Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 22(16)2021 Aug 15.
Article in English | MEDLINE | ID: mdl-34445455

ABSTRACT

Glycerol is used in many skin care products because it improves skin function. Anecdotal reports by patients on the National Psoriasis Foundation website also suggest that glycerol may be helpful for the treatment of psoriasis, although to date no experimental data confirm this idea. Glycerol entry into epidermal keratinocytes is facilitated by aquaglyceroporins like aquaporin-3 (AQP3), and its conversion to phosphatidylglycerol, a lipid messenger that promotes keratinocyte differentiation, requires the lipid-metabolizing enzyme phospholipase-D2 (PLD2). To evaluate whether glycerol inhibits inflammation and psoriasiform lesion development in the imiquimod (IMQ)-induced mouse model of psoriasis, glycerol's effect on psoriasiform skin lesions was determined in IMQ-treated wild-type and PLD2 knockout mice, with glycerol provided either in drinking water or applied topically. Psoriasis area and severity index, ear thickness and ear biopsy weight, epidermal thickness, and inflammatory markers were quantified. Topical and oral glycerol ameliorated psoriasiform lesion development in wild-type mice. Topical glycerol appeared to act as an emollient to induce beneficial effects, since even in PLD2 knockout mice topical glycerol application improved skin lesions. In contrast, the beneficial effects of oral glycerol required PLD2, with no improvement in psoriasiform lesions observed in PLD2 knockout mice. Our findings suggest that the ability of oral glycerol to improve psoriasiform lesions requires its PLD2-mediated conversion to phosphatidylglycerol, consistent with our previous report that phosphatidylglycerol itself improves psoriasiform lesions in this model. Our data also support anecdotal evidence that glycerol can ameliorate psoriasis symptoms and therefore might be a useful therapy alone or in conjunction with other treatments.


Subject(s)
Glycerol/pharmacology , Imiquimod/adverse effects , Psoriasis/drug therapy , Skin/metabolism , Animals , Aquaporin 3/genetics , Aquaporin 3/metabolism , Disease Models, Animal , Humans , Imiquimod/pharmacology , Mice , Mice, Knockout , Phospholipase D/deficiency , Phospholipase D/metabolism , Psoriasis/chemically induced , Psoriasis/genetics , Psoriasis/metabolism
2.
Int J Mol Sci ; 21(9)2020 May 01.
Article in English | MEDLINE | ID: mdl-32370031

ABSTRACT

BACKGROUND: Phospholipase (PL)D1 is crucial for integrin αIIbß3 activation of platelets in arterial thrombosis and TNF-α-mediated inflammation and TGF-ß-mediated collagen scar formation after myocardial infarction (MI) in mice. Enzymatic activity of PLD is not responsible for PLD-mediated TNF-α signaling and myocardial healing. The impact of PLD2 in ischemia reperfusion injury is unknown. METHODS: PLD2-deficient mice underwent myocardial ischemia and reperfusion (I/R). RESULTS: Enhanced integrin αIIbß3 activation of platelets resulted in elevated interleukin (IL)-6 release from endothelial cells in vitro and enhanced IL-6 plasma levels after MI in PLD2-deficient mice. This was accompanied by enhanced migration of inflammatory cells into the infarct border zone and reduced TGF-ß plasma levels after 72 h that might account for enhanced inflammation in PLD2-deficient mice. In contrast to PLD1, TNF-α signaling, infarct size and cardiac function 24 h after I/R were not altered when PLD2 was deleted. Furthermore, TGF-ß plasma levels, scar formation and heart function were comparable between PLD2-deficient and control mice 21 days post MI. CONCLUSIONS: The present study contributes to our understanding about the role of PLD isoforms and altered platelet signaling in the process of myocardial I/R injury.


Subject(s)
Blood Platelets/metabolism , Integrins/metabolism , Myocardial Infarction/complications , Myocarditis/etiology , Myocarditis/metabolism , Phospholipase D/deficiency , Animals , Biomarkers , Cell Survival , Cytokines/metabolism , Disease Susceptibility , Endothelial Cells/metabolism , Gene Expression , Integrins/chemistry , Male , Mice , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocarditis/pathology
3.
Prog Lipid Res ; 78: 101018, 2020 04.
Article in English | MEDLINE | ID: mdl-31830503

ABSTRACT

Despite being discovered over 60 years ago, the precise role of phospholipase D (PLD) is still being elucidated. PLD enzymes catalyze the hydrolysis of the phosphodiester bond of glycerophospholipids producing phosphatidic acid and the free headgroup. PLD family members are found in organisms ranging from viruses, and bacteria to plants, and mammals. They display a range of substrate specificities, are regulated by a diverse range of molecules, and have been implicated in a broad range of cellular processes including receptor signaling, cytoskeletal regulation and membrane trafficking. Recent technological advances including: the development of PLD knockout mice, isoform-specific antibodies, and specific inhibitors are finally permitting a thorough analysis of the in vivo role of mammalian PLDs. These studies are facilitating increased recognition of PLD's role in disease states including cancers and Alzheimer's disease, offering potential as a target for therapeutic intervention.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Phospholipase D/antagonists & inhibitors , Phospholipase D/metabolism , Animals , Humans , Phospholipase D/deficiency
4.
FASEB J ; 33(3): 3167-3179, 2019 03.
Article in English | MEDLINE | ID: mdl-30399323

ABSTRACT

Oleoylethanolamide (OEA), a fatty acid ethanolamide (FAE), is a lipid mediator that controls food intake and lipid metabolism. Accumulating data imply the importance of intestinal OEA in controlling satiety in addition to gastrointestinal peptide hormones. Although the biochemical pathway of FAE production has been illustrated, the enzymes responsible for the cleavage of OEA from its precursor N-acyl-phosphatidylethanolamine (NAPE) must be identified among reported candidates in the gut. In this study, we assessed the involvement of NAPE-specific phospholipase D (NAPE-PLD), which can directly release FAEs from NAPE, in intestinal OEA synthesis and lipid metabolism. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPER-associated protein 9 (Cas9)-mediated deletion of the NAPE-PLD gene in intestinal epithelial-like Caco-2 cells reduced OEA levels, regardless of their differentiation states. Transcriptome analysis revealed that deletion of NAPE-PLD activates a transcriptional program for nutrient transportation, including lipids and lipoproteins, and inactivates cell-cycle or mitosis-related genes in Caco-2 cells. In addition, the basolateral secretion of lipoproteins was increased in NAPE-PLD-deleted cells although lipoprotein size was not affected. By contrast, cellular lipid levels were reduced in NAPE-PLD-deleted cells. Overall, these results indicate that NAPE-PLD plays important roles in OEA synthesis and fat absorption by regulating lipoprotein production in the intestinal epithelial cells.-Igarashi, M., Watanabe, K., Tsuduki, T., Kimura, I., Kubota, N. NAPE-PLD controls OEA synthesis and fat absorption by regulating lipoprotein synthesis in an in vitro model of intestinal epithelial cells.


Subject(s)
Dietary Fats/metabolism , Endocannabinoids/biosynthesis , Intestinal Mucosa/metabolism , Oleic Acids/biosynthesis , Phospholipase D/metabolism , CD36 Antigens/metabolism , Caco-2 Cells , Cell Differentiation , Gene Expression Profiling , Gene Knockout Techniques , Humans , Intestinal Absorption/genetics , Intestinal Absorption/physiology , Intestinal Mucosa/cytology , Lipid Metabolism , Lipoproteins/biosynthesis , Models, Biological , Phospholipase D/deficiency , Phospholipase D/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Sci Rep ; 8(1): 10006, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29968773

ABSTRACT

Sepsis is a systemic inflammatory disorder with organ dysfunction and represents the leading cause of mortality in non-coronary intensive care units. A key player in septic shock is Tumor Necrosis Factor-alpha (TNF-α). Phospholipase (PL)D1 is involved in the regulation of TNF-α upon ischemia/reperfusion injury in mice. In this study we analyzed the impact of PLD1 in the regulation of TNF-α, inflammation and organ damage in experimental sepsis. PLD1 deficiency increased survival of mice and decreased vital organ damage after LPS injections. Decreased TNF-α plasma levels and reduced migration of leukocytes and platelets into lungs was associated with reduced apoptosis in lung and liver tissue of PLD1 deficient mice. PLD1 deficient platelets contribute to preserved outcome after LPS-induced sepsis because platelets exhibit an integrin activation defect suggesting reduced platelet activation in PLD1 deficient mice. Furthermore, reduced thrombin generation of PLD1 deficient platelets might be responsible for reduced fibrin formation in lungs suggesting reduced disseminated intravascular coagulation (DIC). The analysis of Pld1fl/fl-PF4-Cre mice revealed that migration of neutrophils and cell apoptosis in septic animals is not due to platelet-mediated processes. The present study has identified PLD1 as a regulator of innate immunity that may be a new target to modulate sepsis.


Subject(s)
Lipopolysaccharides/toxicity , Phospholipase D/metabolism , Shock, Septic/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/immunology , Blood Platelets/metabolism , Cell Movement/genetics , Cells, Cultured , Disease Models, Animal , Fibrin/metabolism , Immunity, Innate/immunology , Inflammation/pathology , Leukocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Phospholipase D/deficiency , Phospholipase D/genetics , Platelet Activation/genetics
6.
J Biochem ; 162(6): 449-458, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28992041

ABSTRACT

N-acylethanolamines (NAEs), a class of lipid mediators, are produced from N-acyl-phosphatidylethanolamine (NAPE) by several pathways, including the direct release by NAPE-specific phospholipase D (NAPE-PLD) or the multistep pathway via sn-glycero-3-phospho-N-acylethanolamine (Gp-NAE). Using liquid chromatography-tandem mass spectrometry, we compared peripheral tissue levels of NAPE, Gp-NAE and NAE in NAPE-PLD-deficient (NAPE-PLD-/-) and wild type (WT) mice. NAPE-PLD was suggested to play a major role in the NAPE degradation in heart, kidney, and liver, but not in jejunum, because the NAPE levels except jejunum were significantly higher in NAPE-PLD-/- mice than in WT mice. The deletion of NAPE-PLD failed to alter the NAE levels of these tissues, suggesting its limited role in the NAE production. The enzyme assays with tissue homogenates confirmed the presence of NAPE-PLD-independent pathways in these peripheral tissues. Gp-NAE species having an acyl moiety with 22 carbons and 6 double bonds was enriched in these peripheral tissues. As for sn-2 acyl species of NAPE, 18:2-acyl-containing NAPE species were predominant over 18:1-containing species in heart, liver, and jejunum. Our results show that both molecular species composition of NAPE, NAE and Gp-NAE and their dependencies on Napepld are different among the peripheral tissues, suggesting that each tissue has distinct metabolic pathways and these NAE-containing lipids play tissue-specific roles.


Subject(s)
Phosphatidylethanolamines/chemistry , Phospholipase D/metabolism , Animals , Brain , Ethanolamines/chemistry , Ethanolamines/metabolism , Heart , Jejunum/chemistry , Kidney/chemistry , Lipids/analysis , Liver/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Phosphatidylethanolamines/metabolism , Phospholipase D/deficiency
7.
Sci Rep ; 7(1): 9112, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28831159

ABSTRACT

The Phospholipase D (PLD) superfamily is linked to neurological disease, cancer, and fertility, and a recent report correlated a potential loss-of-function PLD2 polymorphism with hypotension. Surprisingly, PLD2 -/- mice exhibit elevated blood pressure accompanied by associated changes in cardiac performance and molecular markers, but do not have findings consistent with the metabolic syndrome. Instead, expression of endothelial nitric oxide synthase (eNOS), which generates the potent vasodilator nitric oxide (NO), is decreased. An eNOS inhibitor phenocopied PLD2 loss and had no further effect on PLD2 -/- mice, confirming the functional relationship. Using a human endothelial cell line, PLD2 loss of function was shown to lower intracellular free cholesterol, causing upregulation of HMG Co-A reductase, the rate-limiting enzyme in cholesterol synthesis. HMG Co-A reductase negatively regulates eNOS, and the PLD2-deficiency phenotype of decreased eNOS expression and activity could be rescued by cholesterol supplementation and HMG Co-A reductase inhibition. Together, these findings identify a novel pathway through which the lipid signaling enzyme PLD2 regulates blood pressure, creating implications for on-going therapeutic development of PLD small molecule inhibitors. Finally, we show that the human PLD2 polymorphism does not trigger eNOS loss, but rather creates another effect, suggesting altered functioning for the allele.


Subject(s)
Blood Pressure/genetics , Nitric Oxide Synthase Type III/metabolism , Phospholipase D/deficiency , Signal Transduction , Animals , Cholesterol/metabolism , Gene Expression , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Male , Mice , Mice, Knockout , Mutation , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/genetics , Obesity/etiology , Obesity/metabolism
8.
Cell Signal ; 38: 171-181, 2017 10.
Article in English | MEDLINE | ID: mdl-28711718

ABSTRACT

Glycoprotein (GP)Ib is not only required for stable thrombus formation but for platelet-mediated inflammatory responses. Phospholipase (PL)D1 is essential for GPIb-dependent aggregate formation under high shear conditions while nothing is known about PLD1-induced regulation of GPIb in platelet-mediated inflammation and the underlying mechanisms. This study aimed to investigate the relevance of PLD1 for platelet-mediated endothelial and leukocyte recruitment and activation in vitro and in vivo. Pld1-/- platelets showed strongly reduced adhesion to TNFα stimulated endothelial cells (ECs) under high shear conditions ex vivo. Normal cytoskeletal reorganization of Pld1-/- platelets but reduced integrin activation after adhesion to inflamed ECs confirmed that defective integrin activation is responsible for reduced platelet adhesion to ECs. This, together with significantly reduced CD40L expression on platelets led to reduced chemotactic and adhesive properties of ECs in vitro. Under flow conditions, recruitment of leukocytes to collagen-adherent platelets was reduced. Under inflammatory conditions in vivo, reduced platelet and leukocyte recruitment and arrest to the injured carotid artery was observed in Pld1-/- mice. In a second in vivo model of venous thrombosis, platelet adhesion to activated endothelial cells was reduced while leukocyte recruitment was attenuated in PLD1 deficient mice. Mechanistically, PLD1 modulates PLCγ2 phosphorylation and integrin activation via Src kinases without affecting vWF binding to GPIb. Thus, PLD1 is important for GPIb-induced inflammatory processes of platelets and might be a promising target to reduce platelet-mediated inflammation.


Subject(s)
Blood Platelets/enzymology , Blood Platelets/pathology , Inflammation/enzymology , Inflammation/pathology , Phospholipase D/metabolism , Animals , Cell Adhesion , Chemotaxis , Cytoskeleton/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Leukocytes/pathology , Mice , Phospholipase C gamma/metabolism , Phospholipase D/deficiency , Phosphorylation , Platelet Glycoprotein GPIb-IX Complex , Shear Strength , Signal Transduction , src-Family Kinases/metabolism
9.
Sci Rep ; 6: 39170, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27976696

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is characterized by the accumulation of triglycerides (TG) as lipid droplets in the liver. Although lipid-metabolizing enzymes are considered important in NAFLD, the involvement of phospholipase D1 (PLD1) has not yet been studied. Here, we show that the genetic ablation of PLD1 in mice induces NAFLD due to an autophagy defect. PLD1 expression was decreased in high-fat diet-induced NAFLD. Subsequently, PLD1 deficiency led to an increase in hepatic TGs and liver weight. Autophagic flux was blocked in Pld1-/- hepatocytes, with decreased ß-oxidation rate, reduced oxidation-related gene expression, and swollen mitochondria. The dynamics of autophagy was restored by treatment with the PLD product, phosphatidic acid (PA) or adenoviral PLD1 expression in Pld1-/- hepatocytes, confirming that lysosomal PA produced by PLD1 regulates autophagy. Notably, PLD1 expression in Pld1-/- liver significantly reduced hepatic lipid accumulation, compared with Pld1-/- liver. Thus, PLD1 plays an important role in hepatic steatosis via the regulation of autophagy.


Subject(s)
Autophagy , Phospholipase D/genetics , Animals , Autophagy/drug effects , Benzimidazoles/pharmacology , Cells, Cultured , Chromatography, High Pressure Liquid , Diet, High-Fat , Genetic Vectors/genetics , Genetic Vectors/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipid Metabolism/drug effects , Lipid Peroxidation/drug effects , Liver/metabolism , Lysosomes/metabolism , Mice , Mice, Knockout , Microscopy, Confocal , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oxygen Consumption/drug effects , Phosphatidic Acids/analysis , Phosphatidic Acids/pharmacology , Phospholipase D/deficiency , Phospholipase D/metabolism , Piperidines/pharmacology , Tandem Mass Spectrometry , Triglycerides/blood
10.
BMB Rep ; 49(3): 191-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26818087

ABSTRACT

Vascular endothelial growth factor (VEGF) is a key mediator of angiogenesis and critical for normal embryonic development and repair of pathophysiological conditions in adults. Although phospholipase D (PLD) activity has been implicated in angiogenic processes, its role in VEGF signaling during angiogenesis in mammals is unclear. Here, we found that silencing of PLD2 by siRNA blocked VEGF-mediated signaling in immortalized human umbilical vein endothelial cells (iHUVECs). Also, VEGF-induced endothelial cell survival, proliferation, migration, and tube formation were inhibited by PLD2 silencing. Furthermore, while Pld2-knockout mice exhibited normal development, loss of PLD2 inhibited VEGF-mediated ex vivo angiogenesis. These findings suggest that PLD2 functions as a key mediator in the VEGF-mediated angiogenic functions of endothelial cells. [BMB Reports 2016; 49(3): 191-196].


Subject(s)
Neovascularization, Physiologic , Phospholipase D/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Aorta/physiology , Cell Line, Transformed , Cell Movement , Cell Proliferation , Cell Survival , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Isoenzymes/metabolism , Mice, Knockout , Phospholipase D/deficiency , Signal Transduction
11.
Sci Rep ; 5: 14778, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26437780

ABSTRACT

The brain-derived neurotrophic factor BDNF plays a critical role in neuronal development and the induction of L-LTP at glutamatergic synapses in several brain regions. However, the cellular and molecular mechanisms underlying these BDNF effects have not been firmly established. Using in vitro cultures of cortical neurons from knockout mice for Pld1 and Rsk2, BDNF was observed to induce a rapid RSK2-dependent activation of PLD and to stimulate BDNF ERK1/2-CREB and mTor-S6K signalling pathways, but these effects were greatly reduced in Pld1(-/-) neurons. Furthermore, phospho-CREB did not accumulate in the nucleus, whereas overexpression of PLD1 amplified the BDNF-dependent nuclear recruitment of phospho-ERK1/2 and phospho-CREB. This BDNF retrograde signalling was prevented in cells silenced for the scaffolding protein PEA15, a protein which complexes with PLD1, ERK1/2, and RSK2 after BDNF treatment. Finally PLD1, ERK1/2, and RSK2 partially colocalized on endosomal structures, suggesting that these proteins are part of the molecular module responsible for BDNF signalling in cortical neurons.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/metabolism , Neurons/metabolism , Phospholipase D/genetics , Signal Transduction , Animals , Apoptosis Regulatory Proteins , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/pharmacology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Endosomes/metabolism , Gene Expression Regulation , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/cytology , Neurons/drug effects , Phospholipase D/deficiency , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , Primary Cell Culture , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
12.
J Immunol ; 195(9): 4492-502, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26392467

ABSTRACT

Phospholipase D (PLD) proteins are enzymes that catalyze the hydrolysis of phosphatidylcholine to generate an important signaling lipid, phosphatidic acid. Phosphatidic acid is a putative second messenger implicated in the regulation of vesicular trafficking and cytoskeletal reorganization. Previous studies using inhibitors and overexpression of PLD proteins indicate that PLD1 and PLD2 play positive roles in FcεRI-mediated signaling and mast cell function. We used mice deficient in PLD1, PLD2, or both to study the function of these enzymes in mast cells. In contrast to published studies, we found that PLD1 deficiency impaired FcεRI-mediated mast cell degranulation; however, PLD2 deficiency enhanced it. Biochemical analysis showed that PLD deficiency affected activation of the PI3K pathway and RhoA. Furthermore, our data indicated that, although PLD1 deficiency impaired F-actin disassembly, PLD2 deficiency enhanced microtubule formation. Together, our results suggested that PLD1 and PLD2, two proteins that catalyze the same enzymatic reaction, regulate different steps in mast cell degranulation.


Subject(s)
Mast Cells/immunology , Phospholipase D/immunology , Receptors, IgE/immunology , Signal Transduction/immunology , Actins/immunology , Actins/metabolism , Animals , Blotting, Western , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Cell Degranulation/immunology , Cells, Cultured , Cytoskeleton/immunology , Cytoskeleton/metabolism , Mast Cells/metabolism , Mast Cells/physiology , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase D/deficiency , Phospholipase D/genetics , Receptors, IgE/metabolism , rhoA GTP-Binding Protein/immunology , rhoA GTP-Binding Protein/metabolism
13.
Eur J Pharmacol ; 761: 398-404, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25967349

ABSTRACT

Phospholipase D (PLD) activity has been linked to proliferation in many cell types including tumor cells. In the present study, we investigated the effects of genetic deletion of PLD1 and PLD2 and of specific PLD1 and PLD2 inhibitors on PLD activity and cell proliferation in primary mouse astrocytes. Basal and stimulated PLD activity was negligible in PLD1/2 double knockouts. PLD activity was significantly reduced in PLD1-deficient cells when fetal calf serum (FCS), insulin-like growth factor 1 (IGF-1) or phorbol ester was used as a stimulant. The specificity of PLD inhibitors VU0359595 and VU0285655-1 at 500nM was confirmed in phorbol ester-stimulated cells. Significant reductions of cell proliferation were observed in PLD-deficient cell lines under basal and stimulated conditions. At 500nM, the PLD1 inhibitor VU0359595 reduced proliferation in PLD2-deficient cells, but also in PLD1-deficient cells stimulated by IGF-1 or phorbol ester. Vice versa, at 500nM, the PLD2 inhibitor VU0285655-1 reduced proliferation in PLD1-deficient cells, but also in PLD2-deficient cells exposed to IGF-1. At 5µM, both inhibitors showed non-specific effects because they inhibited cell proliferation even in PLD1/2 double knockouts. Summarizing, inhibition of PLD occurs in parallel with reduced cell proliferation in astrocytes which are deficient in PLD1 or PLD2. Synthetic PLD inhibitors show high specificity for PLD in low (nanomolar) concentrations, but have additional, non-specific effects on cell proliferation when used at high (micromolar) concentrations.


Subject(s)
Astrocytes/enzymology , Cell Proliferation , Gene Deletion , Phospholipase D/antagonists & inhibitors , Phospholipase D/metabolism , Animals , Astrocytes/drug effects , Astrocytes/pathology , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Genotype , Insulin-Like Growth Factor I/pharmacology , Mice, Knockout , Phenotype , Phorbol Esters/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phospholipase D/deficiency , Phospholipase D/genetics , Quinolines/pharmacology , Signal Transduction/drug effects , Spiro Compounds/pharmacology
14.
Nat Commun ; 6: 6495, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25757720

ABSTRACT

Obesity is a pandemic disease associated with many metabolic alterations and involves several organs and systems. The endocannabinoid system (ECS) appears to be a key regulator of energy homeostasis and metabolism. Here we show that specific deletion of the ECS synthesizing enzyme, NAPE-PLD, in adipocytes induces obesity, glucose intolerance, adipose tissue inflammation and altered lipid metabolism. We report that Napepld-deleted mice present an altered browning programme and are less responsive to cold-induced browning, highlighting the essential role of NAPE-PLD in regulating energy homeostasis and metabolism in the physiological state. Our results indicate that these alterations are mediated by a shift in gut microbiota composition that can partially transfer the phenotype to germ-free mice. Together, our findings uncover a role of adipose tissue NAPE-PLD on whole-body metabolism and provide support for targeting NAPE-PLD-derived bioactive lipids to treat obesity and related metabolic disorders.


Subject(s)
Adipose Tissue, Brown/metabolism , Gastrointestinal Microbiome/physiology , Glucose Intolerance/metabolism , Obesity/metabolism , Phospholipase D/genetics , Adipose Tissue, Brown/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Body Fat Distribution , Cold Temperature , Endocannabinoids/metabolism , Energy Metabolism/physiology , Gene Expression , Glucose Intolerance/genetics , Glucose Intolerance/microbiology , Glucose Intolerance/pathology , Inflammation , Male , Mice , Mice, Knockout , Obesity/genetics , Obesity/microbiology , Obesity/pathology , Phospholipase D/deficiency
15.
Arterioscler Thromb Vasc Biol ; 34(8): 1697-703, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24947526

ABSTRACT

OBJECTIVE: Aberrant regulation of the proliferation, survival, and migration of endothelial cells (ECs) is closely related to the abnormal angiogenesis that occurs in hypoxia-induced pathological situations, such as cancer and vascular retinopathy. Hypoxic conditions and the subsequent upregulation of hypoxia-inducible factor-1α and target genes are important for the angiogenic functions of ECs. Phospholipase D2 (PLD2) is a crucial signaling mediator that stimulates the production of the second messenger phosphatidic acid. PLD2 is involved in various cellular functions; however, its specific roles in ECs under hypoxia and in vivo angiogenesis remain unclear. In the present study, we investigated the potential roles of PLD2 in ECs under hypoxia and in hypoxia-induced pathological angiogenesis in vivo. APPROACH AND RESULTS: Pld2 knockout ECs exhibited decreased hypoxia-induced cellular responses in survival, migration, and thus vessel sprouting. Analysis of hypoxia-induced gene expression revealed that PLD2 deficiency disrupted the upregulation of hypoxia-inducible factor-1α target genes, including VEGF, PFKFB3, HMOX-1, and NTRK2. Consistent with this, PLD2 contributed to hypoxia-induced hypoxia-inducible factor-1α expression at the translational level. The roles of PLD2 in hypoxia-induced in vivo pathological angiogenesis were assessed using oxygen-induced retinopathy and tumor implantation models in endothelial-specific Pld2 knockout mice. Pld2 endothelial-specific knockout retinae showed decreased neovascular tuft formation, despite a larger avascular region. Tumor growth and tumor blood vessel formation were also reduced in Pld2 endothelial-specific knockout mice. CONCLUSIONS: Our findings demonstrate a novel role for endothelial PLD2 in the survival and migration of ECs under hypoxia via the expression of hypoxia-inducible factor-1α and in pathological retinal angiogenesis and tumor angiogenesis in vivo.


Subject(s)
Carcinoma, Lewis Lung/blood supply , Endothelial Cells/enzymology , Hypoxia/complications , Neovascularization, Pathologic , Phospholipase D/deficiency , Retinal Neovascularization/enzymology , Retinal Vessels/enzymology , Animals , Animals, Newborn , Cell Hypoxia , Cell Movement , Cell Proliferation , Cell Survival , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phospholipase D/genetics , RNA Interference , Retinal Neovascularization/etiology , Retinal Neovascularization/genetics , Retinal Neovascularization/pathology , Retinal Vessels/pathology , Time Factors , Tissue Culture Techniques , Transfection
16.
Nature ; 505(7484): 550-554, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24336208

ABSTRACT

Genome-wide association studies (GWAS) have identified several risk variants for late-onset Alzheimer's disease (LOAD). These common variants have replicable but small effects on LOAD risk and generally do not have obvious functional effects. Low-frequency coding variants, not detected by GWAS, are predicted to include functional variants with larger effects on risk. To identify low-frequency coding variants with large effects on LOAD risk, we carried out whole-exome sequencing (WES) in 14 large LOAD families and follow-up analyses of the candidate variants in several large LOAD case-control data sets. A rare variant in PLD3 (phospholipase D3; Val232Met) segregated with disease status in two independent families and doubled risk for Alzheimer's disease in seven independent case-control series with a total of more than 11,000 cases and controls of European descent. Gene-based burden analyses in 4,387 cases and controls of European descent and 302 African American cases and controls, with complete sequence data for PLD3, reveal that several variants in this gene increase risk for Alzheimer's disease in both populations. PLD3 is highly expressed in brain regions that are vulnerable to Alzheimer's disease pathology, including hippocampus and cortex, and is expressed at significantly lower levels in neurons from Alzheimer's disease brains compared to control brains. Overexpression of PLD3 leads to a significant decrease in intracellular amyloid-ß precursor protein (APP) and extracellular Aß42 and Aß40 (the 42- and 40-residue isoforms of the amyloid-ß peptide), and knockdown of PLD3 leads to a significant increase in extracellular Aß42 and Aß40. Together, our genetic and functional data indicate that carriers of PLD3 coding variants have a twofold increased risk for LOAD and that PLD3 influences APP processing. This study provides an example of how densely affected families may help to identify rare variants with large effects on risk for disease or other complex traits.


Subject(s)
Alzheimer Disease/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Phospholipase D/genetics , Black or African American/genetics , Age of Onset , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Case-Control Studies , Europe/ethnology , Exome/genetics , Female , Humans , Male , Peptide Fragments/metabolism , Phospholipase D/deficiency , Phospholipase D/metabolism , Protein Processing, Post-Translational/genetics , Proteolysis
17.
J Lipid Res ; 54(11): 3151-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24018423

ABSTRACT

Lipid aldehydes including isolevuglandins (IsoLGs) and 4-hydroxynonenal modify phosphatidylethanolamine (PE) to form proinflammatory and cytotoxic adducts. Therefore, cells may have evolved mechanisms to degrade and prevent accumulation of these potentially harmful compounds. To test if cells could degrade isolevuglandin-modified phosphatidylethanolamine (IsoLG-PE), we generated IsoLG-PE in human embryonic kidney 293 (HEK293) cells and human umbilical cord endothelial cells and measured its stability over time. We found that IsoLG-PE levels decreased more than 75% after 6 h, suggesting that IsoLG-PE was indeed degraded. Because N-acyl phosphatidylethanolamine-hydrolyzing phospholipase D (NAPE-PLD) has been described as a key enzyme in the hydrolysis of N-acyl phosphatidylethanoamine (NAPE) and both NAPE and IsoLG-PE have large aliphatic headgroups, we considered the possibility that this enzyme might also hydrolyze IsoLG-PE. We found that knockdown of NAPE-PLD expression using small interfering RNA (siRNA) significantly increased the persistence of IsoLG-PE in HEK293 cells. IsoLG-PE competed with NAPE for hydrolysis by recombinant mouse NAPE-PLD, with the catalytic efficiency (V(max)/K(m)) for hydrolysis of IsoLG-PE being 30% of that for hydrolysis of NAPE. LC-MS/MS analysis confirmed that recombinant NAPE-PLD hydrolyzed IsoLG-PE to IsoLG-ethanolamine. These results demonstrate that NAPE-PLD contributes to the degradation of IsoLG-PE and suggest that a major physiological role of NAPE-PLD may be to degrade aldehyde-modified PE, thereby preventing the accumulation of these harmful compounds.


Subject(s)
Aldehydes/metabolism , Phosphatidylethanolamines/metabolism , Phospholipase D/metabolism , Animals , Gene Silencing , HEK293 Cells , Humans , Hydrolysis , Mice , Phospholipase D/deficiency , Phospholipase D/genetics
18.
Arterioscler Thromb Vasc Biol ; 33(9): 2212-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23868933

ABSTRACT

OBJECTIVE: We recently showed that mice lacking the lipid signaling enzyme phospholipase (PL) D1 or both PLD isoforms (PLD1 and PLD2) were protected from pathological thrombus formation and ischemic stroke, whereas hemostasis was not impaired in these animals. We sought to assess whether pharmacological inhibition of PLD activity affects hemostasis, thrombosis, and thrombo-inflammatory brain infarction in mice. APPROACH AND RESULTS: Treatment of platelets with the reversible, small molecule PLD inhibitor, 5-fluoro-2-indolyl des-chlorohalopemide (FIPI), led to a specific blockade of PLD activity that was associated with reduced α-granule release and integrin activation. Mice that received FIPI at a dose of 3 mg/kg displayed reduced occlusive thrombus formation upon chemical injury of carotid arteries or mesenterial arterioles. Similarly, FIPI-treated mice had smaller infarct sizes and significantly better motor and neurological function 24 hours after transient middle cerebral artery occlusion. This protective effect was not associated with major intracerebral hemorrhage or prolonged tail bleeding times. CONCLUSIONS: These results provide the first evidence that pharmacological PLD inhibition might provide a safe therapeutic strategy to prevent arterial thrombosis and ischemic stroke.


Subject(s)
Blood Platelets/drug effects , Carotid Artery Diseases/prevention & control , Domperidone/analogs & derivatives , Enzyme Inhibitors/pharmacology , Fibrinolytic Agents/pharmacology , Indoles/pharmacology , Infarction, Middle Cerebral Artery/prevention & control , Phospholipase D/antagonists & inhibitors , Thrombosis/prevention & control , Animals , Blood Platelets/enzymology , Carotid Artery Diseases/blood , Carotid Artery Diseases/enzymology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/physiopathology , Disease Models, Animal , Domperidone/pharmacology , Dose-Response Relationship, Drug , Hemostasis/drug effects , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/physiopathology , Integrins/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Phospholipase D/deficiency , Phospholipase D/genetics , Recovery of Function , Thrombosis/blood , Thrombosis/enzymology , Thrombosis/genetics , Thrombosis/physiopathology , Time Factors
19.
PLoS One ; 8(1): e55325, 2013.
Article in English | MEDLINE | ID: mdl-23383154

ABSTRACT

Cell migration and phagocytosis ensue from extracellular-initiated signaling cascades that orchestrate dynamic reorganization of the actin cytoskeleton. The reorganization is mediated by effector proteins recruited to the site of activity by locally-generated lipid second messengers. Phosphatidic acid (PA), a membrane phospholipid generated by multiple enzyme families including Phospholipase D (PLD), has been proposed to function in this role. Here, we show that macrophages prepared from mice lacking either of the classical PLD isoforms PLD1 or PLD2, or wild-type macrophages whose PLD activity has been pharmacologically inhibited, display isoform-specific actin cytoskeleton abnormalities that likely underlie decreases observed in phagocytic capacity. Unexpectedly, PA continued to be detected on the phagosome in the absence of either isoform and even when all PLD activity was eliminated. However, a disorganized phagocytic cup was observed as visualized by imaging PA, F-actin, Rac1, an organizer of the F-actin network, and DOCK2, a Rac1 activator, suggesting that PLD-mediated PA production during phagocytosis is specifically critical for the integrity of the process. The abnormal F-actin reorganization additionally impacted neutrophil migration and extravasation from the vasculature into interstitial tissues. Although both PLD1 and PLD2 were important in these processes, we also observed isoform-specific functions. PLD1-driven processes in particular were observed to be critical in transmigration of macrophages exiting the vasculature during immune responses such as those seen in acute pancreatitis or irritant-induced skin vascularization.


Subject(s)
Cytoskeleton/physiology , Macrophages/immunology , Neutrophil Infiltration/immunology , Phagocytosis/immunology , Phospholipase D/deficiency , Animals , Blotting, Western , Cytoskeleton/immunology , Mice , Neuropeptides/metabolism , Pancreatitis/immunology , Phosphatidic Acids/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
20.
J Lipid Res ; 54(2): 379-85, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23213138

ABSTRACT

Functional defects in ATPase class I type 8B membrane 1 (ATP8B1 or familial intrahepatic cholestasis 1, FIC1) lead to cholestasis by mechanism(s) that are not fully understood. One proposed pathophysiology involves aberrant signaling to the bile acid sensor, the farnesoid X receptor (FXR), via protein kinase C ζ (PKCζ). The following cell line-based studies investigated whether phospholipase D2 may transduce a signal from FIC1 to FXR. PLD2 gain of function led to activation of the bile salt export pump (BSEP) promoter, a well-characterized FXR response. BSEP activation by PLD2 could be blocked by abrogating either PKCζ or FXR signaling. PLD2 loss of function led to a reduction in BSEP promoter activity. In addition, a variety of proteins that are activated by FXR, including BSEP, were reduced in HepG2 cells treated with PLD2 siRNA. Similar effects were observed in freshly isolated human hepatocytes. Activation of BSEP by FIC1 gain of function was blocked when PLD2 but not PLD1 was silenced. Overexpression of wild-type but not Byler mutant FIC1 led to an increase in membrane associated PLD activity. An intermediate level of activation of PLD activity was induced when a benign recurrent intrahepatic cholestasis FIC1 mutant construct was expressed. These studies show that FIC1 signals to FXR via a signaling pathway including PLD2 and PKCζ.


Subject(s)
Adenosine Triphosphatases/metabolism , Phospholipase D/metabolism , Signal Transduction , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/genetics , Animals , CHO Cells , Cricetinae , Cricetulus , Gene Silencing , Hep G2 Cells , Humans , Phospholipase D/deficiency , Phospholipase D/genetics , Promoter Regions, Genetic/genetics , Protein Kinase C/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL