Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 33(7): 7942-7952, 2019 07.
Article in English | MEDLINE | ID: mdl-30922124

ABSTRACT

Hypersecretion of hepatic very LDL (VLDL)-associated triglyceride (TG) is the hallmark of hypertriglyceridemia. The estrogen-related receptor γ (ERRγ), an orphan nuclear receptor, plays crucial roles in the regulation of metabolic homeostasis, including TG formation in the liver. It remains unclear whether ERRγ regulates hepatic VLDL-TG secretion. We demonstrated that knockdown of ERRγ impairs hepatic VLDL-TG secretion in mice, whereas overexpression of ERRγ favors the secretion, indicating a novel role of ERRγ in hepatic TG metabolism. We found that ERRγ transcriptionally regulates the expression of PLA2G12B by binding to the promoter region of the Pla2g12b gene. In Pla2g12b-null mice, ERRγ fails to regulate hepatic VLDL-TG secretion. There is an apparent accumulation of large lipid droplets in the liver of Pla2g12b-null mice. These data suggest that ERRγ is a novel regulator of hepatic VLDL-TG secretion, which is mediated through the action on PLA2G12B.-Chen, L., Wu, M., Zhang, S., Tan, W., Guan, M., Feng, L., Chen, C., Tao, J., Chen, L., Qu, L. Estrogen-related receptor γ regulates hepatic triglyceride metabolism through phospholipase A2 G12B.


Subject(s)
Group X Phospholipases A2/physiology , Lipoproteins, VLDL/metabolism , Liver/metabolism , Receptors, Estrogen/physiology , Triglycerides/metabolism , Animals , Cell Line , Cholesterol/blood , Gene Knockdown Techniques , Group X Phospholipases A2/deficiency , Group X Phospholipases A2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Receptors, Estrogen/genetics , Recombinant Proteins/metabolism , Transcription, Genetic , Triglycerides/blood , Up-Regulation
2.
Virology ; 454-455: 78-92, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24725934

ABSTRACT

The role of Group X secreted phospholipase A2 (GX-sPLA2) during influenza infection has not been previously investigated. We examined the role of GX-sPLA2 during H1N1 pandemic influenza infection in a GX-sPLA2 gene targeted mouse (GX(-/-)) model and found that survival after infection was significantly greater in GX(-/-) mice than in GX(+/+) mice. Downstream products of GX-sPLA2 activity, PGD2, PGE2, LTB4, cysteinyl leukotrienes and Lipoxin A4 were significantly lower in GX(-/-) mice BAL fluid. Lung microarray analysis identified an earlier and more robust induction of T and B cell associated genes in GX(-/-) mice. Based on the central role of sPLA2 enzymes as key initiators of inflammatory processes, we propose that activation of GX-sPLA2 during H1N1pdm infection is an early step of pulmonary inflammation and its inhibition increases adaptive immunity and improves survival. Our findings suggest that GX-sPLA2 may be a potential therapeutic target during influenza.


Subject(s)
Group X Phospholipases A2/deficiency , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Animals , B-Lymphocytes/immunology , Disease Models, Animal , Gene Expression Profiling , Group X Phospholipases A2/genetics , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Survival Analysis , T-Lymphocytes/immunology
3.
Arterioscler Thromb Vasc Biol ; 33(3): 466-73, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23349189

ABSTRACT

OBJECTIVE: Several secreted phospholipases A2 (sPLA2s), including group IIA, III, V, and X, have been linked to the development of atherosclerosis, which led to the clinical testing of A-002 (varespladib), a broad sPLA2 inhibitor for the treatment of coronary artery disease. Group X sPLA2 (PLA2G10) has the most potent hydrolyzing activity toward phosphatidylcholine and is believed to play a proatherogenic role. METHODS AND RESULTS: Here, we show that Ldlr(-/-) mice reconstituted with bone marrow from mouse group X-deficient mice (Pla2g10(-/-)) unexpectedly display a doubling of plaque size compared with Pla2g10(+/+) chimeric mice. Macrophages of Pla2g10(-/-) mice are more susceptible to apoptosis in vitro, which is associated with a 4-fold increase of plaque necrotic core in vivo. In addition, chimeric Pla2g10(-/-) mice show exaggerated T lymphocyte (Th)1 immune response, associated with enhanced T-cell infiltration in atherosclerotic plaques. Interestingly, overexpression of human PLA2G10 in murine bone marrow cells leads to significant reduction of Th1 response and to 50% reduction of lesion size. CONCLUSIONS: PLA2G10 expression in bone marrow cells controls a proatherogenic Th1 response and limits the development of atherosclerosis. The results may provide an explanation for the recently reported inefficacy of A-002 (varespladib) to treat patients with coronary artery disease. Indeed, A-002 is a nonselective sPLA2 inhibitor that inhibits both proatherogenic (groups IIA and V) and antiatherogenic (group X) sPLA2s. Our results suggest that selective targeting of individual sPLA2 enzymes may be a better strategy to treat cardiovascular diseases.


