Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.547
Filter
1.
Elife ; 122024 May 24.
Article in English | MEDLINE | ID: mdl-38787378

ABSTRACT

Severe dengue infections are characterized by endothelial dysfunction shown to be associated with the secreted nonstructural protein 1 (sNS1), making it an attractive vaccine antigen and biotherapeutic target. To uncover the biologically relevant structure of sNS1, we obtained infection-derived sNS1 (isNS1) from dengue virus (DENV)-infected Vero cells through immunoaffinity purification instead of recombinant sNS1 (rsNS1) overexpressed in insect or mammalian cell lines. We found that isNS1 appeared as an approximately 250 kDa complex of NS1 and ApoA1 and further determined the cryoEM structures of isNS1 and its complex with a monoclonal antibody/Fab. Indeed, we found that the major species of isNS1 is a complex of the NS1 dimer partially embedded in a high-density lipoprotein (HDL) particle. Crosslinking mass spectrometry studies confirmed that the isNS1 interacts with the major HDL component ApoA1 through interactions that map to the NS1 wing and hydrophobic domains. Furthermore, our studies demonstrated that the sNS1 in sera from DENV-infected mice and a human patient form a similar complex as isNS1. Our results report the molecular architecture of a biological form of sNS1, which may have implications for the molecular pathogenesis of dengue.


Subject(s)
Dengue Virus , Dengue , Lipoproteins, HDL , Viral Nonstructural Proteins , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Animals , Dengue Virus/genetics , Dengue Virus/metabolism , Chlorocebus aethiops , Mice , Humans , Lipoproteins, HDL/metabolism , Vero Cells , Dengue/virology , Dengue/metabolism , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/chemistry , Protein Multimerization , Cryoelectron Microscopy
2.
Int J Mol Sci ; 25(10)2024 May 20.
Article in English | MEDLINE | ID: mdl-38791598

ABSTRACT

CIGB-258, a 3 kDa peptide from heat shock protein 60, exhibits synergistic anti-inflammatory activity with apolipoprotein A-I (apoA-I) in reconstituted high-density lipoproteins (rHDLs) via stabilization of the rHDL structure. This study explored the interactions between CIGB-258 and apoA-I in the lipid-free state to assess their synergistic effects in the structural and functional enhancement of apoA-I and HDL. A co-treatment of lipid-free apoA-I and CIGB-258 inhibited the cupric ion-mediated oxidation of low-density lipoprotein (LDL) and a lowering of oxidized species in the dose-responsive manner of CIGB-258. The co-presence of CIGB-258 caused a blue shift in the wavelength of maximum fluorescence (WMF) of apoA-I with protection from proteolytic degradation. The addition of apoA-I:CIGB-258, with a molar ratio of 1:0.1, 1:0.5, and 1:1, to HDL2 and HDL3 remarkably enhanced the antioxidant ability against LDL oxidation up to two-fold higher than HDL alone. HDL-associated paraoxonase activities were elevated up to 28% by the co-addition of apoA-I and CIGB-258, which is linked to the suppression of Cu2+-mediated HDL oxidation with the slowest electromobility. Isothermal denaturation by a urea treatment showed that the co-presence of CIGB-258 attenuated the exposure of intrinsic tryptophan (Trp) and increased the mid-points of denaturation from 2.33 M for apoA-I alone to 2.57 M for an apoA-I:CIGB-258 mixture with a molar ratio of 1:0.5. The addition of CIGB-258 to apoA-I protected the carboxymethyllysine (CML)-facilitated glycation of apoA-I with the prevention of Trp exposure. A co-treatment of apoA-I and CIGB-258 synergistically safeguarded zebrafish embryos from acute death by CML-toxicity, suppressing oxidative stress and apoptosis. In adult zebrafish, the co-treatment of apoA-I+CIGB-258 exerted the highest anti-inflammatory activity with a higher recovery of swimming ability and survivability than apoA-I alone or CIGB-258 alone. A co-injection of apoA-I and CIGB-258 led to the lowest infiltration of neutrophils and interleukin (IL)-6 generation in hepatic tissue, with the lowest serum triglyceride, aspartate transaminase, and alanine transaminase levels in plasma. In conclusion, the co-presence of CIGB-258 ameliorated the beneficial functionalities of apoA-I, such as antioxidant and anti-glycation activities, by enhancing the structural stabilization and protection of apoA-I. The combination of apoA-I and CIGB-258 synergistically enforced the anti-inflammatory effect against CML toxicity in embryos and adult zebrafish.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Apolipoprotein A-I , Lipoproteins, HDL , Zebrafish , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/chemistry , Animals , Antioxidants/pharmacology , Antioxidants/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Lipoproteins, HDL/metabolism , Lipoproteins, HDL/chemistry , Lipoproteins, LDL/metabolism , Oxidation-Reduction/drug effects , Drug Synergism
3.
J Am Chem Soc ; 146(22): 15096-15107, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38773940

ABSTRACT

For cationic nanoparticles, the spontaneous nanoparticle-protein corona formation and aggregation in biofluids can trigger unexpected biological reactions. Herein, we present a biomimetic strategy for camouflaging the cationic peptide/siRNA nanocomplex (P/Si) with single or dual proteins, which exploits the unique properties of endogenous proteins and stabilizes the cationic P/Si complex for safe and targeted delivery. An in-depth study of the P/Si protein corona (P/Si-PC) formation and protein binding was conducted. The results provided insights into the biochemical and toxicological properties of cationic nanocomplexes and the rationales for engineering biomimetic protein camouflages. Based on this, the human serum albumin (HSA) and apolipoprotein AI (Apo-AI) ranked within the top 20 abundant protein species of P/Si-PC were selected to construct biomimetic HSA-dressed P/Si (P/Si@HSA) and dual protein (HSA and Apo-AI)-dressed P/Si (P/Si@HSA_Apo), given that the dual-protein camouflage plays complementary roles in efficient delivery. A branched cationic peptide (b-HKR) was tailored for siRNA delivery, and their nanocomplexes, including the cationic P/Si and biomimetic protein-dressed P/Si, were produced by a precise microfluidic technology. The biomimetic anionic protein camouflage greatly enhanced P/Si biostability and biocompatibility, which offers a reliable strategy for overcoming the limitation of applying cationic nanoparticles in biofluids and systemic delivery.


Subject(s)
Biomimetic Materials , Nanoparticles , Peptides , RNA, Small Interfering , Serum Albumin, Human , Humans , RNA, Small Interfering/chemistry , Peptides/chemistry , Biomimetic Materials/chemistry , Nanoparticles/chemistry , Serum Albumin, Human/chemistry , Protein Engineering , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Protein Corona/chemistry , Biomimetics/methods
4.
PLoS Comput Biol ; 20(5): e1012137, 2024 May.
Article in English | MEDLINE | ID: mdl-38805510

ABSTRACT

Lecithin:cholesterol acyltransferase (LCAT) exhibits α-activity on high-density and ß-activity on low-density lipoproteins. However, the molecular determinants governing LCAT activation by different apolipoproteins remain elusive. Uncovering these determinants would offer the opportunity to design and explore advanced therapies against dyslipidemias. Here, we have conducted coarse-grained and all-atom molecular dynamics simulations of LCAT with nanodiscs made with α-helical amphiphilic peptides either derived from apolipoproteins A1 and E (apoA1 and apoE) or apoA1 mimetic peptide 22A that was optimized to activate LCAT. This study aims to explore what drives the binding of peptides to our previously identified interaction site in LCAT. We hypothesized that this approach could be used to screen for binding sites of LCAT in different apolipoproteins and would provide insights to differently localized LCAT activities. Our screening approach was able to discriminate apoA1 helixes 4, 6, and 7 as key contributors to the interaction with LCAT supporting the previous research data. The simulations provided detailed molecular determinants driving the interaction with LCAT: the formation of hydrogen bonds or salt bridges between peptides E4 or D4 and LCAT S236 or K238 residues. Additionally, salt bridging between R7 and D73 was observed, depending on the availability of R7. Expanding our investigation to diverse plasma proteins, we detected novel LCAT binding helixes in apoL1, apoB100, and serum amyloid A. Our findings suggest that the same binding determinants, involving E4 or D4 -S236 and R7-D73 interactions, influence LCAT ß-activity on low-density lipoproteins, where apoE and or apoB100 are hypothesized to interact with LCAT.


Subject(s)
Apolipoprotein A-I , Apolipoproteins , Molecular Dynamics Simulation , Phosphatidylcholine-Sterol O-Acyltransferase , Phosphatidylcholine-Sterol O-Acyltransferase/chemistry , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Binding Sites , Apolipoproteins/chemistry , Apolipoproteins/metabolism , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/metabolism , Humans , Peptides/chemistry , Peptides/metabolism , Nanostructures/chemistry , Protein Binding , Apolipoproteins E/chemistry , Apolipoproteins E/metabolism
5.
Vet Microbiol ; 294: 110106, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38776767

ABSTRACT

Glaesserella parasuis (G. parasuis) is the causative agent of porcine Glässer's disease, resulting in high mortality rates in pigs due to excessive inflammation-induced tissue damage. Previous studies investigating the protective effects of G. parasuis vaccination indicated a possible role of ApoA1 in reflecting disease progression following G. parasuis infection. However, the mechanisms of ApoA1 expression and its role in these infections are not well understood. In this investigation, newborn porcine tracheal (NPTr) epithelial cells infected with G. parasuis were used to elucidate the molecular mechanism and role of ApoA1. The study revealed that the AMPK pathway activation inhibited ApoA1 expression in NPTr cells infected with G. parasuis for the first time. Furthermore, Egr1 was identified as a core transcription factor regulating ApoA1 expression using a CRISPR/Cas9-based system. Importantly, it was discovered that APOA1 protein significantly reduced apoptosis, pyroptosis, necroptosis, and inflammatory factors induced by G. parasuis in vivo. These findings not only enhance our understanding of ApoA1 in response to bacterial infections but also highlight its potential in mitigating tissue damage caused by G. parasuis infection.


