApoA-I-Milano (M) is a mutated variant, which showed increased vasoprotective properties compared to ApoA-I-wild type in models of atherosclerosis and cardiovascular damage. We speculated that ApoA-I-M may also protect Aß-affected vasculature and reverse some of the pathological features associated with Alzheimer's disease (AD). For this purpose, we produced and characterized human recombinant ApoA-I-wild type and ApoA-I-M proteins. Both of them were able to avoid the aggregation of Aß in vitro, even though recombinant ApoA-I-M was significantly more effective in protecting endothelial cells from Aß(1-42)-toxicity. Next, we determined the effect of chronic intravenous administration of rApoA-I-M in the APP23-transgenic mouse model of AD. We found reduced cerebral Aß levels in mice that received rApoA-I-M, which were accompanied by a lower expression of astrocyte and microglia neuroinflammatory markers. Our results suggest an applicability of this molecule as a therapeutic candidate for protecting the brain in AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoprotein A-I/administration & dosage , Brain/metabolism , Animals , Apolipoprotein A-I/pharmacology , Apolipoprotein A-I/physiology , Disease Models, Animal , Infusions, Intravenous , Mice, Transgenic , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology
12.
J Endocrinol ; 233(2): R95-R107, 2017 05.
Article in English | MEDLINE | ID: mdl-28314771

ABSTRACT

It is well appreciated that high-density lipoprotein (HDL) and bone physiology and pathology are tightly linked. Studies, primarily in mouse models, have shown that dysfunctional and/or disturbed HDL can affect bone mass through many different ways. Specifically, reduced HDL levels have been associated with the development of an inflammatory microenvironment that affects the differentiation and function of osteoblasts. In addition, perturbation in metabolic pathways of HDL favors adipoblastic differentiation and restrains osteoblastic differentiation through, among others, the modification of specific bone-related chemokines and signaling cascades. Increased bone marrow adiposity also deteriorates bone osteoblastic function and thus bone synthesis, leading to reduced bone mass. In this review, we present the current knowledge and the future directions with regard to the HDL-bone mass connection. Unraveling the molecular phenomena that underline this connection will promote the deeper understanding of the pathophysiology of bone-related pathologies, such as osteoporosis or bone metastasis, and pave the way toward the development of novel and more effective therapies against these conditions.


Subject(s)
Bone and Bones/physiology , Lipoproteins, HDL/physiology , Adiposity , Animals , Apolipoprotein A-I/physiology , Apolipoproteins/physiology , Apolipoproteins E/physiology , Bone Density , Bone Marrow/physiology , Bone and Bones/metabolism , Cartilage/physiology , Cell Differentiation , Disease Models, Animal , Homeostasis , Humans , Lipoproteins, HDL/metabolism , Mice , Obesity , Osteoblasts/physiology , Osteoclasts/physiology , Osteogenesis
13.
Biochem Biophys Res Commun ; 484(2): 403-408, 2017 03 04.
Article in English | MEDLINE | ID: mdl-28137586

ABSTRACT

Saturated fatty acids (SFAs) activate toll-like receptor 4 (TLR4) signal transduction in macrophages and are involved in the chronic inflammation accompanying obesity. High-density lipoprotein (HDL) and apolipoprotein A-I (apoA-I) produce anti-inflammatory effects via reverse cholesterol transport. However, the underlying mechanisms by which HDL and apoA-I inhibit inflammatory responses in adipocytes remain to be determined. Here we examined whether palmitate increases the translocation of TLR4 into lipid rafts and whether HDL and apoA-I inhibit inflammation in adipocytes. Palmitate exposure (250 µM, 24 h) increased interleukin-6 and tumor necrosis factor-α gene expressions and translocation of TLR4 into lipid rafts in 3T3-L1 adipocytes. Pretreatment with HDL and apoA-I (50 µg/mL, 6 h) suppressed palmitate-induced inflammatory cytokine expression and TLR4 translocation into lipid rafts. Moreover, HDL and apoA-I inhibited palmitate-induced phosphorylation of nuclear factor-kappa B. HDL showed an anti-inflammatory effect via ATP-binding cassette transporter G1 and scavenger receptor class B, member 1, whereas apoA-I showed an effect via ATP-binding cassette transporter A1. These results demonstrated that HDL and apoA-I reduced palmitate-potentiated TLR4 trafficking into lipid rafts and its related inflammation in adipocytes via these specific transporters.


Subject(s)
Apolipoprotein A-I/physiology , Cytokines/metabolism , Inflammation Mediators/metabolism , Lipoproteins, HDL/physiology , Membrane Microdomains/metabolism , Palmitates/pharmacology , Toll-Like Receptor 4/metabolism , 3T3-L1 Cells , Animals , Mice , Protein Transport
14.
Reprod Biomed Online ; 33(5): 635-645, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27567428

ABSTRACT

It is known that apolipoprotein A1 (apoA1) is a stimulator of endothelial nitric oxide synthase (eNOS), and that heterogeneous nuclear ribonucleoprotein E1 (hnRNP-E1)-containing RNP complexes is a key protector of basal stabilization of eNOS mRNA. Recently, we found that apoA1 and hnRNP-E1 were up-regulated during peri-implantation period, and the purpose of this study was to explore the roles of apoA1 and hnRNP-E1 during this period in the mouse. It was found that the up-regulation of apoA1 and hnRNP-E1 were dependent on the presence and status of blastocysts, on endometrial decidualization and on the progesterone and 17ß-oestradiol status. Knockdown of apoA1 or hnRNP-E1 both resulted in reduced numbers of embryo implantations and neonates (P < 0.01), and lipid peroxidation was found to be involved. On pregnancy day 5 eNOS expression and superoxidase dismutase (SOD) quantity were increased, and malondialdehyde (MDA) quantity was decreased at implantation sites. The knockdown of either apoA1 or hnRNP-E1 led to down-regulation of eNOS (P < 0.01) and to an increase in the quantity of MDA (P < 0.05), and a decrease in the amount of SOD (P < 0.01). These findings suggest that apoA1 and hnRNP-E1 may play roles in embryo implantation by inhibiting lipid peroxidation.


Subject(s)
Apolipoprotein A-I/physiology , Embryo Implantation/genetics , Heterogeneous-Nuclear Ribonucleoproteins/physiology , Lipid Peroxidation/genetics , Animals , Apolipoprotein A-I/genetics , Endometrium/metabolism , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism
15.
Biochemistry ; 55(27): 3752-62, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27332083

ABSTRACT

In addition to high-density lipoprotein cholesterol (HDL-C) levels, HDL quality also appears to be very important for atheroprotection. Analysis of various clinical paradigms suggests that the lipid and apolipoprotein composition of HDL defines its size, shape, and functions and may determine its beneficial effects on human health. Previously, we reported that like apolipoprotein A-I (Apoa1), apolipoprotein E (Apoe) is also capable of promoting the de novo biogenesis of HDL with the participation of ATP binding cassette A lipid transporter member 1 (Abca1) and plasma enzyme lecithin:cholesterol acyltransferase (Lcat), in a manner independent of a functional Apoa1. Here, we performed a comparative analysis of the functions of these HDL subpopulations. Specifically, Apoe and Apoa1 double-deficient (Apoe(-/-) × Apoa1(-/-)) mice were infected with APOA1- or APOE3-expressing adenoviruses, and APOA1-containing HDL (APOA1-HDL) and APOE3-containing HDL (APOE3-HDL), respectively, were isolated and analyzed by biochemical and physicochemical methods. Western blot and lipidomic analyses indicated significant differences in the apolipoprotein and lipid composition of the two HDL species. Moreover APOE3-HDL presented a markedly reduced antioxidant potential and Abcg1-mediated cholesterol efflux capacity. Surprisingly, APOE3-HDL but not APOA1-HDL attenuated LPS-induced production of TNFα in RAW264.7 cells, suggesting that the anti-inflammatory effects of APOA1 are dependent on APOE expression. Taken together, our data indicate that APOA1 and APOE3 recruit different apolipoproteins and lipids on the HDL particle, leading to structurally and functionally distinct HDL subpopulations. The distinct role of these two apolipoproteins in the modulation of HDL functionality may pave the way toward the development of novel pharmaceuticals that aim to improve HDL functionality.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apolipoprotein A-I/physiology , Apolipoproteins E/physiology , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/pharmacology , Animals , Blotting, Western , Cells, Cultured , Cholesterol/metabolism , Female , Humans , Lipids/blood , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Exp Biol Med (Maywood) ; 241(17): 1933-1942, 2016 11.
Article in English | MEDLINE | ID: mdl-27354333

ABSTRACT

In sickle cell disease (SCD) cholesterol metabolism appears dysfunctional as evidenced by abnormal plasma cholesterol content in a subpopulation of SCD patients. Specific activity of the high density lipoprotein (HDL)-bound lecithin cholesterol acyltransferase (LCAT) enzyme, which catalyzes esterification of cholesterol, and generates lysoPC (LPC) was significantly lower in sickle plasma compared to normal. Inhibitory amounts of LPC were present in sickle plasma, and the red blood cell (RBC) lysophosphatidylcholine acyltransferase (LPCAT), essential for the removal of LPC, displayed a broad range of activity. The functionality of sickle HDL appeared to be altered as evidenced by a decreased HDL-Apolipoprotein A-I exchange in sickle plasma as compared to control. Increased levels of oxidized proteins including ApoA-I were detected in sickle plasma. In vitro incubation of sickle plasma with washed erythrocytes affected the ApoA-I-exchange supporting the view that the RBC blood compartment can affect cholesterol metabolism in plasma. HDL functionality appeared to decrease during acute vaso-occlusive episodes in sickle patients and was associated with an increase of secretory PLA2, a marker for increased inflammation. Simvastatin treatment to improve the anti-inflammatory function of HDL did not ameliorate HDL-ApoA-I exchange in sickle patients. Thus, the cumulative effect of an inflammatory and highly oxidative environment in sickle blood contributes to a decrease in cholesterol esterification and HDL function, related to hypocholesterolemia in SCD.


Subject(s)
Anemia, Sickle Cell/blood , Anemia, Sickle Cell/enzymology , Apolipoprotein A-I/physiology , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Adult , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/physiopathology , Biomarkers/blood , Case-Control Studies , Erythrocytes/chemistry , Fetal Hemoglobin/analysis , Humans , Phosphatidylcholine-Sterol O-Acyltransferase/physiology
17.
Front Biosci (Elite Ed) ; 8(3): 460-77, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27100352

ABSTRACT

Limited axonal regeneration after traumatic injuries to the CNS presents a challenge in neuroscience. Investigation of CSF from subjects with spinal cord injury (SCI) has found that the lipid catabolism pathway is implicated in the post injury scenario. Sequestration of the CNS by the blood brain barrier ensures a mechanism of cholesterol metabolism and recycling distinct from that in the peripheral tissues. Apolipoprotein A1, the protein component of high density lipoprotein (HDL), is an abundant protein in the mammalian cerebrospinal fluid. Interaction of ApoA1 with its cellular receptor, ABCA1, gives rise to several signaling events, such as the activation of Cdc42 protein leading to actin polymerisation. Emerging evidences suggest that ApoA1 mediates anti-inflammatory effects and conversely, is negatively regulated by inflammatory cytokines. Collating these findings, added to the clinical evidences of using HDL as a therapeutic target for cardio vascular diseases, we hypothesize that ApoA1 could be useful in neurite outgrowth after mechanical injury by 1) mediating polymerisation of actin and 2) restricting inflammatory responses after injury which are deleterious to healing.


Subject(s)
Apolipoprotein A-I/physiology , Neurons/pathology , Apolipoprotein A-I/metabolism , Axons/metabolism , Axons/physiology , Cell Death , Humans , Models, Theoretical , Nerve Regeneration , Neurons/metabolism , Neurons/physiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
18.
PLoS One ; 11(3): e0151767, 2016.
Article in English | MEDLINE | ID: mdl-26986486

ABSTRACT

OBJECTIVE: HDL and its apolipoproteins protect against atherosclerotic disease partly by removing excess cholesterol from macrophage foam cells. But the underlying mechanisms of cholesterol clearance are still not well defined. We investigated roles of vesicle trafficking of coatomer ß-COP in delivering cholesterol to the cell surface during apoA-1 and apoE-mediated lipid efflux from fibroblasts and THP-1 macrophages. METHODS: shRNA knockout, confocal and electron microscopy and biochemical analysis were used to investigate the roles of ß-COP in apolipoprotein-mediated cholesterol efflux in fibroblasts and THP-1 macrophages. RESULTS: We showed that ß-COP knockdown by lentiviral shRNA resulted in reduced apoA-1-mediated cholesterol efflux, while increased cholesterol accumulation and formation of larger vesicles were observed in THP-1 macrophages by laser scanning confocal microscopy. Immunogold electron microscopy showed that ß-COP appeared on the membrane protrusion complexes and colocalized with apoA-1 or apoE during cholesterol efflux. This was associated with releasing heterogeneous sizes of small particles into the culture media of THP-1 macrophage. Western blotting also showed that apoA-1 promotes ß-COP translocation to the cell membrane and secretion into culture media, in which a total of 17 proteins were identified by proteomics. Moreover, ß-COP exclusively associated with human plasma HDL fractions. CONCLUSION: ApoA-1 and apoE promoted transport vesicles consisting of ß-COP and other candidate proteins to exocytose cholesterol, forming the protrusion complexes on cell surface, which were then released from the cell membrane as small particles to media.


