Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
Add more filters

Publication year range
2.
J Cell Physiol ; 236(6): 4750-4763, 2021 06.
Article in English | MEDLINE | ID: mdl-33615471

ABSTRACT

Excessively high cholesterol content in the blood leads to nonalcohol fatty liver disease (NAFLD) and arteriosclerosis. Although there are increasing publications and patent applications to lower blood cholesterol with small chemical molecules, limited effective drugs can be available in clinic. It is necessary to uncover new targets and drugs to alleviate high cholesterol. Esterase D (ESD) is abundant in liver and it remains unknown about its role in cholesterol metabolism. Here we reported that small chemical molecule fluorescigenic pyrazoline derivative 5 (FPD5), a new ESD activator, could effectively reverse high blood cholesterol level and prevent fatty liver and arteriosclerosis in apoE-/- mice fed the high-fat diet. We also observed that FPD5 could reduce oxidized low density lipoprotein (oxLDL)-induced formation of foam cells. To further investigate the mechanism of FPD5 action on blood cholesterol modulation, we found that ESD trigged by FPD5 was aggregated in lysosome and interacted with Jun activation domain binding protein 1 (JAB1). ESD served as a deacetylase to remove Thr89 acetylation of JAB1 and increased its activity; thus, promoting the ATP-binding cassette transporters A1 (ABCA1) to accelerate cholesterol efflux. Our findings demonstrate that FPD5 decreases blood cholesterol level to ameliorate NAFLD and arteriosclerosis through ESD/JAB1/ABCA1 pathway, and ESD functions as a novel nonclassical deacetylase that hydrolyzes serine/threonine acetyl group. Our findings not only highlight that FPD5 may be a pioneer drug for alleviating blood cholesterol but also indicate that ESD is a potential drug target that promotes cholesterol metabolism.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Anticholesteremic Agents/pharmacology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , COP9 Signalosome Complex/metabolism , Cholesterol/blood , Enzyme Inhibitors/pharmacology , Foam Cells/drug effects , Peptide Hydrolases/metabolism , Thiolester Hydrolases/antagonists & inhibitors , Acetylation , Animals , Aortic Diseases/blood , Aortic Diseases/enzymology , Aortic Diseases/pathology , Atherosclerosis/blood , Atherosclerosis/enzymology , Atherosclerosis/pathology , Biomarkers/blood , Diet, High-Fat , Disease Models, Animal , Down-Regulation , Foam Cells/enzymology , Foam Cells/pathology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/prevention & control , Plaque, Atherosclerotic , Protein Processing, Post-Translational , RAW 264.7 Cells , Thiolester Hydrolases/metabolism
3.
Arterioscler Thromb Vasc Biol ; 40(5): 1155-1167, 2020 05.
Article in English | MEDLINE | ID: mdl-32212851

ABSTRACT

OBJECTIVES: During the advancement of atherosclerosis, plaque cellularity is governed by the influx of monocyte-derived macrophages and their turnover via apoptotic and nonapoptotic forms of cell death. Previous reports have demonstrated that programmed necrosis, or necroptosis, of plaque macrophages contribute to necrotic core formation. Knockdown or inhibition of the necrosome components RIPK1 (receptor-interacting protein kinase 1) and RIPK3 (receptor-interacting protein kinase 3) slow atherogenesis, and activation of the terminal step of necroptosis, MLKL (mixed lineage kinase domain-like protein), has been demonstrated in advanced human atherosclerotic plaques. However, whether MLKL directly contributes to lesion development and necrotic core formation has not been investigated. Approaches and Results: MLKL expression was knocked down in atherogenic Apoe-knockout mice via the administration of antisense oligonucleotides. During atherogenesis, Mlkl knockdown decreased both programmed cell death and the necrotic core in the plaque. However, total lesion area remained unchanged. Furthermore, treatment with the MLKL antisense oligonucleotide unexpectedly reduced circulating cholesterol levels compared with control antisense oligonucleotide but increased the accumulation of lipids within the plaque and in vitro in macrophage foam cells. MLKL colocalized with the late endosome and multivesicular bodies in peritoneal macrophages incubated with atherogenic lipoproteins. Transfection with MLKL antisense oligonucleotide increased lipid localization with the multivesicular bodies, suggesting that upon Mlkl knockdown, lipid trafficking becomes defective leading to enhanced lipid accumulation in macrophages. CONCLUSIONS: These studies confirm the requirement for MLKL as the executioner of necroptosis, and as such a significant contributor to the necrotic core during atherogenesis. We also identified a previously unknown role for MLKL in regulating endosomal trafficking to facilitate lipid handling in macrophages during atherogenesis.


Subject(s)
Aortic Diseases/enzymology , Atherosclerosis/enzymology , Cholesterol/metabolism , Foam Cells/enzymology , Macrophages, Peritoneal/enzymology , Plaque, Atherosclerotic , Protein Kinases/deficiency , Animals , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Disease Models, Animal , Endosomes/metabolism , Female , Foam Cells/pathology , Macrophages, Peritoneal/pathology , Male , Mice, Knockout, ApoE , Necroptosis , Necrosis , Oligonucleotides, Antisense/administration & dosage , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction
4.
J Cardiovasc Pharmacol ; 77(4): 508-518, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33136767