Subject(s)
Aorta, Thoracic/enzymology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Group X Phospholipases A2/metabolism , Receptors, LDL/deficiency , Adaptive Immunity , Animals , Aorta, Thoracic/immunology , Aorta, Thoracic/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Apoptosis , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Bone Marrow Transplantation , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Group X Phospholipases A2/deficiency , Group X Phospholipases A2/genetics , Humans , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Necrosis , Plaque, Atherosclerotic , Receptors, LDL/genetics , Th1 Cells/immunology , Time Factors
4.
Am J Physiol Heart Circ Physiol ; 302(1): H95-104, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21984544

ABSTRACT

Group X secretory PLA(2) (sPLA(2)-X) is expressed in neutrophils and plays a role in the pathogenesis of neutrophil-mediated tissue inflammation and injury. This study tested the hypothesis that sPLA(2)-X in neutrophils may contribute to the pathogenesis of abdominal aortic aneurysms (AAA) using sPLA(2)-X(-/-) mice. AAA was created by application of CaCl(2) to external surface of aorta. As a result, the aortas of sPLA(2)-X(-/-) mice had smaller diameters (percent increase from baseline; 24.8 ± 3.5% vs. 49.9 ± 9.1%, respectively; P < 0.01), a reduced grade of elastin degradation, and lower activities of elastase and gelatinase (26% and 19% lower, respectively) after CaCl(2) treatment compared with sPLA(2)-X(+/+) mice. In sPLA(2)-X(+/+) mice, immunofluorescence microscopic images showed that the immunoreactivity of sPLA(2)-X was detected only in neutrophils within aortic walls 3 days, 1, 2, and 6 wk after CaCl(2) treatment, whereas the immunoreactivity was not detected in macrophages or mast cells in aortic walls. sPLA(2)-X immunoreactivity also was colocalized in cells expressing matrix metalloproteinase (MMP)-9. Neutrophils isolated from sPLA(2)-X(-/-) mice had lower activities of elastase, gelatinase, and MMP-9 in response to stimuli compared with sPLA(2)-X(+/+) mice. The attenuated release of elastase and gelatinase from sPLA(2)-X(-/-) neutrophils was reversed by exogenous addition of mouse sPLA(2)-X protein. The adoptive transfer of sPLA(2)-X(+/+) neutrophils days 0 and 3 after CaCl(2) treatment reversed aortic diameters and elastin degradation grades in the lethally irradiated sPLA(2)-X(+/+) mice reconstituted with sPLA(2)-X(-/-) bone marrow to an extent similar to that seen in sPLA(2)-X(+/+) mice. In conclusion, sPLA(2)-X in neutrophils plays a pathogenic role in AAA in a mice model.


Subject(s)
Aorta/enzymology , Aortic Aneurysm, Abdominal/enzymology , Group X Phospholipases A2/metabolism , Neutrophils/enzymology , Adoptive Transfer , Animals , Aorta/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/pathology , Bone Marrow Transplantation , Calcium Chloride , Disease Models, Animal , Elastin/metabolism , Gelatinases/metabolism , Group X Phospholipases A2/deficiency , Group X Phospholipases A2/genetics , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Neutrophils/transplantation , Pancreatic Elastase/metabolism , Time Factors
5.
J Immunol ; 187(1): 482-9, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21622863

ABSTRACT

Secretory phospholipase A(2)s (sPLA(2)) hydrolyze glycerophospholipids to liberate lysophospholipids and free fatty acids. Although group X (GX) sPLA(2) is recognized as the most potent mammalian sPLA(2) in vitro, its precise physiological function(s) remains unclear. We recently reported that GX sPLA(2) suppresses activation of the liver X receptor in macrophages, resulting in reduced expression of liver X receptor-responsive genes including ATP-binding cassette transporters A1 (ABCA1) and G1 (ABCG1), and a consequent decrease in cellular cholesterol efflux and increase in cellular cholesterol content (Shridas et al. 2010. Arterioscler. Thromb. Vasc. Biol. 30: 2014-2021). In this study, we provide evidence that GX sPLA(2) modulates macrophage inflammatory responses by altering cellular cholesterol homeostasis. Transgenic expression or exogenous addition of GX sPLA(2) resulted in a significantly higher induction of TNF-α, IL-6, and cyclooxygenase-2 in J774 macrophage-like cells in response to LPS. This effect required GX sPLA(2) catalytic activity, and was abolished in macrophages that lack either TLR4 or MyD88. The hypersensitivity to LPS in cells overexpressing GX sPLA(2) was reversed when cellular free cholesterol was normalized using cyclodextrin. Consistent with results from gain-of-function studies, peritoneal macrophages from GX sPLA(2)-deficient mice exhibited a significantly dampened response to LPS. Plasma concentrations of inflammatory cytokines were significantly lower in GX sPLA(2)-deficient mice compared with wild-type mice after LPS administration. Thus, GX sPLA(2) amplifies signaling through TLR4 by a mechanism that is dependent on its catalytic activity. Our data indicate this effect is mediated through alterations in plasma membrane free cholesterol and lipid raft content.


Subject(s)
Group X Phospholipases A2/physiology , Macrophages/enzymology , Macrophages/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/physiology , Animals , Cell Line , Cholesterol/metabolism , Female , Group X Phospholipases A2/deficiency , Group X Phospholipases A2/genetics , Homeostasis/genetics , Homeostasis/immunology , Lipopolysaccharides/physiology , Macrophages/pathology , Male , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics
6.
Arterioscler Thromb Vasc Biol ; 30(10): 2014-21, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20844270

ABSTRACT

OBJECTIVE: GX sPLA(2) potently hydrolyzes plasma membranes to generate lysophospholipids and free fatty acids; it has been implicated in inflammatory diseases, including atherosclerosis. To identify a novel role for group X (GX) secretory phospholipase A(2) (sPLA(2)) in modulating ATP binding casette transporter A1 (ABCA1) and ATP binding casette transporter G1 (ABCG1) expression and, therefore, macrophage cholesterol efflux. METHODS AND RESULTS: The overexpression or exogenous addition of GX sPLA(2) significantly reduced ABCA1 and ABCG1 expression in J774 macrophage-like cells, whereas GX sPLA(2) deficiency in mouse peritoneal macrophages was associated with enhanced expression. Altered ABC transporter expression led to reduced cholesterol efflux in GX sPLA(2)-overexpressing J774 cells and increased efflux in GX sPLA(2)-deficient mouse peritoneal macrophages. Gene regulation was dependent on GX sPLA(2) catalytic activity, mimicked by arachidonic acid and abrogated when liver X receptor (LXR)α/ß expression was suppressed, and partially reversed by the LXR agonist T0901317. Reporter assays indicated that GX sPLA(2) suppresses the ability of LXR to transactivate its promoters through a mechanism involving the C-terminal portion of LXR spanning the ligand-binding domain. CONCLUSIONS: GX sPLA(2) modulates gene expression in macrophages by generating lipolytic products that suppress LXR activation. GX sPLA(2) may play a previously unrecognized role in atherosclerotic lipid accumulation by negatively regulating the genes critical for cellular cholesterol efflux.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Cholesterol/metabolism , Group X Phospholipases A2/metabolism , Lipoproteins/genetics , Macrophages/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , Animals , Arachidonic Acid/pharmacology , Base Sequence , Biological Transport, Active/drug effects , Cell Line , Female , Gene Expression/drug effects , Group X Phospholipases A2/deficiency , Group X Phospholipases A2/pharmacology , Humans , Hydrocarbons, Fluorinated/pharmacology , In Vitro Techniques , Liver X Receptors , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...