Subject(s)
Apolipoprotein A-I/physiology , Apolipoproteins E/physiology , Cholesterol/metabolism , Coatomer Protein/physiology , Exocytosis/physiology , Transport Vesicles/physiology , Blotting, Western , Cells, Cultured , Fibroblasts/metabolism , Gene Knockout Techniques , Humans , Macrophages/metabolism , Microscopy, Confocal , Microscopy, Electron , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transport Vesicles/metabolism
19.
PLoS One ; 10(12): e0144372, 2015.
Article in English | MEDLINE | ID: mdl-26680360

ABSTRACT

High density lipoprotein has anti-inflammatory effects in addition to mediating reverse cholesterol transport. While many of the chronic anti-inflammatory effects of high density lipoprotein (HDL) are attributed to changes in cell adhesion molecules, little is known about acute signal transduction events elicited by HDL in endothelial cells. We now show that high density lipoprotein decreases endothelial cell exocytosis, the first step in leukocyte trafficking. ApoA-I, a major apolipoprotein of HDL, mediates inhibition of endothelial cell exocytosis by interacting with endothelial scavenger receptor-BI which triggers an intracellular protective signaling cascade involving protein kinase C (PKC). Other apolipoproteins within the HDL particle have only modest effects upon endothelial exocytosis. Using a human primary culture of endothelial cells and murine apo-AI knockout mice, we show that apo-AI prevents endothelial cell exocytosis which limits leukocyte recruitment. These data suggest that high density lipoprotein may inhibit diseases associated with vascular inflammation in part by blocking endothelial exocytosis.


Subject(s)
Inflammation/prevention & control , Lipoproteins, HDL/physiology , Weibel-Palade Bodies , Apolipoprotein A-I/physiology , Cell Adhesion/physiology , Cells, Cultured , Exocytosis , Humans
20.
Mol Cells ; 38(6): 573-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25997739

ABSTRACT

Apolipoprotein A-I and A-IV are protein constituents of high-density lipoproteins although their functional difference in lipoprotein metabolism is still unclear. To compare anti-atherogenic properties between apoA-I and apoA-4, we characterized both proteins in lipid-free and lipid-bound state. In lipid-free state, apoA4 showed two distinct bands, around 78 and 67 Å on native gel electrophoresis, while apoA-I showed scattered band pattern less than 71 Å. In reconstituted HDL (rHDL) state, apoA-4 showed three major bands around 101 Å and 113 Å, while apoA-I-rHDL showed almost single band around 98 Å size. Lipid-free apoA-I showed 2.9-fold higher phospholipid binding ability than apoA-4. In lipid-free state, BS3-crosslinking revealed that apoA-4 showed less multimerization tendency upto dimer, while apoA-I showed pentamerization. In rHDL state (95:1), apoA-4 was existed as dimer as like as apoA-I. With higher phospholipid content (255:1), five apoA-I and three apoA-4 were required to the bigger rHDL formation. Regardless of particle size, apoA-I-rHDL showed superior LCAT activation ability than apoA-4-rHDL. Uptake of acetylated LDL was inhibited by apoA-I in both lipid-free and lipid-bound state, while apoA-4 inhibited it only lipid-free state. ApoA-4 showed less anti-atherogenic activity with more sensitivity to glycation. In conclusion, apoA-4 showed inferior physiological functions in lipid-bound state, compared with those of apoA-I, to induce more pro-atherosclerotic properties.


Subject(s)
Apolipoprotein A-I/chemistry , Apolipoprotein A-I/physiology , Apolipoproteins A/chemistry , Apolipoproteins A/physiology , Antioxidants/chemistry , Antioxidants/physiology , Apolipoprotein A-I/isolation & purification , Apolipoprotein A-I/pharmacology , Apolipoproteins A/isolation & purification , Apolipoproteins A/pharmacology , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Cell Line , Circular Dichroism , Humans , Lipoproteins, HDL/chemistry , Male , Phospholipids/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...