Subject(s)
AMP-Activated Protein Kinases , Apolipoprotein A-I , Early Growth Response Protein 1 , Haemophilus parasuis , Signal Transduction , Swine Diseases , Animals , Swine , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Haemophilus parasuis/genetics , Swine Diseases/microbiology , Swine Diseases/genetics , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Haemophilus Infections/veterinary , Haemophilus Infections/microbiology , Epithelial Cells/microbiology , Gene Expression Regulation , Trachea/microbiology , Trachea/metabolism , Apoptosis , Animals, Newborn
6.
Protein Sci ; 33(5): e4983, 2024 May.
Article in English | MEDLINE | ID: mdl-38659173

ABSTRACT

Serum amyloid A (SAA) is a highly conserved acute-phase protein that plays roles in activating multiple pro-inflammatory pathways during the acute inflammatory response and is commonly used as a biomarker of inflammation. It has been linked to beneficial roles in tissue repair through improved clearance of lipids and cholesterol from sites of damage. In patients with chronic inflammatory diseases, elevated levels of SAA may contribute to increased severity of the underlying condition. The majority of circulating SAA is bound to lipoproteins, primarily high-density lipoprotein (HDL). Interaction with HDL not only stabilizes SAA but also alters its functional properties, likely through altered accessibility of protein-protein interaction sites on SAA. While high-resolution structures for lipid-free, or apo-, forms of SAA have been reported, their relationship with the HDL-bound form of the protein, and with other possible mechanisms of SAA binding to lipids, has not been established. Here, we have used multiple biophysical techniques, including SAXS, TEM, SEC-MALS, native gel electrophoresis, glutaraldehyde crosslinking, and trypsin digestion to characterize the lipid-free and lipid-bound forms of SAA. The SAXS and TEM data show the presence of soluble octamers of SAA with structural similarity to the ring-like structures reported for lipid-free ApoA-I. These SAA octamers represent a previously uncharacterized structure for lipid-free SAA and are capable of scaffolding lipid nanodiscs with similar morphology to those formed by ApoA-I. The SAA-lipid nanodiscs contain four SAA molecules and have similar exterior dimensions as the lipid-free SAA octamer, suggesting that relatively few conformational rearrangements may be required to allow SAA interactions with lipid-containing particles such as HDL. This study suggests a new model for SAA-lipid interactions and provides new insight into how SAA might stabilize protein-lipid nanodiscs or even replace ApoA-I as a scaffold for HDL particles during inflammation.


Subject(s)
Serum Amyloid A Protein , Serum Amyloid A Protein/chemistry , Serum Amyloid A Protein/metabolism , Humans , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/metabolism , Nanostructures/chemistry , Models, Molecular , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/metabolism , Protein Binding
7.
J Lipid Res ; 65(5): 100543, 2024 May.
Article in English | MEDLINE | ID: mdl-38641010

ABSTRACT

AapoA-I, the main protein of high-density lipoprotein, plays a key role in the biogenesis and atheroprotective properties of high-density lipoprotein. We showed previously that a naturally occurring apoA-I mutation, L178P, induces major defects in protein's structural integrity and functions that may underlie the increased cardiovascular risk observed in carriers of the mutation. Here, a library of marketed drugs (956 compounds) was screened against apoA-I[L178P] to identify molecules that can stabilize the normal conformation of apoA-I. Screening was performed by the thermal shift assay in the presence of fluorescent dye SYPRO Orange. As an orthogonal assay, we monitored the change in fluorescence intensity of 8-anilinonaphthalene-1-sulfonic acid upon binding on hydrophobic sites on apoA-I. Screening identified four potential structure correctors. Subsequent analysis of the concentration-dependent effect of these compounds on secondary structure and thermodynamic stability of WT apoA-I and apoA-I[L178P] (assessed by thermal shift assay and circular dichroism spectroscopy), as well as on macrophage viability, narrowed the potential structure correctors to two, the drugs atorvastatin and bexarotene. Functional analysis showed that these two compounds can restore the defective capacity of apoA-I[L178P] to promote cholesterol removal from macrophages, an important step for atheroprotection. Computational docking suggested that both drugs target a positively charged cavity in apoA-I, formed between helix 1/2 and helix 5, and make extensive interactions that could underlie thermodynamic stabilization. Overall, our findings indicate that small molecules can correct defective apoA-I structure and function and may lead to novel therapeutic approaches for apoA-I-related dyslipidemias and increased cardiovascular risk.


Subject(s)
Apolipoprotein A-I , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/chemistry , Humans , Animals , Mice , Drug Evaluation, Preclinical , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemistry
8.
Biophys J ; 123(9): 1116-1128, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38555508

ABSTRACT

The physicochemical characteristics of the various subpopulations of high-density lipoproteins (HDLs) and, in particular, their surface properties determine their ability to scavenge lipids and interact with specific receptors and peptides. Five representative spheroidal HDL subpopulation models were mapped from a previously reported equilibrated coarse-grained (CG) description to an atomistic representation for subsequent molecular dynamics simulation. For each HDL model a range of finer-level analyses was undertaken, including the component-wise characterization of HDL surfaces, the average size and composition of hydrophobic surface patches, dynamic protein secondary structure monitoring, and the proclivity for solvent exposure of the proposed ß-amyloid (Aß) binding region of apolipoprotein A-I (apoA-I), "LN." This study reveals that previously characterized ellipsoidal HDL3a and HDL2a models revert to a more spherical geometry in an atomistic representation due to the enhanced conformational flexibility afforded to the apoA-I protein secondary structure, allowing for enhanced surface lipid packing and lower overall surface hydrophobicity. Indeed, the proportional surface hydrophobicity and apoA-I exposure reduced with increasing HDL size, consistent with previous characterizations. Furthermore, solvent exposure of the "LN" region of apoA-I was exclusively limited to the smallest HDL3c model within the timescale of the simulations, and typically corresponded to a distinct loss in secondary structure across the "LN" region to form part of a significant contiguous hydrophobic patch on the HDL surface. Taken together, these findings provide preliminary evidence for a subpopulation-specific interaction between HDL3c particles and circulating hydrophobic species such as Aß via the exposed "LN" region of apoA-I.


Subject(s)
Apolipoprotein A-I , Hydrophobic and Hydrophilic Interactions , Lipoproteins, HDL , Molecular Dynamics Simulation , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/metabolism , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/metabolism , Protein Structure, Secondary , Humans
9.
J Lipid Res ; 65(4): 100530, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38479648

ABSTRACT

Atherosclerosis results from the deposition and oxidation of LDL and immune cell infiltration in the sub-arterial space leading to arterial occlusion. Studies have shown that transcytosis transports circulating LDL across endothelial cells lining blood vessels. LDL transcytosis is initiated by binding to either scavenger receptor B1 (SR-B1) or activin A receptor-like kinase 1 on the apical side of endothelial cells leading to its transit and release on the basolateral side. HDL is thought to partly protect individuals from atherosclerosis due to its ability to remove excess cholesterol and act as an antioxidant. Apolipoprotein A1 (APOA1), an HDL constituent, can bind to SR-B1, raising the possibility that APOA1/HDL can compete with LDL for SR-B1 binding, thereby limiting LDL deposition in the sub-arterial space. To examine this possibility, we used in vitro approaches to quantify the internalization and transcytosis of fluorescent LDL in coronary endothelial cells. Using microscale thermophoresis and affinity capture, we find that SR-B1 and APOA1 interact and that binding is enhanced when using the cardioprotective variant of APOA1 termed Milano (APOA1-Milano). In male mice, transiently increasing the levels of HDL reduced the acute deposition of fluorescently labeled LDL in the atheroprone inner curvature of the aorta. Reduced LDL deposition was also observed when increasing circulating wild-type APOA1 or the APOA1-Milano variant, with a more robust inhibition from the APOA1-Milano. The results suggest that HDL may limit SR-B1-mediated LDL transcytosis and deposition, adding to the mechanisms by which it can act as an atheroprotective particle.


Subject(s)
Apolipoprotein A-I , Lipoproteins, HDL , Lipoproteins, LDL , Transcytosis , Animals , Humans , Male , Mice , Apolipoprotein A-I/metabolism , Atherosclerosis/metabolism , Endothelial Cells/metabolism , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Protein Binding , Scavenger Receptors, Class B/metabolism
10.
Prostaglandins Other Lipid Mediat ; 172: 106817, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38331090

ABSTRACT

Cardiovascular disease (CVD) has been the leading cause of death worldwide. As a chronic inflammatory disease, atherosclerosis (AS) acts as the initiating factor for CVD and reactive oxygen species (ROS) play a vital role in its development. Superoxide dismutases (SOD) can alleviate the detrimental effects of ROS and serve as the first line of defense through detoxifying the products derived from oxidative stress in vivo. Considering the potential preventive effects of high-density lipoprotein (HDL) on AS and the close relationship between CuZn superoxide dismutase (CuZnSOD) and HDL, the present work investigated whether CuZnSOD overexpression in swine could improve the function of HDL. Seven CuZnSOD transgenic swine, constructed by sperm and magnetic nanoparticles, demonstrated overexpressed CuZnSOD in the liver (P < 0.01) but comparable level to control in plasma (P > 0.05). CuZnSOD overexpression significantly down-regulated the levels of triglyceride (TG), apolipoprotein A-I (apoA-I) (P < 0.05), and high-density lipoprotein cholesterol (HDL-C) (P < 0.01) in plasma. In the presence of CuZnSOD overexpression, HDL3 significantly inhibited levels of IL-6 and TNF-α induced by oxidized low-density lipoprotein (oxLDL) (P < 0.05), indicating enhanced anti-inflammatory activity of HDL. At the same time, HDL-mediated cholesterol efflux did not decrease (P > 0.05). CuZnSOD overexpression improves the anti-inflammatory function of HDL despite decreased levels of HDL-C. In Conclusion, CuZnSOD overexpression improves HDL function in swine.


Subject(s)
Lipoproteins, HDL , Superoxide Dismutase , Animals , Swine , Superoxide Dismutase/metabolism , Superoxide Dismutase/genetics , Lipoproteins, HDL/metabolism , Animals, Genetically Modified , Interleukin-6/metabolism , Interleukin-6/genetics , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/genetics , Male , Liver/metabolism , Triglycerides/metabolism , Triglycerides/blood
11.
Arterioscler Thromb Vasc Biol ; 44(2): e20-e38, 2024 02.
Article in English | MEDLINE | ID: mdl-38095105

ABSTRACT

BACKGROUND: High cholesterol levels in pancreatic ß-cells cause oxidative stress and decrease insulin secretion. ß-cells can internalize apo (apolipoprotein) A-I, which increases insulin secretion. This study asks whether internalization of apoA-I improves ß-cell insulin secretion by reducing oxidative stress. METHODS: Ins-1E cells were cholesterol-loaded by incubation with cholesterol-methyl-ß-cyclodextrin. Insulin secretion in the presence of 2.8 or 25 mmol/L glucose was quantified by radioimmunoassay. Internalization of fluorescently labeled apoA-I by ß-cells was monitored by flow cytometry. The effects of apoA-I internalization on ß-cell gene expression were evaluated by RNA sequencing. ApoA-I-binding partners on the ß-cell surface were identified by mass spectrometry. Mitochondrial oxidative stress was quantified in ß-cells and isolated islets with MitoSOX and confocal microscopy. RESULTS: An F1-ATPase ß-subunit on the ß-cell surface was identified as the main apoA-I-binding partner. ß-cell internalization of apoA-I was time-, concentration-, temperature-, cholesterol-, and F1-ATPase ß-subunit-dependent. ß-cells with internalized apoA-I (apoA-I+ cells) had higher cholesterol and cell surface F1-ATPase ß-subunit levels than ß-cells without internalized apoA-I (apoA-I- cells). The internalized apoA-I colocalized with mitochondria and was associated with reduced oxidative stress and increased insulin secretion. The IF1 (ATPase inhibitory factor 1) attenuated apoA-I internalization and increased oxidative stress in Ins-1E ß-cells and isolated mouse islets. Differentially expressed genes in apoA-I+ and apoA-I- Ins-1E cells were related to protein synthesis, the unfolded protein response, insulin secretion, and mitochondrial function. CONCLUSIONS: These results establish that ß-cells are functionally heterogeneous, and apoA-I restores insulin secretion in ß-cells with elevated cholesterol levels by improving mitochondrial redox balance.


Subject(s)
Insulin-Secreting Cells , Insulin , Mice , Animals , Insulin/pharmacology , Apolipoprotein A-I/metabolism , Insulin-Secreting Cells/metabolism , Cholesterol/metabolism , Glucose/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/pharmacology
12.
J Lipid Res ; 65(1): 100482, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38052254

ABSTRACT

Excess cholesterol originating from nonhepatic tissues is transported within HDL particles to the liver for metabolism and excretion. Cholesterol efflux is initiated by lipid-free or lipid-poor apolipoprotein A1 interacting with the transmembrane protein ABCA1, a key player in cholesterol homeostasis. Defective ABCA1 results in reduced serum levels of HDL cholesterol, deposition of cholesterol in arteries, and an increased risk of early onset CVD. Over 300 genetic variants in ABCA1 have been reported, many of which are associated with reduced HDL cholesterol levels. Only a few of these have been functionally characterized. In this study, we have analyzed 51 previously unclassified missense variants affecting the extracellular domains of ABCA1 using a sensitive, easy, and low-cost fluorescence-based assay. Among these, only 12 variants showed a distinct loss-of-function phenotype, asserting their direct association with severe HDL disorders. These findings emphasize the crucial role of functional characterization of genetic variants in pathogenicity assessment and precision medicine. The functional rescue of ABCA1 loss-of-function variants through proteasomal inhibition or by the use of the chemical chaperone 4-phenylbutyric acid was genotype specific. Genotype-specific responses were also observed for the ability of apolipoprotein A1 to stabilize the different ABCA1 variants. In view of personalized medicine, this could potentially form the basis for novel therapeutic strategies.


Subject(s)
Apolipoprotein A-I , Cholesterol , Cholesterol, HDL , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Fluorescence , ATP Binding Cassette Transporter 1/genetics , Cholesterol/metabolism , Mutation, Missense
13.
Circulation ; 149(10): 774-787, 2024 03 05.
Article in English | MEDLINE | ID: mdl-38018436

ABSTRACT

BACKGROUND: Cholesterol efflux capacity (CEC) predicts cardiovascular disease independently of high-density lipoprotein (HDL) cholesterol levels. Isolated small HDL particles are potent promoters of macrophage CEC by the ABCA1 (ATP-binding cassette transporter A1) pathway, but the underlying mechanisms are unclear. METHODS: We used model system studies of reconstituted HDL and plasma from control and lecithin-cholesterol acyltransferase (LCAT)-deficient subjects to investigate the relationships among the sizes of HDL particles, the structure of APOA1 (apolipoprotein A1) in the different particles, and the CECs of plasma and isolated HDLs. RESULTS: We quantified macrophage and ABCA1 CEC of 4 distinct sizes of reconstituted HDL. CEC increased as particle size decreased. Tandem mass spectrometric analysis of chemically cross-linked peptides and molecular dynamics simulations of APOA1, the major protein of HDL, indicated that the mobility of C-terminus of that protein was markedly higher and flipped off the surface in the smallest particles. To explore the physiological relevance of the model system studies, we isolated HDL from LCAT-deficient subjects, whose small HDLs (like reconstituted HDLs) are discoidal and composed of APOA1, cholesterol, and phospholipid. Despite their very low plasma levels of HDL particles, these subjects had normal CEC. In both the LCAT-deficient subjects and control subjects, the CEC of isolated extra-small HDL (a mixture of extra-small and small HDL by calibrated ion mobility analysis) was 3- to 5-fold greater than that of the larger sizes of isolated HDL. Incubating LCAT-deficient plasma and control plasma with human LCAT converted extra-small and small HDL particles into larger particles, and it markedly inhibited CEC. CONCLUSIONS: We present a mechanism for the enhanced CEC of small HDLs. In smaller particles, the C-termini of the 2 antiparallel molecules of APOA1 are "flipped" off the lipid surface of HDL. This extended conformation allows them to engage with ABCA1. In contrast, the C-termini of larger HDLs are unable to interact productively with ABCA1 because they form a helical bundle that strongly adheres to the lipid on the particle. Enhanced CEC, as seen with the smaller particles, predicts decreased cardiovascular disease risk. Thus, extra-small and small HDLs may be key mediators and indicators of the cardioprotective effects of HDL.


Subject(s)
Apolipoprotein A-I , Cardiovascular Diseases , Humans , Apolipoprotein A-I/metabolism , Cardiovascular Diseases/metabolism , Lipoproteins, HDL/metabolism , Cholesterol , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Macrophages/metabolism , Cholesterol, HDL
14.
J Invest Dermatol ; 144(3): 645-658.e7, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37832842

ABSTRACT

Infantile hemangioma (IH) is the most frequent vascular tumor of infancy with unclear pathogenesis; disordered angiogenesis is considered to be involved in its formation. Apolipoprotein A-I binding protein (AIBP)-also known as NAXE (NAD [P]HX epimerase)-a regulator of cholesterol metabolism, plays a critical role in the pathological angiogenesis of mammals. In this study, we found that AIBP had much lower expression levels in both tissues from patients with IH and hemangioma endothelial cells (HemECs) than in adjacent normal tissues and human dermal vascular endothelial cells, respectively. Knockout of NAXE by CRISPR-Cas9 in HemECs enhanced tube formation and migration, and NAXE overexpression impaired tube formation and migration of HemECs. Interestingly, AIBP suppressed the proliferation of HemECs in hypoxia. We then found that reduced expression of AIBP correlated with increased hypoxia-inducible factor 1α levels in tissues from patients with IH and HemECs. Further mechanistic investigation demonstrated that AIBP disrupted hypoxia-inducible factor 1α signaling through cholesterol metabolism under hypoxia. Notably, AIBP significantly inhibited the development of IH in immunodeficient mice. Furthermore, using the validated mouse endothelial cell (ie, EOMA cells) and Naxe-/- mouse models, we demonstrated that both endogenous AIBP from tumors and AIBP in the tumor microenvironment limit the formation of hemangioma. These findings suggested that AIBP was a player in the pathogenesis of IH and could be a potential pharmacological target for treating IH.


Subject(s)
Endothelial Cells , Hemangioma , Humans , Animals , Mice , Endothelial Cells/metabolism , Apolipoprotein A-I/metabolism , Mice, Knockout , Hemangioma/genetics , Cholesterol/metabolism , Racemases and Epimerases/metabolism , Hypoxia/metabolism , Mammals , Tumor Microenvironment
15.
Cell Mol Gastroenterol Hepatol ; 17(4): 539-551, 2024.
Article in English | MEDLINE | ID: mdl-38122985

ABSTRACT

BACKGROUND & AIMS: Apolipoprotein A-1 (ApoA-1), the main apolipoprotein of high-density lipoprotein, has been well studied in the area of lipid metabolism and cardiovascular diseases. In this project, we clarify the function and mechanism of ApoA-1 in liver regeneration. METHODS: Seventy percent of partial hepatectomy was applied in male ApoA-1 knockout mice and wild-type mice to investigate the effects of ApoA-1 on liver regeneration. D-4F (ApoA-1 mimetic peptide), autophagy activator, and AMPK activator were used to explore the mechanism of ApoA-1 on liver regeneration. RESULTS: We demonstrated that ApoA-1 levels were highly expressed during the early stage of liver regeneration. ApoA-1 deficiency greatly impaired liver regeneration after hepatectomy. Meanwhile, we found that ApoA-1 deficiency inhibited autophagy during liver regeneration. The activation of autophagy protected against ApoA-1 deficiency in inhibiting liver regeneration. Furthermore, ApoA-1 deficiency impaired autophagy through AMPK-ULK1 pathway, and AMPK activation significantly improved liver regeneration. The administration of D-4F could accelerated liver regeneration after hepatectomy. CONCLUSIONS: These findings suggested that ApoA-1 played an essential role in liver regeneration through promoting autophagy in hepatocytes via AMPK-ULK1 pathway. Our findings enrich the understanding of the underlying mechanism of liver regeneration and provide a potential therapeutic strategy for liver injury.


Subject(s)
AMP-Activated Protein Kinases , Apolipoprotein A-I , Animals , Male , Mice , AMP-Activated Protein Kinases/metabolism , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/pharmacology , Autophagy , Liver/metabolism , Liver Regeneration
16.
Antivir Ther ; 28(6): 13596535231219639, 2023 12.
Article in English | MEDLINE | ID: mdl-38037795

ABSTRACT

BACKGROUND: Previously, we have demonstrated that Apolipoprotein A-I (ApoA-I) could inhibit the secretion of Hepatitis B virus (HBV), suggesting that stimulation of ApoA-I may block particle production. In the present study, we evaluated the anti-HBV effect of RVX-208, a small-molecule stimulator of ApoA-I gene expression. METHODS: RVX-208 was used to treat HepG2.2.15 cell, a HepG2 derived cell line stably producing HBV virus. Real-time PCR was performed to examine the HBV DNA levels. Magnetic particles, which were coated with anti-HBS or anti-HBE antibody, were used to examine the HBsAg and HBeAg levels in the supernatant of cultured HepG2.2.15 cells in combination with the enzyme conjugates that were prepared with horseradish peroxidase labelled anti-HBS or anti-HBE antibody in a double antibody sandwich manner. RNA-seq, immunoblots and real-time PCR were used to analyze the functional mechanism of RVX-208. RESULTS: RVX-208 could elevate the ApoA-I protein levels in HepG2.2.15 cells. In the meantime, RVX-208 significantly repressed HBV DNA, HBsAg and HBeAg levels in the supernatants of HepG2.2.15 cells. RNA-seq data revealed that RVX-208 treatment not only affected the cholesterol metabolism, which is closely related to ApoA-I, but also regulated signalling pathways that are associated with antiviral immune response. Moreover, mechanistic studies demonstrated that RVX-208 could activate cGAS-STING pathway and upregulate the transcription of a series of interferons, pro-inflammatory cytokines and chemokines with antiviral potential that are at the downstream of cGAS-STING pathway. CONCLUSION: Our study demonstrated that RVX-208, an inducer of ApoA-I, could suppress HBV particle production through activation of cGAS-STING pathway.


Subject(s)
Apolipoprotein A-I , Hepatitis B virus , Humans , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Hepatitis B Surface Antigens , DNA, Viral , Hepatitis B e Antigens , Hep G2 Cells , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/pharmacology
17.
Int J Mol Sci ; 24(22)2023 Nov 15.
Article in English | MEDLINE | ID: mdl-38003549

ABSTRACT

Preeclampsia (PE) is one of the pregnancy complications, leading to major maternal and fetal morbidity and mortality; however, the underlying mechanisms of PE still remain unclear. We aimed to explore the role of apolipoprotein A1 (APOA1) in the pathophysiology of PE. The expression of APOA1 was elevated in both plasma and placental tissues, as detected by Western blotting, immunohistochemistry, and a qRT-PCR assay. Importantly, we detected the concentration of APOA1 using the ELISA assay in normal control women (n = 30) and women with preeclampsia (n = 29) from a prospective cohort study. The concentration of APOA1 was not significantly altered in plasma during early and mid-term gestation of the PE patients compared to the NP patients; however, it was elevated during late gestation. Additionally, the concentration of APOA1 was positively associated with systolic blood pressure during late gestation. The proliferation and invasion of trophoblast were all increased in HTR8/SVneo cells transfected with APOA1 siRNA and decreased in HTR8/SVneo cells treated with the recombinant human APOA1 protein (rhAPOA1). Additionally, we used public datasets to investigate the downstream genes of APOA1 and qRT-PCR for validation. Furthermore, we explored the transcriptional activity of peroxisome proliferator-activated receptor gamma (PPARγ) in APOA1 by using a luciferase assay, which showed that the APOA1 promoter was activated by PPARγ. Additionally, the inhibitory effect of rhAPOA1 on the ability of trophoblast invasion and proliferation can be rescued by the PPARγ inhibitor. Our findings suggest the crucial role of APOA1 in PE, which might provide a new strategy for the prevention and treatment of PE.


Subject(s)
Placenta , Pre-Eclampsia , Pregnancy , Humans , Female , Placenta/metabolism , Pre-Eclampsia/metabolism , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , PPAR gamma/metabolism , Prospective Studies , Trophoblasts/metabolism , Cell Movement , Cell Proliferation/genetics
18.
Environ Sci Pollut Res Int ; 30(59): 123148-123163, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37979116

ABSTRACT

Although previous studies have indicated polycyclic aromatic hydrocarbons (PAHs) as cardiovascular health risk factors, evidence linking exposure to PAHs and blood lipids is still lacking, and the mechanism remains largely unknown. In this study, we evaluated the association between human internal exposure to PAHs and blood lipid levels in adults, as well as the indirect effects of inflammation and oxidative stress. The internal exposure of PAHs was assessed by determining serum PAHs and their hydroxylated metabolites (OH-PAHs) in the paired urine samples. Multivariable linear regression results demonstrated significant positive associations of individual PAHs and OH-PAHs with blood lipid biomarkers. The Bayesian kernel machine regression model revealed positive joint effects of PAH internal exposure on the fasting blood glucose, low-density lipoprotein cholesterol, total cholesterol, and total triglyceride, as well as an increased ratio of apolipoprotein B to apolipoprotein A1. In evaluating individual effects, serum phenanthrene played the most significant role in the association of increased PAH exposure with elevated fasting blood glucose. Quantile g-computation demonstrated the significant change in the levels of apolipoprotein B, ratio of apolipoprotein B to apolipoprotein A1, low-density lipoprotein cholesterol, and total cholesterol per quartile increase in PAH internal exposure. The restricted cubic spline analysis demonstrated the non-linear relationship between individual PAHs and OH-PAHs on blood lipid biomarkers. The mediation analysis indicated that PAH exposure may affect blood lipids not directly, but rather indirectly through intermediate inflammation and oxidative stress. The results demonstrated a significant association between increased PAH exposure levels and elevated blood lipids, highlighting the indirect effects of inflammation and oxidative stress.


Subject(s)
Polycyclic Aromatic Hydrocarbons , Adult , Humans , Polycyclic Aromatic Hydrocarbons/analysis , Apolipoprotein A-I/metabolism , Bayes Theorem , Blood Glucose , Inflammation , Oxidative Stress , Lipids , Biomarkers/metabolism , Lipoproteins, LDL , Cholesterol
19.
Sci Rep ; 13(1): 18514, 2023 10 28.
Article in English | MEDLINE | ID: mdl-37898709

ABSTRACT

The N-terminal fragment of apolipoprotein A-I (apoA-I), comprising residues 1-83, contains three segments prone to aggregation: residues 14-22, 53-58, and 67-72. We previously demonstrated that residues 14-22 are critical in apoA-I fibril formation while residues 53-58 entropically drove the nucleation process. Here, we investigated the impact of amyloidogenic mutations (Δ60-71/VT, Δ70-72, and F71Y) located around residues 67-72 on fibril formation by the apoA-I 1-83 fragment. Thioflavin T fluorescence assay demonstrated that the Δ60-71/VT mutation significantly enhances both nucleation and fibril elongation rates, whereas the Δ70-72 and F71Y mutations had minimal effects. Circular dichroism measurements and microscopic observations revealed that all variant fragments formed straight fibrils, transitioning from random coils to ß-sheet structures. Kinetic analysis demonstrated that primary nucleation is the dominant step in fibril formation, with fibril elongation reaching saturation at high protein concentrations. Thermodynamically, both nucleation and fibril elongation were enthalpically and entropically unfavorable in all apoA-I 1-83 variants, in which the entropic barrier of nucleation was almost eliminated for the Δ60-71/VT variant. Taken together, our results suggest the presence of new aggregation-prone segment in the Δ60-71/VT variant that promotes nucleation through entropic effects.


Subject(s)
Amyloid , Apolipoprotein A-I , Apolipoprotein A-I/metabolism , Kinetics , Mutagenesis, Insertional , Amyloid/metabolism , Mutation , Circular Dichroism
20.
Int J Mol Sci ; 24(14)2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37511116

ABSTRACT

A specific feature of dyslipidemia in pregnancy is increased high-density lipoprotein (HDL) cholesterol concentration, which is probably associated with maternal endothelium protection. However, preeclampsia is most often associated with low HDL cholesterol, and the mechanisms behind this change are scarcely explored. We aimed to investigate changes in HDL metabolism in risky pregnancies and those complicated by late-onset preeclampsia. We analyze cholesterol synthesis (cholesterol precursors: desmosterol, 7-dehydrocholesterol, and lathosterol) and absorption markers (phytosterols: campesterol and ß-sitosterol) within HDL particles (NCSHDL), the activities of principal modulators of HDL cholesterol's content, and major HDL functional proteins levels in mid and late pregnancy. On the basis of the pregnancy outcome, participants were classified into the risk group (RG) (70 women) and the preeclampsia group (PG) (20 women). HDL cholesterol was lower in PG in the second trimester compared to RG (p < 0.05) and followed by lower levels of cholesterol absorption markers (p < 0.001 for campesterolHDL and p < 0.05 for ß-sitosterolHDL). Lowering of HDL cholesterol between trimesters in RG (p < 0.05) was accompanied by a decrease in HDL phytosterol content (p < 0.001), apolipoprotein A-I (apoA-I) concentration (p < 0.05), and paraoxonase 1 (PON1) (p < 0.001), lecithin-cholesterol acyltransferase (LCAT) (p < 0.05), and cholesterol ester transfer protein (CETP) activities (p < 0.05). These longitudinal changes were absent in PG. Development of late-onset preeclampsia is preceded by the appearance of lower HDL cholesterol and NCSHDL in the second trimester. We propose that reduced capacity for intestinal HDL synthesis, decreased LCAT activity, and impaired capacity for HDL-mediated cholesterol efflux could be the contributing mechanisms resulting in lower HDL cholesterol.


Subject(s)
Pre-Eclampsia , Humans , Female , Pregnancy , Cholesterol, HDL/metabolism , Cholesterol/metabolism , Cholesterol Ester Transfer Proteins/metabolism , Biological Transport , Apolipoprotein A-I/metabolism , Aryldialkylphosphatase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...