Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 301
Filter
1.
Biochemistry (Mosc) ; 86(10): 1201-1213, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34903152

ABSTRACT

Apolipoprotein A-I (ApoA-I) is a key component of reverse cholesterol transport in humans. In the previous studies, we demonstrated expression of the apoA-I gene in human monocytes and macrophages; however, little is known on the regulation of the apoA-I expression in macrophages during the uptake of modified low-density lipoprotein (LDL), which is one of the key processes in the early stages of atherogenesis leading to formation of foam cells. Here, we demonstrate a complex nature of the apoA-I regulation in human macrophages during the uptake of oxidized LDL (oxLDL). Incubation of macrophages with oxLDL induced expression of the apoA-I gene within the first 24 hours, but suppressed it after 48 h. Both effects depended on the interaction of oxLDL with the TLR4 receptor, rather than on the oxLDL uptake by the macrophages. The oxLDL-mediated downregulation of the apoA-I gene depended on the ERK1/2 and JNK cascades, as well as on the NF-κB cascade.


Subject(s)
Apolipoprotein A-I/genetics , Gene Expression Regulation/drug effects , Lipoproteins, LDL/pharmacology , Macrophages/drug effects , Toll-Like Receptor 4/metabolism , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/metabolism , Humans , MAP Kinase Signaling System , Macrophages/metabolism , NF-kappa B/metabolism , THP-1 Cells
2.
Br J Haematol ; 193(3): 628-632, 2021 05.
Article in English | MEDLINE | ID: mdl-33216373

ABSTRACT

Sickle cell disease (SCD) and apolipoprotein L1 (APOL1) G1/G2 variants increase chronic kidney disease (CKD) risk in African Americans by poorly understood mechanisms. We applied bioinformatics to identify new candidate genes associated with SCD-related CKD. An interaction network demonstrated APOA1 connecting haemoglobin subunit ß (HBB) and APOL1 with 36 other candidate genes. Gene expression revealed upregulation of engulfment and cell motility 1 (ELMO1) and downregulation of APOA1 in the kidney cortex of SCD versus non-SCD mice. Analysis of candidate genes identified ELMO1 rs10951509 to be associated with albuminuria and APOA1 rs11216132 with haemoglobinuria in patients with SCD. A bioinformatic approach highlights ELMO1 and APOA1 as potentially associated with SCD nephropathy.


Subject(s)
Adaptor Proteins, Signal Transducing , Anemia, Sickle Cell , Apolipoprotein A-I , Cell Movement/genetics , Down-Regulation , Gene Regulatory Networks , Renal Insufficiency, Chronic , Up-Regulation , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Adult , Albuminuria/genetics , Albuminuria/metabolism , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Animals , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/genetics , Female , Humans , Male , Mice , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism
3.
Subcell Biochem ; 94: 399-420, 2020.
Article in English | MEDLINE | ID: mdl-32189309

ABSTRACT

High-density lipoprotein (HDL) and its main protein component apolipoprotein (apo)A-I, play an important role in cholesterol homeostasis. It has been demonstrated that HDLs comprise of a very heterogeneous group of particles, not only regarding size but also composition. HDL's best described function is its role in the reverse cholesterol transport, where lipid-free apoA-I or small HDLs can accept and take up cholesterol from peripheral cells and subsequently transport this to the liver for excretion. However, several other functions have also been described, like anti-oxidant, anti-inflammatory and anti-thrombotic effects. In this article, the general features, synthesis and metabolism of apoA-I and HDLs will be discussed. Additionally, an overview of HDL functions will be given, especially in the context of some major pathologies like cardiovascular disease, cancer and diabetes mellitus. Finally, the therapeutic potential of raising HDL will be discussed, focussing on the difficulties of the past and the promises of the future.


Subject(s)
Apolipoprotein A-I/metabolism , Lipoproteins, HDL/metabolism , Apolipoprotein A-I/biosynthesis , Cardiovascular Diseases/metabolism , Cholesterol/metabolism , Diabetes Mellitus/metabolism , Humans , Neoplasms/metabolism
4.
Clin Chim Acta ; 505: 55-59, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32092318

ABSTRACT

ApoA-I is a major protein component of high-density lipoprotein (HDL) that is widely known for regulating cholesterol trafficking and inflammatory and immune responses and for protecting against atherosclerosis. ApoA-I is generally considered to be synthesized in the liver (hepatocytes) and small intestine (enterocytes). However, computer analysis of ApoA-I has shown that the ApoA-I gene may be expressed in not only hepatocytes and enterocytes but also monocyte-macrophage cells, dendritic cells (DCs) and T cells. ApoA-I expression has been detected in THP-1 monocytes and macrophages, peripheral blood mononuclear cells (PBMCs) from postmenopausal women, human PBMC-derived monocytes and macrophages, mouse peritoneal macrophages, etc. Endogenous ApoA-I in macrophages has anti-inflammatory and cholesterol efflux effects. However, our understanding of the detailed roles of macrophage-synthesized ApoA-I is still at an early stage and very limited. More experiments are needed to elucidate the exact roles of endogenous ApoA-I in macrophages. Several lines of evidence indicate that recombinant exogenous human ApoA-I in mouse macrophages increases cholesterol efflux and thus reduces atherosclerosis development. Considering the antiatherogenic effect of exogenous ApoA-I overexpression in mouse macrophages, better understanding the role and mechanisms underlying macrophage-synthesized ApoA-I in atherosclerosis will enable macrophage-synthesized ApoA-I therapy to open new avenues for reducing the risk of atherosclerosis.


Subject(s)
Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/genetics , Atherosclerosis/prevention & control , Macrophages/metabolism , Animals , Cholesterol/metabolism , Humans
5.
Metabolism ; 105: 154186, 2020 04.
Article in English | MEDLINE | ID: mdl-32084429

ABSTRACT

BACKGROUND: Apolipoprotein A-I (ApoA-I) is involved in reverse cholesterol transport as a major component of HDL, but also conveys anti-thrombotic, anti-oxidative, anti-inflammatory and immune-regulatory properties that are pertinent to its protective roles in cardiovascular, inflammatory and malignant pathologies. Despite the pleiotropy in ApoA-I functions, the regulation of intracellular ApoA-I levels remains poorly explored. METHODS: HepG2 hepatoma cells and primary mouse hepatocytes were used as in vitro models to study the impact of genetic and chemical inhibitors of autophagy and the proteasome on ApoA-I by immunoblot, immunofluorescence and electron microscopy. Different growth conditions were implemented in conjunction with mTORC inhibitors to model the influence of nutrient scarcity versus sufficiency on ApoA-I regulation. Hepatic ApoA-I expression was also evaluated in high fat diet-fed mice displaying blockade in autophagy. RESULTS: Under nutrient-rich conditions, basal ApoA-I levels in liver cells are sustained by the balancing act of autophagy and of mTORC1-dependent de novo protein synthesis. ApoA-I proteolysis occurs through a canonical autophagic pathway involving Beclin1 and ULK1 and the receptor protein p62/SQSTM1 that targets ApoA-I to autophagosomes. However, upon aminoacid insufficiency, suppression of ApoA-I synthesis prevails, rendering mTORC1 inactivation dispensable for autophagy-mediated ApoA-I proteolysis. CONCLUSION: These data underscore the major contribution of post-transcriptional mechanisms to ApoA-I levels which differentially involve mTORC1-dependent signaling to protein synthesis and autophagy, depending on nutrient availability. Given the established role of ApoA-I in HDL-mediated reverse cholesterol transport, this mode of ApoA-I regulation may reflect a hepatocellular response to the organismal requirement for maintenance of cholesterol and lipid reserves under conditions of nutrient scarcity.


Subject(s)
Apolipoprotein A-I/biosynthesis , Autophagy/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Animals , Apolipoprotein A-I/genetics , Cholesterol/metabolism , Diet, High-Fat , Gene Expression Regulation , Hep G2 Cells , Hepatocytes/metabolism , Humans , Lipid Metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mice , Non-alcoholic Fatty Liver Disease/genetics , Nutritional Status , Proteasome Endopeptidase Complex/genetics
6.
Article in English | MEDLINE | ID: mdl-31461686

ABSTRACT

Apolipoprotein A-I (ApoA-I) plays an important role in lipid transport and performs an antimicrobial activity in vertebrates. However, the role of ApoA-I in invertebrates remains largely unexplored. In this study, an ApoA-I gene named BbApoA-I having an open reading frame of 1466 bp and encoding a polypeptide of 345 amino acids was cloned from a cephalochordate (Branchiostoma belcheri). The predicted polypeptide revealed conserved features, including α-helical secondary structures and coiled-coil regions in known vertebrate ApoA-I sequences. Quantitative real-time PCR analysis indicated that BbApoA-I was detected in all of the tested tissues, and the highest expression was found in the hepatic cecum. The BbApoA-I expression was upregulated after the specimens were injected with lipopolysaccharide. Recombinant BbApoA-I produced in an E. coli expression system was able to bind to a wide range of Gram-positive and Gram-negative bacteria as well as lipopolysaccharide and lipoteichoic acid. In addition, rBbApoA-I exhibited in vitro antibacterial activity against Gram-negative bacteria. These results indicated that BbApoA-I performed a protective function against bacterial infection in amphioxus.


Subject(s)
Anti-Bacterial Agents , Apolipoprotein A-I , Lancelets , Animals , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/chemistry , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/genetics , Bacteria/chemistry , Bacteria/metabolism , Gene Expression Regulation , Lancelets/genetics , Lancelets/metabolism , Protein Structure, Secondary
7.
Protein Sci ; 27(12): 2101-2109, 2018 12.
Article in English | MEDLINE | ID: mdl-30291643

ABSTRACT

Apolipoprotein A-I (ApoA-I)-related amyloidosis is a rare disease caused by missense mutations in the APOA1 gene. These mutations lead to protein aggregation and abnormal accumulation of ApoA-I amyloid fibrils in heart, liver, kidneys, skin, nerves, ovaries, or testes. Consequently, the carriers are at risk of single- or multi-organ failure and of need of organ transplantation. Understanding the basic molecular structure and function of ApoA-I amyloidogenic variants, as well as their biological effects, is, therefore, of great interest. However, the intrinsic low stability of this type of proteins makes their overexpression and purification difficult. To overcome this barrier, we here describe an optimized production and purification procedure for human ApoA-I amyloidogenic proteins that efficiently provides between 46 mg and 91 mg (depending on the protein variant) of pure protein per liter of Escherichia coli culture. Structural integrity of the amyloidogenic and native ApoA-I proteins were verified by circular dichroism spectroscopy and intrinsic fluorescence analysis, and preserved functionality was demonstrated by use of a lipid clearance assay as well as by reconstitution of high-density lipoprotein (HDL) particles. In conclusion, the use of the described high-yield protein production system to obtain amyloidogenic ApoA-I proteins, and their native counterpart, will enable molecular and cellular experimental studies aimed to explain the molecular basis for this rare disease.


Subject(s)
Apolipoprotein A-I/biosynthesis , Escherichia coli/metabolism , Genetic Variation , Recombinant Proteins/biosynthesis , Apolipoprotein A-I/genetics , Apolipoprotein A-I/isolation & purification , Escherichia coli/genetics , Genetic Variation/genetics , Humans , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
8.
Mol Cell Biochem ; 448(1-2): 211-223, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29442267

ABSTRACT

Apolipoprotein A-I (ApoA-I) is the main structural and functional protein component of high-density lipoprotein. ApoA-I has been shown to regulate lipid metabolism and inflammation in macrophages. Recently, we found the moderate expression of endogenous apoA-I in human monocytes and macrophages and showed that pro-inflammatory cytokine tumor necrosis factor α (TNFα) increases apoA-I mRNA and stimulates ApoA-I protein secretion by human monocytes and macrophages. Here, we present data about molecular mechanisms responsible for the TNFα-mediated activation of apoA-I gene in human monocytes and macrophages. This activation depends on JNK and MEK1/2 signaling pathways in human monocytes, whereas inhibition of NFκB, JNK, or p38 blocks an increase of apoA-I gene expression in the macrophages treated with TNFα. Nuclear receptor PPARα is a ligand-dependent regulator of apoA-I gene, whereas LXRs stimulate apoA-I mRNA transcription and ApoA-I protein synthesis and secretion by macrophages. Treatment of human macrophages with PPARα or LXR synthetic ligands as well as knock-down of LXRα, and LXRß by siRNAs interfered with the TNFα-mediated activation of apoA-I gene in human monocytes and macrophages. At the same time, TNFα differently regulated the levels of PPARα, LXRα, and LXRß binding to the apoA-I gene promoter in THP-1 cells. Obtained results suggest a novel tissue-specific mechanism of the TNFα-mediated regulation of apoA-I gene in monocytes and macrophages and show that endogenous ApoA-I might be positively regulated in macrophage during inflammation.


Subject(s)
Apolipoprotein A-I/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Liver X Receptors/metabolism , Macrophages/metabolism , Monocytes/metabolism , NF-kappa B/metabolism , PPAR alpha/metabolism , Tumor Necrosis Factor-alpha/metabolism , Humans , Macrophages/cytology , Monocytes/cytology , THP-1 Cells
9.
Arterioscler Thromb Vasc Biol ; 38(1): 206-217, 2018 01.
Article in English | MEDLINE | ID: mdl-29122817

ABSTRACT

OBJECTIVE: Gene therapy that expresses apo A-I (apolipoprotein A-I) from vascular wall cells has promise for preventing and reversing atherosclerosis. Previously, we reported that transduction of carotid artery endothelial cells with a helper-dependent adenoviral (HDAd) vector expressing apo A-I reduced early (4 weeks) fatty streak development in fat-fed rabbits. Here, we tested whether the same HDAd could provide long-term protection against development of more complex lesions. APPROACH AND RESULTS: Fat-fed rabbits (n=25) underwent bilateral carotid artery gene transfer, with their left and right common carotids randomized to receive either a control vector (HDAdNull) or an apo A-I-expressing vector (HDAdApoAI). Twenty-four additional weeks of high-fat diet yielded complex intimal lesions containing lipid-rich macrophages as well as smooth muscle cells, often in a lesion cap. Twenty-four weeks after gene transfer, high levels of apo A-I mRNA (median ≥250-fold above background) were present in all HDAdApoAI-treated arteries. Compared with paired control HDAdNull-treated arteries in the same rabbit, HDAdApoAI-treated arteries had 30% less median intimal lesion volume (P=0.03), with concomitant reductions (23%-32%) in intimal lipid, macrophage, and smooth muscle cell content (P≤0.05 for all). HDAdApoAI-treated arteries also had decreased intimal inflammatory markers. VCAM-1 (vascular cell adhesion molecule-1)-stained area was reduced by 36% (P=0.03), with trends toward lower expression of ICAM-1 (intercellular adhesion molecule-1), MCP-1 (monocyte chemoattractant protein 1), and TNF-α (tumor necrosis factor-α; 13%-39% less; P=0.06-0.1). CONCLUSIONS: In rabbits with severe hyperlipidemia, transduction of vascular endothelial cells with an apo A-I-expressing HDAd yields at least 24 weeks of local apo A-I expression that durably reduces atherosclerotic lesion growth and intimal inflammation.


Subject(s)
Apolipoprotein A-I/genetics , Atherosclerosis/prevention & control , Carotid Arteries/metabolism , Carotid Artery Diseases/prevention & control , Endothelial Cells/metabolism , Genetic Therapy/methods , Hyperlipidemias/therapy , Animals , Apolipoprotein A-I/biosynthesis , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Biomarkers/blood , Carotid Arteries/pathology , Carotid Artery Diseases/blood , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Disease Models, Animal , Endothelial Cells/pathology , Hyperlipidemias/blood , Hyperlipidemias/genetics , Intercellular Adhesion Molecule-1/metabolism , Lipids/blood , Male , Neointima , Plaque, Atherosclerotic , Rabbits , Transduction, Genetic , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
10.
FASEB J ; 32(3): 1602-1612, 2018 03.
Article in English | MEDLINE | ID: mdl-29183962

ABSTRACT

The intestine is involved in whole-body lipid and cholesterol homeostasis and secretes lipoproteins containing apolipoprotein (Apo)B48 and discrete ApoA-I into the mesenteric lymph. The lymphatic system has been proposed to have a significant role in the reverse cholesterol transport pathway associated with HDL-ApoA-I. In conditions of insulin resistance (IR), there is intestinal overproduction of chylomicrons containing ApoB48; however, there is limited data on the intestinal synthesis and secretion of HDL-ApoA-I. microRNA (miR)-223 has been shown to regulate peripheral HDL metabolism and may impact intestinal-derived HDL. Niacin (nicotinic acid; vitamin B3) is known to regulate lipid metabolism, but the role of niacin in modulating intestinal lipid and lipoprotein (ApoB48 and ApoA-I) metabolism is unknown. The aim of this study was to determine the secretion of intestinal lymphatic HDL-ApoA-I and the effect of dietary intervention with niacin on these pathways in a rodent model of IR. HDL was isolated from intestinal mesenteric lymph by density ultracentrifugation, and subsequent HDL miR analysis was developed in collaboration with Exiqon Services. Insulin-resistant rodents were fed chow or chow with niacin (1% w/w) for 6 wk. Intestinal lymph HDL-ApoA-I and miR-223 expression were lower by at least 45 and 60%, respectively, and lymph HDL was associated with 85% higher triglyceride (TG) content in IR compared to non-IR control group. Niacin was found to increase secretion of lymph HDL and miR-223 by at least 50-60% and to deplete the TGs associated with HDL compared with the nontreated IR group. Niacin significantly increased peroxisome proliferator-activating nuclear receptor α and carnitine palmitoyltransferase I α mRNA and annulled Tnf-α mRNA expression in intestinal (jejunal) explants. Altered intestinal lymphatic HDL-ApoA-I and miR-223 metabolism in IR and modulation by niacin may provide insight into the intestinal-mediated regulation of the reverse cholesterol transport pathway.-Mangat, R., Borthwick, F., Haase, T., Jacome, M., Nelson, R., Kontush, A., Vine, D. F., Proctor, S. D. Intestinal lymphatic HDL miR-223 and ApoA-I are reduced during insulin resistance and restored with niacin.


Subject(s)
Apolipoprotein A-I/biosynthesis , Gene Expression Regulation/drug effects , Insulin Resistance/ethnology , Intestinal Mucosa/metabolism , Lipoproteins, HDL/biosynthesis , Lymph Nodes/metabolism , MicroRNAs/biosynthesis , Niacin/pharmacology , Animals , Apolipoprotein A-I/genetics , Lipoproteins, HDL/genetics , Male , Mesentery/metabolism , Mice , Mice, Transgenic , MicroRNAs/genetics
11.
Assay Drug Dev Technol ; 15(8): 362-371, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28742374

ABSTRACT

Apolipoprotein A-I (apo A-I) is the primary antiatherogenic protein in high-density lipoprotein (HDL). Despite the controversy as to the clinical effectiveness of raising HDL, the search is ongoing for safe and effective drugs that increase HDL and apo A-I levels. To identify novel compounds that can increase hepatic apo A-I production, two drug libraries were screened. The NIH clinical collection (NCC) and the NIH clinical collection 2 (NCC2) were purchased from Evotec (San Francisco, CA). The NCC library contains 446 compounds and the NCC2 library contains 281 compounds, all dissolved in dimethylsulfoxide at a concentration of 10 mM. Hepatoma-derived cells (HepG2) and primary hepatocytes in culture were treated with various compounds for 24 h and apo A-I in media samples was measured by enzyme immunoassay. Samples with significant changes in apo A-I concentrations were retested in independent experiments by Western blot analysis to confirm the immunoassay findings. Of a total of 727 compounds screened at a concentration of 50 µM, 15 compounds increased hepatic apo A-I production by 35%-54%, and 9 compounds lowered hepatic apo A-I concentrations in the culture media by 25%-52%. Future trials should explore the clinical effectiveness of these agents when standard doses of these drugs are used in humans.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apolipoprotein A-I/biosynthesis , High-Throughput Screening Assays , Small Molecule Libraries/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
12.
Protein Expr Purif ; 137: 13-19, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28624493

ABSTRACT

Human apolipoprotein A-I (apoA-I) is the most abundant protein in high-density lipoprotein, an anti-atherogenic lipid-protein complex responsible for reverse cholesterol transport. The protein is composed of an N-terminal helix bundle domain, and a small C-terminal (CT) domain. To facilitate study of CT-apoA-I, a novel strategy was employed to produce this small domain in a bacterial expression system. A protein construct was designed of insect apolipophorin III (apoLp-III) and residues 179-243 of apoA-I, with a unique methionine residue positioned between the two proteins and an N-terminal His-tag to facilitate purification. The chimera was expressed in E. coli, purified by Ni-affinity chromatography, and cleaved by cyanogen bromide. SDS-PAGE revealed the presence of three proteins with masses of 7 kDa (CT-apoA-I), 18 kDa (apoLp-III), and a minor 26 kDa band of uncleaved chimera. The digest was reloaded on the Ni-affinity column to bind apoLp-III and uncleaved chimera, while CT-apoA-I was washed from the column and collected. Alternatively, CT-apoA-I was isolated from the digest by reversed-phase HPLC. CT-apoA-I was α-helical, highly effective in solubilizing phospholipid vesicles and disaggregating LPS micelles. However, CT-apoA-I was less active compared to full-length apoA-I in protecting lipolyzed low density lipoproteins from aggregating, and disrupting phosphatidylglycerol bilayer vesicles. Thus the novel expression system produced mg quantities of functional CT-apoA-I, facilitating structural and functional studies of this critical domain of apoA-I.


Subject(s)
Apolipoprotein A-I , Escherichia coli/metabolism , Gene Expression , Recombinant Fusion Proteins , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/genetics , Apolipoprotein A-I/isolation & purification , Escherichia coli/genetics , Humans , Protein Domains , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification
13.
Cardiovasc Ther ; 35(4)2017 Aug.
Article in English | MEDLINE | ID: mdl-28423226

ABSTRACT

Low-density cholesterol (LDL) has been the prime target of currently available lipid-lowering therapies although current research is expanding the focus beyond LDL lowering and has included high-density cholesterol (HDL) also as the target. Bromo and extra-terminal (BET) proteins are implicated in the regulation of transcription of several regulatory genes and regulation of proinflammatory pathways. As atherosclerosis is an inflammatory pathway and studies showed that BET inhibition has a role in inhibiting inflammation, the concept of BET inhibition came in the field of atherosclerosis. RVX 208 is a novel, orally active, BET protein inhibitor and the only BET inhibitor currently available in the field of atherosclerosis. RVX 208 acts primarily by increasing apo A-I (apolipoprotein A-I) and HDL levels. RVX 208 has a novel action of increasing larger, more cardio-protective HDL particles. Post hoc analysis of Phase II trials also showed that RVX 208 reduced major adverse cardiovascular events (MACE) in treated patients, over and above that of apo A-I/HDL increasing action. This MACE reducing actions of RVX 208 were largely due to its novel anti-inflammatory actions. Currently, a phase III trial, BETonMACE, is recruiting patients to look for the effects of RVX 208 in patients with increased risk of atherosclerotic cardiovascular disease. So BET inhibitors act in multiple ways to inhibit and modulate atherosclerosis and would be an emerging and potential option in the management of multifactorial disease like coronary artery disease by inhibiting a single substrate. But we need long-term phase III trial data's to look for effects on real-world patients.


Subject(s)
Apolipoprotein A-I/biosynthesis , Lipoproteins, HDL/biosynthesis , Protein Synthesis Inhibitors/pharmacology , Proteins/antagonists & inhibitors , Quinazolines/pharmacology , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Humans , Quinazolinones
14.
Protein Expr Purif ; 134: 18-24, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28336201

ABSTRACT

Apolipoprotein (apo) A-I is the major protein component of high-density lipoprotein (HDL) and plays key roles in the Reverse Cholesterol Transport pathway. In the past decade, reconstituted HDL (rHDL) has been employed as a therapeutic agent for treatment of atherosclerosis. The ability of rHDL to promote cholesterol efflux from peripheral cells has been documented to reduce the size of atherosclerotic plaque lesions. However, development of apoA-I rHDL-based therapeutics for human use requires a cost effective process to generate an apoA-I product that meets "Good Manufacturing Practice" standards. Methods available for production and isolation of unmodified recombinant human apoA-I at scale are cumbersome, laborious and complex. To overcome this obstacle, a streamlined two-step procedure has been devised for isolation of recombinant untagged human apoA-I from E. coli that takes advantage of its ability to re-fold to a native conformation following denaturation. Heat treatment of a sonicated E. coli supernatant fraction induced precipitation of a large proportion of host cell proteins (HCP), yielding apoA-I as the major soluble protein. Reversed-phase HPLC of this material permitted recovery of apoA-I largely free of HCP and endotoxin. Purified apoA-I possessed α-helix secondary structure, formed rHDL upon incubation with phospholipid and efficiently promoted cholesterol efflux from cholesterol loaded J774 macrophages.


Subject(s)
Apolipoprotein A-I , Escherichia coli/metabolism , Protein Refolding , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/isolation & purification , Apolipoprotein A-I/pharmacokinetics , Biological Transport, Active/drug effects , Cell Line , Cholesterol/metabolism , Escherichia coli/chemistry , Escherichia coli/genetics , Humans , Macrophages/metabolism , Protein Structure, Secondary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
15.
J Cell Biochem ; 118(2): 382-396, 2017 02.
Article in English | MEDLINE | ID: mdl-27404023

ABSTRACT

Apolipoprotein A-I (ApoA-I) is a key component of high density lipoproteins which possess anti-atherosclerotic and anti-inflammatory properties. Insulin is a crucial mediator of the glucose and lipid metabolism that has been implicated in atherosclerotic and inflammatory processes. Important mediators of insulin signaling such as Liver X Receptors (LXRs) and Forkhead Box A2 (FOXA2) are known to regulate apoA-I expression in liver. Forkhead Box O1 (FOXO1) is a well-known target of insulin signaling and a key mediator of oxidative stress response. Low doses of insulin were shown to activate apoA-I expression in human hepatoma HepG2 cells. However, the detailed mechanisms for these processes are still unknown. We studied the possible involvement of FOXO1, FOXA2, LXRα, and LXRß transcription factors in the insulin-mediated regulation of apoA-I expression. Treatment of HepG2 cells with high doses of insulin (48 h, 100 nM) suppresses apoA-I gene expression. siRNAs against FOXO1, FOXA2, LXRß, or LXRα abrogated this effect. FOXO1 forms a complex with LXRß and insulin treatment impairs FOXO1/LXRß complex binding to hepatic enhancer and triggers its nuclear export. Insulin as well as LXR ligand TO901317 enhance the interaction between FOXA2, LXRα, and hepatic enhancer. These data suggest that high doses of insulin downregulate apoA-I gene expression in HepG2 cells through redistribution of FOXO1/LXRß complex, FOXA2, and LXRα on hepatic enhancer of apoA-I gene. J. Cell. Biochem. 118: 382-396, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Apolipoprotein A-I/biosynthesis , Carcinoma, Hepatocellular/metabolism , Down-Regulation/drug effects , Forkhead Box Protein O1/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Insulin/pharmacology , Liver Neoplasms/metabolism , Liver X Receptors/metabolism , Neoplasm Proteins/metabolism , Carcinoma, Hepatocellular/pathology , Hep G2 Cells , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver Neoplasms/pathology , Sulfonamides/pharmacology
16.
Mol Cell Biochem ; 424(1-2): 45-55, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27734225

ABSTRACT

ApoA1 is a player in reverse cholesterol transport that initiates multiple cellular pathways on binding to its receptor ABCA1. Its relation to neuronal injury is however unclear. We found ApoA1 to be increasingly abundant at a later time point in the secondary phase of traumatic spinal cord injury. In a cellular injury model of neuroblastoma, ApoA1 showed an initial diminished expression after infliction of injury, which sharply increased thereafter. Subsequently, ApoA1 was shown to alter wound healing dynamics in neuroblastoma injury model. It was observed that an initial lag in scratch wound closure was followed by rapid healing in the ApoA1 treatment group. Activation of ERK pathway and Actin polymerisation by ApoA1 corroborated its role in healing after neuronal injury. We propose that ApoA1 is increasingly expressed and secreted as a delayed response to neuronal injury, and this is a self-protecting mechanism of the injured system.


Subject(s)
Apolipoprotein A-I/biosynthesis , Gene Expression Regulation , MAP Kinase Signaling System , Regeneration , Spinal Cord Injuries/metabolism , Adult , Cell Line, Tumor , Female , Humans , Male , Middle Aged , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
17.
Arterioscler Thromb Vasc Biol ; 37(2): 316-327, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27932352

ABSTRACT

OBJECTIVE: Gene therapy, delivered directly to the blood vessel wall, could potentially prevent atherosclerotic lesion growth and promote atherosclerosis regression. Previously, we reported that a helper-dependent adenoviral (HDAd) vector expressing apolipoprotein A-I (apoA-I) in carotid endothelium of fat-fed rabbits reduced early (4 weeks) atherosclerotic lesion growth. Here, we tested whether the same HDAd-delivered to the existing carotid atherosclerotic lesions-could promote regression. APPROACH AND RESULTS: Rabbits (n=26) were fed a high-fat diet for 7 months, then treated with bilateral carotid gene transfer. One carotid was infused with an HDAd expressing apoA-I (HDAdApoAI) and the other with a control nonexpressing HDAd (HDAdNull). The side with HDAdApoAI was randomized. Rabbits were then switched to regular chow, lowering their plasma cholesterols by over 70%. ApoA-I mRNA and protein were detected in HDAdApoAI-transduced arteries. After 7 weeks of gene therapy, compared with HDAdNull-treated arteries in the same rabbits, HDAdApoAI-treated arteries had significantly less vascular cell adhesion molecule-1 expression (28%; P=0.04) along with modest but statistically insignificant trends toward decreased intimal lesion volume, lipid and macrophage content, and intercellular adhesion molecule-1 expression (9%-21%; P=0.1-0.4). Post hoc subgroup analysis of rabbits with small-to-moderate-sized lesions (n=20) showed that HDAdApoAI caused large reductions in lesion volume, lipid content, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1 expression (30%-50%; P≤0.04 for all). Macrophage content was reduced by 30% (P=0.06). There was a significant interaction (P=0.02) between lesion size and treatment efficacy. CONCLUSIONS: Even when administered on a background of aggressive lowering of plasma cholesterol, local HDAdApoAI vascular gene therapy may promote rapid regression of small-to-moderate-sized atherosclerotic lesions.


Subject(s)
Apolipoprotein A-I/biosynthesis , Atherosclerosis/therapy , Carotid Artery Diseases/therapy , Carotid Artery, Common/metabolism , Genetic Therapy/methods , Transduction, Genetic , Adenoviridae/genetics , Animals , Apolipoprotein A-I/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Carotid Artery, Common/pathology , Diet, Atherogenic , Disease Models, Animal , Genetic Vectors , Intercellular Adhesion Molecule-1/metabolism , Lipids/blood , Macrophages/metabolism , Male , Muscle, Smooth, Vascular , Neointima , Plaque, Atherosclerotic , Rabbits , Remission Induction , T-Lymphocytes/metabolism , Time Factors , Vascular Cell Adhesion Molecule-1/metabolism
18.
J Cell Biochem ; 118(4): 754-763, 2017 04.
Article in English | MEDLINE | ID: mdl-27618583

ABSTRACT

Increasing apolipoproteinA-I (apoA-I) production may be anti-atherogenic. Thus, there is a need to identify regulatory factors involved. Transcription of apoA-I involves peroxisome-proliferator-activated-receptor-alpha (PPARα) activation, but endoplasmic reticulum (ER) -stress and inflammation also influence apoA-I production. To unravel why PPARα agonist GW7647 increased apoA-I production compared to PPARα agonist fenofibric acid (FeAc) in human hepatocellular carcinoma (HepG2) and colorectal adenocarcinoma (CaCo-2) cells, gene expression profiles were compared. Microarray analyses suggested CCAAT/enhancer-binding-protein-beta (C/EBP-ß) involvement in the FeAc condition. Therefore, C/EBP-ß silencing and isoform-specific overexpression experiments were performed under ER-stressed, inflammatory and non-inflammatory conditions. mRNA expression of C/EBP-ß, ATF3, NF-IL3 and GDF15 were upregulated by FeAc compared to GW7647 in both cell lines, while DDIT3 and DDIT4 mRNA were only upregulated in HepG2 cells. This ER-stress related signature was associated with decreased apoA-I secretion. After ER-stress induction by thapsigargin or FeAc addition, intracellular apoA-I concentrations decreased, while ER-stress marker expression (CHOP, XBP1s, C/EBP-ß) increased. Cytokine addition increased intracellular C/EBP-ß levels and lowered apoA-I concentrations. Although a C/EBP binding place is present in the apoA-I promoter, C/EBP-ß silencing or isoform-specific overexpression did not affect apoA-I production in inflammatory, non-inflammatory and ER-stressed conditions. Therefore, C/EBP-ß is not a target to influence hepatic apoA-I production. J. Cell. Biochem. 118: 754-763, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Apolipoprotein A-I/biosynthesis , Butyrates/pharmacology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Fenofibrate/analogs & derivatives , PPAR alpha/agonists , Phenylurea Compounds/pharmacology , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , CCAAT-Enhancer-Binding Protein-beta/antagonists & inhibitors , CCAAT-Enhancer-Binding Protein-beta/genetics , Caco-2 Cells , Endoplasmic Reticulum Stress/drug effects , Fenofibrate/pharmacology , Gene Expression Profiling , Gene Silencing , Hep G2 Cells , Humans , Inflammation/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thapsigargin/pharmacology
19.
Gut ; 66(5): 896-907, 2017 05.
Article in English | MEDLINE | ID: mdl-27609828

ABSTRACT

OBJECTIVE: The HCV life cycle and the lipid metabolism are inextricably intertwined. In the blood, HCV virions are associated with lipoproteins, forming lipoviroparticles (LVPs), which are the most infectious form of the virus. Apolipoprotein E (apoE), a key LVP component, plays an essential role in HCV entry, assembly and egress. ApoE is also a cell host factor involved in lipoprotein homeostasis. Although the majority of apoE is associated with lipoproteins, a lipid-free (LF) form exists in blood. However, the role of LF-apoE in both lipid metabolism and HCV life cycle is poorly understood. DESIGN: In this study, using the cell culture-derived HCV model system in human hepatoma Huh7.5.1 cells and primary human hepatocytes (PHH), we investigated the effect of LF-apoE on the early steps of HCV life cycle and on the lipid metabolism of hepatic cells. RESULTS: A dose-dependent decrease in HCV replication was observed when Huh7.5.1 cells and PHH were treated with increasing amounts of LF-apoE. We showed that LF-apoE acts on HCV replication independently of previously described apoE receptors. We observed that LF-apoE induced a marked hepatic cholesterol efflux via the ATP-binding cassette subfamily G member 1 (ABCG1) protein that in turn inhibits HCV replication. LF-apoE also increases both apolipoprotein AI and high-density lipoprotein production. CONCLUSIONS: Our findings highlight a new mechanism in lipid metabolism regulation and interaction of the lipid metabolism with the HCV life cycle, which may be important for viral pathogenesis and might also be explored for antiviral therapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Apolipoproteins E/metabolism , Apolipoproteins E/pharmacology , Cholesterol/metabolism , Hepacivirus/physiology , Virus Replication/drug effects , Apolipoprotein A-I/biosynthesis , Cell Line, Tumor , Dose-Response Relationship, Drug , Hepacivirus/growth & development , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Life Cycle Stages/drug effects , Lipoproteins, HDL/biosynthesis , Membrane Microdomains , Virus Internalization
20.
Indian J Med Res ; 144(2): 238-244, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27934803

ABSTRACT

BACKGROUND & OBJECTIVES: Hepatic scavenger receptor class B1 (SR-B1), a high-density lipoprotein (HDL) receptor, is involved in the selective uptake of HDL-associated esterified cholesterol (EC), thereby regulates cholesterol homoeostasis and improves reverse cholesterol transport. Previously, we reported in euglycaemic obese rats (WNIN/Ob strain) that feeding of vitamin A-enriched diet normalized hypercholesterolaemia, possibly through hepatic SR-B1-mediated pathway. This study was aimed to test whether it would be possible to normalize hypercholesterolaemia in glucose-intolerant obese rat model (WNIN/GR/Ob) through similar mechanism by feeding identical vitamin A-enriched diet. METHODS: In this study, 30 wk old male lean and obese rats of WNIN/GR-Ob strain were divided into two groups and received either stock diet or vitamin A-enriched diet (2.6 mg or 129 mg vitamin A/kg diet) for 14 wk. Blood and other tissues were collected for various biochemical analyses. RESULTS: Chronic vitamin A-enriched diet feeding decreased hypercholesterolaemia and normalized abnormally elevated plasma HDL-cholesterol (HDL-C) levels in obese rats as compared to stock diet-fed obese groups. Further, decreased free cholesterol (FC) and increased esterified cholesterol (EC) contents of plasma cholesterol were observed, which were reflected in higher EC to FC ratio of vitamin A-enriched diet-fed obese rats. However, neither lecithin-cholesterol acyltransferase (LCAT) activity of plasma nor its expression (both gene and protein) in the liver were altered. On the contrary, hepatic cholesterol levels significantly increased in vitamin A-enriched diet fed obese rats. Hepatic SR-B1 expression (both mRNA and protein) remained unaltered among groups. Vitamin A-enriched diet fed obese rats showed a significant increase in hepatic low-density lipoprotein receptor mRNA levels, while the expression of genes involved in HDL synthesis, namely, ATP-binding cassette protein 1 (ABCA1) and apolipoprotein A-I, were downregulated. No such response was seen in vitamin A-supplemented lean rats as compared with their stock diet-fed lean counterparts. INTERPRETATION & CONCLUSIONS: Chronic vitamin A-enriched diet feeding decreased hypercholesterolaemia and normalized HDL-C levels, possibly by regulating pathways involved in HDL synthesis and degradation, independent of hepatic SR-B1 in this glucose-intolerant obese rat model.


Subject(s)
Cholesterol/blood , Hypercholesterolemia/blood , Obesity/blood , Scavenger Receptors, Class B/biosynthesis , Vitamin A/administration & dosage , ATP Binding Cassette Transporter 1/biosynthesis , Animals , Apolipoprotein A-I/biosynthesis , Biological Transport/genetics , Cholesterol/genetics , Cholesterol, HDL/biosynthesis , Cholesterol, HDL/blood , Diet , Disease Models, Animal , Gene Expression Regulation , Humans , Hypercholesterolemia/diet therapy , Hypercholesterolemia/genetics , Liver/metabolism , Male , Metabolic Networks and Pathways/genetics , Obesity/diet therapy , Obesity/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/blood , Rats , Scavenger Receptors, Class B/genetics , Vitamin A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...