ABSTRACT

ABSTRACT: Foam cells are the main pathological components of atherosclerosis. Therapies reducing foam cell formation can effectively prevent atherosclerotic diseases and cardiovascular events. Beyond lowering plasma cholesterol levels, the pleiotropic functions of statins in atherosclerosis have not been fully elucidated. In the present study, atorvastatin reduced cholesterol content and increased cholesterol efflux from foam cells in a concentration-dependent manner. Atorvastatin (10 µM) inhibited foam cell formation within 48 hours. Furthermore, we found that atorvastatin inhibited foam cell formation by promoting lipophagy, which was manifested by increased autophagy-related gene 5 (Atg5) expression, elevated ratio of microtubule-associated protein1 light chain 3 (LC3) II to LC3I, reduced p62 expression, and increased LC3 and lipid droplets colocalization in foam cells treated with atorvastatin. The autophagy inducer, rapamycin (Rap), did not increase the lipophagy enhancement effect of atorvastatin, but the autophagy inhibitor, 3-methyladenine, suppressed the effect of atorvastatin on Atg5 expression and the LC3II/LC3I ratio, as well as the increased p62 expression, suppressed lipophagy, attenuated cholesterol efflux and increased cholesterol content in foam cells. Further analysis revealed that atorvastatin promoted lipophagy by upregulating adenosine 5'-monophosphate-activated protein kinase (AMPK) phosphorylation, and downregulating mammalian target of rapamycin phosphorylation, whereas the AMPK inhibiter, compound C, attenuated these effects. In conclusion, atorvastatin reduced lipid accumulation and promoted cholesterol efflux by enhancing lipophagy in foam cells and thereby inhibited foam cell formation. The enhanced lipophagy of foam cells was exerted through the AMPK/mammalian target of rapamycin signaling pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Atherosclerosis/drug therapy , Atorvastatin/pharmacology , Autophagy/drug effects , Cholesterol/metabolism , Foam Cells/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/metabolism , Atherosclerosis/enzymology , Atherosclerosis/pathology , Autophagy-Related Proteins/metabolism , Foam Cells/enzymology , Foam Cells/pathology , Humans , Lipid Droplets/drug effects , Lipid Droplets/metabolism , Phosphorylation , Signal Transduction , THP-1 Cells
5.
J Cardiovasc Pharmacol ; 75(4): 321-332, 2020 04.
Article in English | MEDLINE | ID: mdl-31895870

ABSTRACT

Artemisinin is an endoperoxide sesquiterpene lactone from Artemisia annua L with multiple beneficial effects, including anti-inflammation, antioxidant, and vascular protection. Recent studies have found that inflammation along with autophagy deficiency in macrophages is the possible reason for foam cell accumulation in the intima, which leads to atherosclerotic plaque formation. The primary aims of this study were to explore the inhibiting effect of artemisinin on atherosclerosis in high-fat diet-fed ApoE mice and investigate the probable mechanism. Artemisinin (50 and 100 mg/kg, intragastric administration) treatment effectively inhibited foamy macrophage transformation and decreased atherosclerotic plaque formation in atherosclerotic mice. Moreover, artemisinin promoted AMP-activated protein kinase (AMPK) activation, inhibited mammalian target of rapamycin (mTOR) and uncoordinated-51-like kinase 1 (ULK1) phosphorylation, and increased LC-3II accumulation and P62 degradation, thereby enhancing macrophage autophagy. Besides, the inhibiting effect of artemisinin on mTOR and ULK1 phosphorylation could be abrogated by AMPK knockdown, suggesting AMPK was the essential target of artemisinin on promoting macrophage autophagy. Our study indicated that artemisinin alleviated atherosclerotic lesions by accelerating macrophage autophagy through the AMPK/mTOR/ULK1 pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anti-Inflammatory Agents/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Artemisinins/pharmacology , Atherosclerosis/prevention & control , Autophagy/drug effects , Foam Cells/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Autophagy-Related Protein-1 Homolog/metabolism , Diet, High-Fat , Disease Models, Animal , Foam Cells/enzymology , Foam Cells/pathology , Lipoproteins, LDL/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Microtubule-Associated Proteins/metabolism , Phosphorylation , Plaque, Atherosclerotic , RAW 264.7 Cells , Sequestosome-1 Protein/metabolism , Signal Transduction
6.
J Cardiovasc Pharmacol ; 75(1): 45-53, 2020 01.
Article in English | MEDLINE | ID: mdl-31895879

ABSTRACT

Atherosclerosis is a chronic inflammation condition resulting from the interaction between lipoproteins, monocyte-derived macrophages, T lymphocytes, and other cellular elements in the arterial wall. Macrophage-derived foam cells play a key role in both early and advanced stage of atherosclerosis. Previous studies have shown that berberine could inhibit foam cell formation and prevent experimental atherosclerosis. However, its underlying molecular mechanisms have not been fully clarified. In this study, we explored the cholesterol-lowering effects of berberine in macrophage-derived foam cells and investigated its possible mechanisms in prevention and treatment of atherosclerosis. Here, we demonstrated that berberine could inhibit atherosclerosis in apolipoprotein E-deficient mice and induce cholesterol reduction as well as decrease the content of macrophages. Berberine can regulate oxLDL uptake and cholesterol efflux, thus suppresses foam cell formation. Mechanisms study showed that berberine can suppress scavenger receptor expression via inhibiting the activity of AP-1 and upregulate ATP-binding cassette transporter via activating Nrf2/HO-1 signaling in human macrophage. In summary, berberine significantly inhibits atherosclerotic disease development by regulating lipid homeostasis and suppressing macrophage foam cell formation.


Subject(s)
Atherosclerosis/prevention & control , Berberine/pharmacology , Cholesterol/metabolism , Cholinergic Antagonists/pharmacology , Foam Cells/drug effects , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/metabolism , Transcription Factor AP-1/metabolism , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Animals , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , CD36 Antigens/metabolism , Disease Models, Animal , Foam Cells/enzymology , Foam Cells/pathology , Heme Oxygenase-1/genetics , Humans , Lipoproteins, LDL/metabolism , Mice, Inbred C57BL , Mice, Knockout, ApoE , NF-E2-Related Factor 2/genetics , Scavenger Receptors, Class A/metabolism , THP-1 Cells
7.
Perfusion ; 35(1): 57-65, 2020 01.
Article in English | MEDLINE | ID: mdl-31170866

ABSTRACT

PURPOSE: The aim of this study was to investigate the role of miR-33-5p in abdominal aortic aneurysm progression, which regulated adenosine triphosphate-binding cassette transporter A1 (ABCA1)-mediated cholesterol efflux and lipid accumulation in THP-1 macrophage-derived foam cells through the PI3K/Akt pathway. METHODS: Quantitative reverse transcription polymerase chain reaction was used to evaluate the expression level of miR-33-5p and ABCA1 mRNA in abdominal aortic aneurysm patient and normal person tissues. The relationship between miR-33-5p and ABCA1 was examined by dual luciferase report assay. High-performance liquid chromatography was used to evaluate the levels of cholesterol contents. Cholesterol efflux detection was performed by liquid scintillator. The expression of inflammatory cytokines was detected by quantitative reverse transcription polymerase chain reaction. Western blot was applied to determine the expression levels of ABCA1, PI3K (p-PI3K), and Akt (p-Akt). RESULTS: The quantitative reverse transcription polymerase chain reaction analysis results revealed miR-33-5p overexpression in abdominal aortic aneurysm tissues, but the expression level of ABCA1 was lower in abdominal aortic aneurysm tissues than non-abdominal aortic aneurysm tissues. Subsequently, the dual luciferase report gene assay confirmed that ABCA1 was a target of miR-33-5p, and miR-33-5p-negative regulated ABCA1 expression. Moreover, the expression levels of p-PI3K, p-Akt, and ABCA1 were decreased in THP-1 cell transferred with ABCA1 siRNA, but knockdown of miR-33-5p had an opposite effect. Furthermore, knockdown of miR-33-5p decreased the expression of MMP-2, MMP-9, TNF-α, total cellular cholesterol, and promoted cholesterol efflux in THP-1-derived foam cells. Importantly, LY294002 (PI3K inhibitor) or si-ABCA1 completely inhibited the stimulatory effects of miR-33-5p inhibitor. CONCLUSION: This study has found that knockdown of miR-33-5p induced ABCA1 expression and promoted inflammatory cytokines and cholesterol efflux likely via activating the PI3K/Akt signaling pathway.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Aorta, Abdominal/enzymology , Aortic Aneurysm, Abdominal/prevention & control , Foam Cells/enzymology , Gene Knockdown Techniques , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , ATP Binding Cassette Transporter 1/genetics , Aged , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/enzymology , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/pathology , Case-Control Studies , Cholesterol/metabolism , Cytokines/metabolism , Disease Progression , Female , Foam Cells/pathology , Humans , Inflammation Mediators/metabolism , Male , MicroRNAs/genetics , Middle Aged , Signal Transduction , THP-1 Cells , Up-Regulation
8.
Int J Mol Sci ; 22(1)2020 Dec 28.
Article in English | MEDLINE | ID: mdl-33379334

ABSTRACT

The development, progression, or stabilization of the atherosclerotic plaque depends on the pro-inflammatory and anti-inflammatory macrophages. The influx of the macrophages and the regulation of macrophage phenotype, inflammatory or anti-inflammatory, are controlled by the small GTPase RhoA and its downstream effectors. Therefore, macrophages and the components of the RhoA pathway are attractive targets for anti-atherosclerotic therapies, which would inhibit macrophage influx and inflammatory phenotype, maintain an anti-inflammatory environment, and promote tissue remodeling and repair. Here, we discuss the recent findings on the role of macrophages and RhoA pathway in the atherosclerotic plaque formation and resolution and the novel therapeutic approaches.


Subject(s)
Arteries/metabolism , Atherosclerosis/immunology , Foam Cells/enzymology , Lipid Metabolism , Plaque, Atherosclerotic/immunology , rhoA GTP-Binding Protein/metabolism , Animals , Humans
9.
Int J Mol Sci ; 21(23)2020 Nov 27.
Article in English | MEDLINE | ID: mdl-33261140

ABSTRACT

The dysregulation of macrophage lipid metabolism drives atherosclerosis. AMP-activated protein kinase (AMPK) is a master regulator of cellular energetics and plays essential roles regulating macrophage lipid dynamics. Here, we investigated the consequences of atherogenic lipoprotein-induced foam cell formation on downstream immunometabolic signaling in primary mouse macrophages. A variety of atherogenic low-density lipoproteins (acetylated, oxidized, and aggregated forms) activated AMPK signaling in a manner that was in part due to CD36 and calcium-related signaling. In quiescent macrophages, basal AMPK signaling was crucial for maintaining markers of lysosomal homeostasis as well as levels of key components in the lysosomal expression and regulation network. Moreover, AMPK activation resulted in targeted upregulation of members of this network via transcription factor EB. However, in lipid-induced macrophage foam cells, neither basal AMPK signaling nor its activation affected lysosomal-associated programs. These results suggest that while the sum of AMPK signaling in cultured macrophages may be anti-atherogenic, atherosclerotic input dampens the regulatory capacity of AMPK signaling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy , Foam Cells/enzymology , Homeostasis , Lysosomes/metabolism , Animals , Atherosclerosis/metabolism , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , CD36 Antigens/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cells, Cultured , Enzyme Activation , Female , Lipid Metabolism , Lipoproteins/metabolism , Male , Mice , Mice, Knockout , Signal Transduction , Transcription, Genetic , Up-Regulation/genetics
10.
J Cell Mol Med ; 23(10): 6611-6621, 2019 10.
Article in English | MEDLINE | ID: mdl-31436906

ABSTRACT

The contribution of soluble epoxide hydrolase (sEH) to atherosclerosis has been well defined. However, less is understood about the role of sEH and its underlying mechanism in the cholesterol metabolism of macrophages. The expression of sEH protein was increased in atherosclerotic aortas of apolipoprotein E-deficient mice, primarily in macrophage foam cells. Oxidized low-density lipoprotein (oxLDL) increased sEH expression in macrophages. Genetic deletion of sEH (sEH-/- ) in macrophages markedly exacerbated oxLDL-induced lipid accumulation and decreased the expression of ATP-binding cassette transporters-A1 (ABCA1) and apolipoprotein AI-dependent cholesterol efflux following oxLDL treatment. The down-regulation of ABCA1 in sEH-/- macrophages was due to an increase in the turnover rate of ABCA1 protein but not in mRNA transcription. Inhibition of phosphatase activity, but not hydrolase activity, of sEH decreased ABCA1 expression and cholesterol efflux following oxLDL challenge, which resulted in increased cholesterol accumulation. Additionally, oxLDL increased the phosphatase activity, promoted the sEH-ABCA1 complex formation and decreased the phosphorylated level of ABCA1 at threonine residues. Overexpression of phosphatase domain of sEH abrogated the oxLDL-induced ABCA1 phosphorylation and further increased ABCA1 expression and cholesterol efflux, leading to the attenuation of oxLDL-induced cholesterol accumulation. Our findings suggest that the phosphatase domain of sEH plays a crucial role in the cholesterol metabolism of macrophages.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , Atherosclerosis/enzymology , Cholesterol/metabolism , Epoxide Hydrolases/genetics , Epoxide Hydrolases/metabolism , Foam Cells/enzymology , Macrophages/enzymology , Phosphoprotein Phosphatases/metabolism , ATP Binding Cassette Transporter 1/metabolism , Animals , Apolipoproteins E/deficiency , Atherosclerosis/genetics , Atherosclerosis/metabolism , Epoxide Hydrolases/antagonists & inhibitors , Foam Cells/metabolism , Lipid Metabolism , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Binding
11.
Arterioscler Thromb Vasc Biol ; 38(12): 2780-2792, 2018 12.
Article in English | MEDLINE | ID: mdl-30571174

ABSTRACT

Objective- Monocyte-derived foam cells are one of the key players in the formation of atherosclerotic plaques. Adenosine receptors and extracellular adenosine have been demonstrated to modulate foam cell formation. ADK (adenosine kinase) is a major enzyme regulating intracellular adenosine levels, but its functional role in myeloid cells remains poorly understood. To enhance intracellular adenosine levels in myeloid cells, ADK was selectively deleted in novel transgenic mice using Cre-LoxP technology, and foam cell formation and the development of atherosclerotic lesions were determined. Approach and Results- ADK was upregulated in macrophages on ox-LDL (oxidized low-density lipoprotein) treatment in vitro and was highly expressed in foam cells in atherosclerotic plaques. Atherosclerotic mice deficient in ADK in myeloid cells were generated by breeding floxed ADK (ADKF/F) mice with LysM-Cre (myeloid-specific Cre recombinase expressing) mice and ApoE-/- (apolipoprotein E deficient) mice. Mice absent ADK in myeloid cells exhibited much smaller atherosclerotic plaques compared with controls. In vitro assays showed that ADK deletion or inhibition resulted in increased intracellular adenosine and reduced DNA methylation of the ABCG1 (ATP-binding cassette transporter G1) gene. Loss of methylation was associated with ABCG1 upregulation, enhanced cholesterol efflux, and eventually decreased foam cell formation. Conclusions- Augmentation of intracellular adenosine levels through ADK knockout in myeloid cells protects ApoE-/- mice against atherosclerosis by reducing foam cell formation via the epigenetic regulation of cholesterol trafficking. ADK inhibition is a promising approach for the treatment of atherosclerotic diseases.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , Adenosine Kinase/deficiency , Aorta/enzymology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Epigenesis, Genetic , Foam Cells/enzymology , Mice, Knockout, ApoE , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Adenosine Kinase/genetics , Animals , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Cholesterol/metabolism , DNA Methylation , Disease Models, Animal , Female , Foam Cells/pathology , Male , Mice, Inbred C57BL , Plaque, Atherosclerotic , Signal Transduction
12.
Arterioscler Thromb Vasc Biol ; 38(2): 324-334, 2018 02.
Article in English | MEDLINE | ID: mdl-29217509

ABSTRACT

OBJECTIVE: Macrophage proinflammatory responses induced by modified low-density lipoproteins (modLDL) contribute to atherosclerotic progression. How modLDL causes macrophages to become proinflammatory is still enigmatic. Macrophage foam cell formation induced by modLDL requires glycerolipid synthesis. Lipin-1, a key enzyme in the glycerolipid synthesis pathway, contributes to modLDL-elicited macrophage proinflammatory responses in vitro. The objective of this study was to determine whether macrophage-associated lipin-1 contributes to atherogenesis and to assess its role in modLDL-mediated signaling in macrophages. APPROACH AND RESULTS: We developed mice lacking lipin-1 in myeloid-derived cells and used adeno-associated viral vector 8 expressing the gain-of-function mutation of mouse proprotein convertase subtilisin/kexin type 9 (adeno-associated viral vector 8-proprotein convertase subtilisin/kexin type 9) to induce hypercholesterolemia and plaque formation. Mice lacking myeloid-associated lipin-1 had reduced atherosclerotic burden compared with control mice despite similar plasma lipid levels. Stimulation of bone marrow-derived macrophages with modLDL activated a persistent protein kinase Cα/ßII-extracellular receptor kinase1/2-jun proto-oncogene signaling cascade that contributed to macrophage proinflammatory responses that was dependent on lipin-1 enzymatic activity. CONCLUSIONS: Our data demonstrate that macrophage-associated lipin-1 is atherogenic, likely through persistent activation of a protein kinase Cα/ßII-extracellular receptor kinase1/2-jun proto-oncogene signaling cascade that contributes to foam cell proinflammatory responses. Taken together, these results suggest that modLDL-induced foam cell formation and modLDL-induced macrophage proinflammatory responses are not independent consequences of modLDL stimulation but rather are both directly influenced by enhanced lipid synthesis.


Subject(s)
Aorta/enzymology , Aortic Diseases/enzymology , Atherosclerosis/enzymology , Inflammation Mediators/metabolism , Inflammation/enzymology , Lipoproteins, LDL/blood , Macrophages/enzymology , Nuclear Proteins/metabolism , Phosphatidate Phosphatase/metabolism , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Foam Cells/enzymology , Foam Cells/pathology , Inflammation/genetics , Inflammation/pathology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phosphatidate Phosphatase/deficiency , Phosphatidate Phosphatase/genetics , Plaque, Atherosclerotic , Protein Kinase C beta/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RAW 264.7 Cells , Signal Transduction
13.
Circ J ; 83(12): 2555-2566, 2019 11 25.
Article in English | MEDLINE | ID: mdl-31666457

ABSTRACT

BACKGROUND: Accumulation of foam cells in the neointima represents an early stage of atherosclerosis. 1-trifluoromethoxyphenyl-3-(1-propionylpiperidine-4-yl) urea (TPPU), a novel soluble epoxide hydrolase inhibitor (sEHi), effectively elevates epoxyeicosatrienoic acid (EET) levels. The effects of EETs on macrophages foam cells formation are poorly understood.Methods and Results:Incubation of foam cells with TPPU markedly ameliorate cholesterol deposition in oxidized low-density lipoprotein (oxLDL)-loaded macrophages by increasing the levels of EETs. Notably, TPPU treatment significantly inhibits oxLDL internalization and promotes cholesterol efflux. The elevation of EETs results in a decrease of class A scavenger receptor (SR-A) expression via downregulation of activator protein 1 (AP-1) expression. Additionally, TPPU selectively increases protein but not the mRNA level of ATP-binding cassette transporter A1 (ABCA1) through the reduction of calpain activity that stabilizes the protein. Moreover, TPPU treatment reduces the cholesterol content of macrophages and inhibits atherosclerotic plaque formation in apolipoprotein E-deficient mice. These changes induced by TPPU are dependent on heme oxygenase-1 (HO-1) activation. CONCLUSIONS: The present study findings elucidate a precise mechanism of regulating cholesterol uptake and efflux in macrophages, which involves the prevention of atherogenesis by increasing the levels of EETs with TPPU.


Subject(s)
Atherosclerosis/drug therapy , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Foam Cells/drug effects , Heme Oxygenase-1/metabolism , Macrophages/drug effects , Membrane Proteins/metabolism , Phenylurea Compounds/pharmacology , Piperidines/pharmacology , ATP Binding Cassette Transporter 1/metabolism , Animals , Arachidonic Acids/metabolism , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Calpain/metabolism , Cholesterol/metabolism , Disease Models, Animal , Epoxide Hydrolases/metabolism , Foam Cells/enzymology , Foam Cells/pathology , Humans , Lipoproteins, LDL/metabolism , Macrophages/enzymology , Macrophages/pathology , Male , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Scavenger Receptors, Class A/metabolism , Signal Transduction , THP-1 Cells
14.
Clin Sci (Lond) ; 132(6): 719-738, 2018 03 30.
Article in English | MEDLINE | ID: mdl-29382697

ABSTRACT

Growing evidence implicates cyclophilin A secreted by vascular wall cells and monocytes as a key mediator in atherosclerosis. Cyclophilin A in addition to its proliferative effects, during hyperglycemic conditions, increases lipid uptake in macrophages by increasing scavenger receptors on the cell's surface. It also promotes macrophage migration across endothelial cells and conversion of macrophages into foam cells. Given the known effects of metformin in reducing vascular complications of diabetes, we investigated the effect of metformin on cyclophilin A action in macrophages. Using an ex vivo model of cultured macrophages isolated from patients with type 2 diabetes with and without coronary artery disease (CAD), we measured the effect of metformin on cyclophilin A expression, lipid accumulation, expression of scavenger receptors, plasma cytokine levels and AMP-activated protein kinase (AMPK) activity in macrophages. In addition, the effects of metformin on migration of monocytes, reactive oxygen species (ROS) formation, lipid uptake in the presence of cyclophilin A inhibitors and comparison with pioglitazone were studied using THP-1 monocytes. Metformin reduced cyclophilin A expression in human monocyte-derived macrophages. Metformin also decreased the effects of cyclophilin A on macrophages such as oxidized low-density lipoprotein (oxLDL) uptake, scavenger receptor expression, ROS formation and secretion of inflammatory cytokines in high-glucose conditions. Metformin reversed cyclophilin A-induced decrease in AMPK-1α activity in macrophages. These effects of metformin were similar to those of cyclophilin A inhibitors. Metformin can thus function as a suppressor of pro-inflammatory effects of cyclophilin A in high-glucose conditions by attenuating its expression and repressing cyclophilin A-induced decrease in AMPK-1α activity in macrophages.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cyclophilin A/blood , Cytokines/metabolism , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Lipid Metabolism/drug effects , Macrophages/drug effects , Metformin/pharmacology , Adult , Aged , Case-Control Studies , Cell Movement/drug effects , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/enzymology , Foam Cells/drug effects , Foam Cells/enzymology , Humans , Lipoproteins, LDL/metabolism , Macrophages/enzymology , Middle Aged , Oxidative Stress/drug effects , Pioglitazone/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , THP-1 Cells
15.
Arterioscler Thromb Vasc Biol ; 36(9): 1937-46, 2016 09.
Article in English | MEDLINE | ID: mdl-27417584

ABSTRACT

OBJECTIVE: Activation of the inflammasome pathway in macrophages results in the secretion of 2 potent proinflammatory and proatherogenic cytokines, interleukin (IL)-1ß, and IL-18. Atherosclerotic lesions are characterized by the presence of various endogenous activators of the NLR family pyrin domain containing 3 (NLRP3) inflammasome, including cholesterol crystals and extracellular ATP. The aim of this study was to comprehensively characterize the expression of inflammasome pathway components and regulators in human atherosclerotic lesions. APPROACH AND RESULTS: Twenty human coronary artery RNA samples from 10 explanted hearts were analyzed using an inflammasome pathway-focused quantitative polymerase chain reaction array. Advanced atherosclerotic plaques, when compared with early-to-intermediate lesions from the same coronary trees, displayed significant upregulation of 12 target genes, including the key inflammasome components apoptosis-associated speck-like protein containing a CARD domain, caspase-1, and IL-18. Immunohistochemical stainings of the advanced plaques revealed macrophage foam cells positive for NLRP3 inflammasome components around the necrotic lipid cores. The polymerase chain reaction array target p38δ mitogen-activated protein kinase was upregulated in advanced plaques and strongly expressed by lesional macrophage foam cells. In cultured human monocyte-derived macrophages, the p38δ mitogen-activated protein kinase was activated by intracellular stress signals triggered during ATP- and cholesterol crystal-induced NLRP3 inflammasome activation and was required for NLRP3-mediated IL-1ß secretion. CONCLUSIONS: Increased expression of the key inflammasome components in advanced coronary lesions implies enhanced activity of the inflammasome pathway in progression of coronary atherosclerosis. The p38δ mitogen-activated protein kinase was identified as a novel regulator of NLRP3 inflammasome activation in primary human macrophages, and thus, represents a potential target for modulation of atherosclerotic inflammation.


Subject(s)
Coronary Artery Disease/enzymology , Coronary Vessels/enzymology , Foam Cells/enzymology , Inflammasomes/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Plaque, Atherosclerotic , Adenosine Triphosphate/metabolism , Cells, Cultured , Cholesterol/metabolism , Coronary Artery Disease/genetics , Coronary Artery Disease/pathology , Coronary Vessels/pathology , Crystallization , Enzyme Activation , Foam Cells/pathology , Gene Expression Profiling/methods , Histocompatibility Antigens Class II/metabolism , Humans , Inflammasomes/genetics , Male , Middle Aged , Mitogen-Activated Protein Kinase 13/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Necrosis , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Primary Cell Culture , Real-Time Polymerase Chain Reaction , Signal Transduction , Time Factors , Trans-Activators/metabolism , Up-Regulation
16.
Cardiovasc Drugs Ther ; 31(1): 19-27, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28084571

ABSTRACT

PURPOSE: Xanthine oxidase catalyzes the oxidation of xanthine to uric acid. This process generates excessive reactive oxygen species (ROS) that play an important role in atherogenesis. Recent studies show that LRR and PYD domains-containing protein 3 (NLRP3), a component of the inflammasome, may be involved in the formation of foam cells, a hallmark of atherosclerosis. This study was designed to study the role of various scavenger receptors and NLRP3 inflammasome in xanthine oxidase and uric acid-induced foam cell formation. METHODS AND RESULTS: Human vascular smooth muscle cells (VSMCs) and THP-1 macrophages were treated with xanthine oxidase or uric acid. Xanthine oxidase treatment (of both VSMCs and THP-1 cells) resulted in foam cell formation in concert with generation of ROS and expression of cluster of differentiation 36 (CD36) and oxidized low density lipoprotein (lectin-like) receptor 1 (LOX-1), but not of scavenger receptor A (SRA). Uric acid treatment resulted in foam cell formation, ROS generation and expression of CD36, but not of LOX-1 or SRA. Further, treatment of cells with xanthine oxidase, but not uric acid, activated NLRP3 and its downstream pro-inflammatory signals- caspase-1, interleukin (IL)-1ß and IL-18. Blockade of LOX-1 or NLRP3 inflammasome with specific siRNAs reduced xanthine oxidase-induced foam cell formation, ROS generation and activation of NLRP3 and downstream signals. CONCLUSIONS: Xanthine oxidase induces foam cell formation in large part through activation of LOX-1 - NLRP3 pathway in both VSMCs and THP-1 cells, but uric acid-induced foam cell formation is exclusively through CD36 pathway. Further, LOX-1 activation is upstream of NLRP3 activation. Graphical Abstract Steps in the formation of foam cells in response to xanthine oxidase and uric acid. Xanthine oxidase stimulates LOX-1 expression on the cell membrane of macrophages and vascular smooth muscle cells (VSMCs) and increases generation of ROS, which activate NLRP3 inflammasome and downstream pro-inflammatory mediators such as Caspase-1, IL-1ß and IL-18. Xanthine oxidase also induces CD36 expression. Activation of both LOX-1 and CD36 (LOX-1> > CD36) participates in the transformation of macrophages and VSMCs into foam cells. Uric acid formed from xanthine-xanthine oxidase interaction stimulates CD36 expression and triggers foam cell formation independent of NLRP3 activation.


Subject(s)
Cell Transdifferentiation/drug effects , Foam Cells/drug effects , Inflammasomes/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Scavenger Receptors, Class E/metabolism , Xanthine Oxidase/pharmacology , CD36 Antigens/metabolism , Cell Line, Tumor , Foam Cells/enzymology , Humans , Inflammasomes/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , RNA Interference , Reactive Oxygen Species/metabolism , Scavenger Receptors, Class A/metabolism , Scavenger Receptors, Class E/genetics , Signal Transduction/drug effects , Transfection , Uric Acid/pharmacology
17.
J Vasc Surg ; 64(4): 1074-83, 2016 10.
Article in English | MEDLINE | ID: mdl-26054589

ABSTRACT

OBJECTIVE: Effective treatments against restenosis after percutaneous transluminal angioplasty and stenting are largely lacking. Human tissue kallikrein gene transfer has been shown to be able to attenuate neointima formation induced by balloon catheter. As a tissue kallikrein in vivo, human urinary kininogenase (HUK) is widely used to prevent ischemia-reperfusion injury. However, the effects of HUK on neointima formation have not been explored. We therefore investigated whether HUK could alleviate balloon catheter-induced intimal hyperplasia in rabbits fed with high-fat diets. METHODS: The effects of HUK on neointima and atherosclerosis formation were analyzed by hematoxylin-eosin staining and immunohistochemical staining in balloon-injured carotid arteries of rabbits. Local inflammatory response was evaluated by detecting the gene expression of tumor necrosis factor α and interleukin 1ß with real-time quantitative polymerase chain reaction plus the invasion of macrophages with immunohistochemical staining. Western blotting was employed to investigate the effects of HUK on activities of endothelial nitric oxide synthase (eNOS), transforming growth factor ß1 (TGF-ß1), and Smad signaling pathway. The long-term effect of HUK on intimal hyperplasia of the injured carotid artery was assessed by angiography. RESULTS: Quantitative image analysis showed that intravenous administration of HUK for 14 days significantly decreased the intimal areas and intima area/media area ratios (day 14, 54% decrease in intimal area and 58% decrease in intima area/media area ratios; day 28, 63% and 85%). Significant decreases were also noted in macrophage foam cell-positive area after 7-day or 14-day administration of HUK (day 7, 69% decrease in intimal area and 78% decrease in media area; day 14, 79% and 60%; day 28, 68% and 44%). Actin staining for smooth muscle cells in neointima at 2 months showed similar results (vascular smooth muscle cell-positive area of neointima, 28.21% ± 5.58% vs 43.78% ± 8.36%; P < .05). Real-time quantitative polymerase chain reaction or Western blot analysis showed that HUK reduced expression of tumor necrosis factor α, interleukin 1ß, TGF-ß1, and p-Smad2/3 but increased the expression of p-eNOS. Angiography analysis showed that 14-day administration of HUK significantly decreased the degree of stenosis (26.8% ± 7.1% vs 47.9% ± 5.7%; P < .01) at 2 months after balloon injury. CONCLUSIONS: Our results indicate that HUK is able to attenuate atherosclerosis formation and to inhibit intimal hyperplasia by downregulating TGF-ß1 expression and Smad2/3 phosphorylation, upregulating eNOS activity. HUK may be a potential therapeutic agent to prevent stenosis after vascular injury.


Subject(s)
Angioplasty, Balloon/adverse effects , Carotid Arteries/drug effects , Carotid Artery Injuries/drug therapy , Carotid Stenosis/prevention & control , Kallikreins/pharmacology , Neointima , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Urine/chemistry , Administration, Intravenous , Angiography, Digital Subtraction , Animals , Carotid Arteries/enzymology , Carotid Arteries/pathology , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Carotid Stenosis/enzymology , Carotid Stenosis/genetics , Carotid Stenosis/pathology , Diet, High-Fat , Disease Models, Animal , Drug Administration Schedule , Foam Cells/drug effects , Foam Cells/enzymology , Foam Cells/pathology , Humans , Hyperplasia , Inflammation Mediators/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Kallikreins/administration & dosage , Kallikreins/urine , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Rabbits , Signal Transduction/drug effects , Time Factors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
Arterioscler Thromb Vasc Biol ; 35(1): 79-86, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25395616

ABSTRACT

OBJECTIVE: The protein degrading activity of cathepsin C (CatC), combined with its role in leukocyte granule activation, suggests a contribution of this cystein protease in atherosclerosis. However, no experimental data are available to validate this concept. APPROACH AND RESULTS: CatC gene and protein expression were increased in ruptured versus advanced stable human carotid artery lesions. To assess causal involvement of CatC in plaque progression and stability, we generated LDLr(-/-)//CatC(-/-) chimeras by bone marrow transplantation. CatC(-/-) chimeras presented attenuated plaque burden in carotids, descending aorta, aortic arch and root, at both the early and advanced plaque stage. CatC was abundantly expressed by plaque macrophages and foam cells. CatC expression and activity were dramatically downregulated in plaques of CatC(-/-) chimeras, supporting a hematopoietic origin of plaque CatC. Our studies unveiled an unexpected feedback of CatC deficiency on macrophage activation programs and T helper cell differentiation in as much as that CatC expression was upregulated in M1 macrophages, whereas its deficiency led to combined M2 (in vitro) and Th2 polarization (in vivo). CONCLUSIONS: Our data implicate CatC has a role in the selective tuning of innate and adaptive immune responses, relevant to a chronic immune disease, such as atherosclerosis.


Subject(s)
Adaptive Immunity , Aorta/enzymology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Carotid Arteries/enzymology , Carotid Artery Diseases/prevention & control , Cathepsin C/metabolism , Immunity, Innate , Leukocytes/enzymology , Animals , Aorta/immunology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Carotid Arteries/immunology , Carotid Arteries/pathology , Carotid Artery Diseases/enzymology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/immunology , Carotid Artery Diseases/pathology , Cathepsin C/genetics , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Disease Progression , Down-Regulation , Female , Foam Cells/enzymology , Foam Cells/immunology , Humans , Leukocytes/immunology , Macrophage Activation , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , T-Lymphocytes, Helper-Inducer/enzymology , T-Lymphocytes, Helper-Inducer/immunology , Time Factors
19.
Arterioscler Thromb Vasc Biol ; 35(4): 948-59, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25810299

ABSTRACT

OBJECTIVE: Activation of liver X receptor (LXR) inhibits atherosclerosis but induces hypertriglyceridemia. In vitro, it has been shown that mitogen-activated protein kinase kinase 1/2 (MEK1/2) inhibitor synergizes LXR ligand-induced macrophage ABCA1 expression and cholesterol efflux. In this study, we determined whether MEK1/2 (U0126) and LXR ligand (T0901317) can have a synergistic effect on the reduction of atherosclerosis while eliminating LXR ligand-induced fatty livers and hypertriglyceridemia. We also set out to identify the cellular mechanisms of the actions. APPROACH AND RESULTS: Wild-type mice were used to determine the effect of U0126 on a high-fat diet or high-fat diet plus T0901317-induced transient dyslipidemia and liver injury. ApoE deficient (apoE(-/-)) mice or mice with advanced lesions were used to determine the effect of the combination of T0901317 and U0126 on atherosclerosis and hypertriglyceridemia. We found that U0126 protected animals against T0901317-induced transient or long-term hepatic lipid accumulation, liver injury, and hypertriglyceridemia. Meanwhile, the combination of T0901317 and U0126 inhibited the development of atherosclerosis in a synergistic manner and reduced advanced lesions. Mechanistically, in addition to synergistic induction of macrophage ABCA1 expression, the combination of U0126 and T0901317 maintained arterial wall integrity, inhibited macrophage accumulation in aortas and formation of macrophages/foam cells, and activated reverse cholesterol transport. The inhibition of T0901317-induced lipid accumulation by the combined U0126 might be attributed to inactivation of lipogenesis and activation of lipolysis/fatty acid oxidation pathways. CONCLUSIONS: Our study suggests that the combination of mitogen-activated protein kinase kinase 1/2 inhibitor and LXR ligand can function as a novel therapy to synergistically reduce atherosclerosis while eliminating LXR-induced deleterious effects.


Subject(s)
Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Butadienes/pharmacology , Hydrocarbons, Fluorinated/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Nitriles/pharmacology , Orphan Nuclear Receptors/agonists , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology , Animals , Aorta/drug effects , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/genetics , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Cholesterol/metabolism , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Fatty Liver/chemically induced , Fatty Liver/enzymology , Fatty Liver/pathology , Fatty Liver/prevention & control , Female , Foam Cells/drug effects , Foam Cells/enzymology , Foam Cells/pathology , Hep G2 Cells , Humans , Hydrocarbons, Fluorinated/toxicity , Hypertriglyceridemia/chemically induced , Hypertriglyceridemia/enzymology , Hypertriglyceridemia/pathology , Hypertriglyceridemia/prevention & control , Liver/drug effects , Liver/metabolism , Liver X Receptors , Male , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Orphan Nuclear Receptors/metabolism , Signal Transduction/drug effects , Sulfonamides/toxicity
20.
J Lipid Res ; 56(5): 1025-33, 2015 May.
Article in English | MEDLINE | ID: mdl-25773887

ABSTRACT

Atherosclerosis stems from imbalances in lipid metabolism and leads to maladaptive inflammatory responses. The AMP-activated protein kinase (Ampk) is a highly conserved serine/threonine kinase that regulates many aspects of lipid and energy metabolism, although its specific role in controlling macrophage cholesterol homeostasis remains unclear. We sought to address this question by testing the effects of direct Ampk activators in primary bone marrow-derived macrophages from Ampk ß1-deficient (ß1(-/-)) mice. Macrophages from Ampk ß1(-/-) mice had enhanced lipogenic capacity and diminished cholesterol efflux, although cholesterol uptake was unaffected. Direct activation of Ampk ß1 via salicylate (the unacetylated form of aspirin) or A-769662 (a small molecule activator), decreased the synthesis of FAs and sterols in WT but not Ampk ß1(-/-) macrophages. In lipid-laden macrophages, Ampk activation decreased cholesterol content (foam cell formation) and increased cholesterol efflux to HDL and apoA-I, effects that occurred in an Ampk ß1-dependent manner. Increased cholesterol efflux was also associated with increased gene expression of the ATP binding cassette transporters, Abcg1 and Abca1. Moreover, in vivo reverse cholesterol transport was suppressed in mice that received Ampk ß1(-/-) macrophages compared with the WT control. Our data highlight the therapeutic potential of targeting macrophage Ampk with new or existing drugs for the possible reduction in foam cell formation during the early stages of atherosclerosis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cholesterol/metabolism , Enzyme Activators/pharmacology , Foam Cells/enzymology , Salicylic Acid/pharmacology , Animals , Apolipoprotein A-I/metabolism , Atherosclerosis , Cells, Cultured , Cholesterol, HDL/metabolism , Drug Evaluation, Preclinical , Enzyme Activation , Foam Cells/drug effects , Homeostasis , Lipogenesis , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL