Abstract

The canonical pathway of eicosanoid production in most mammalian cells is initiated by phospholipase A2-mediated release of arachidonic acid, followed by its enzymatic oxidation resulting in a vast array of eicosanoid products. However, recent work has demonstrated that the major phospholipase in mitochondria, iPLA2γ (patatin-like phospholipase domain containing 8 (PNPLA8)), possesses sn-1 specificity, with polyunsaturated fatty acids at the sn-2 position generating polyunsaturated sn-2-acyl lysophospholipids. Through strategic chemical derivatization, chiral chromatographic separation, and multistage tandem MS, here we first demonstrate that human platelet-type 12-lipoxygenase (12-LOX) can directly catalyze the regioselective and stereospecific oxidation of 2-arachidonoyl-lysophosphatidylcholine (2-AA-LPC) and 2-arachidonoyl-lysophosphatidylethanolamine (2-AA-LPE). Next, we identified these two eicosanoid-lysophospholipids in murine myocardium and in isolated platelets. Moreover, we observed robust increases in 2-AA-LPC, 2-AA-LPE, and their downstream 12-LOX oxidation products, 12(S)-HETE-LPC and 12(S)-HETE-LPE, in calcium ionophore (A23187)-stimulated murine platelets. Mechanistically, genetic ablation of iPLA2γ markedly decreased the calcium-stimulated production of 2-AA-LPC, 2-AA-LPE, and 12-HETE-lysophospholipids in mouse platelets. Importantly, a potent and selective 12-LOX inhibitor, ML355, significantly inhibited the production of 12-HETE-LPC and 12-HETE-LPE in activated platelets. Furthermore, we found that aging is accompanied by significant changes in 12-HETE-LPC in murine serum that were also markedly attenuated by iPLA2γ genetic ablation. Collectively, these results identify previously unknown iPLA2γ-initiated signaling pathways mediated by direct 12-LOX oxidation of 2-AA-LPC and 2-AA-LPE. This oxidation generates previously unrecognized eicosanoid-lysophospholipids that may serve as biomarkers for age-related diseases and could potentially be used as targets in therapeutic interventions.

Highlights

  • We found that aging is accompanied by significant changes in 12(S)-hydroxy-eicosatetraenoic acid (12-HETE)-LPC in murine serum that were markedly attenuated by iPLA2␥ genetic ablation

  • Because calcium activates iPLA2␥ to produce 2-arachidonic acid (AA)-lysophospholipids from arachidonate-containing phospholipids [37], these results indicate that the production of 2–12–HETElysophospholipids in platelets is initiated by iPLA2␥–mediated PLA1 hydrolysis of AA-phospholipids to produce 2-AA-lysophospholipids that can be directly oxidized by 12-LOX to generate 2–12–HETE-lysophospholipids (Fig. 7)

  • We demonstrated that calcium ion induces the activation of iPLA2␥ leading to the production of 2-AA-LPC and 2-AA-LPE [37]

Read more

Summary

The abbreviations used are

12-LOX, 12-lipoxygenase; iPLA2␥, calcium-independent phospholipase A2␥; cPLA2␣, cytosolic phospholipase A2␣; PPH3, triphenylphosphine; AA-LPC, arachidonoyl-lysophosphatidylcholine; AALPE, arachidonoyl-lysophosphatidylethanolamine; 12-HETE, 12-hydroxy-eicosatetraenoic acid; 12-HpETE, 12-hydroperoxy-eicosatetraenoic acid; ML355, N-2-benzothiaoly-4-[[(2-hydroxy-3-methoxyphenyl)methyl]amino]benzenesulfonamide; HRAM, high resolution high mass accuracy; CID, collision-induced dissociation; Ni-NTA, nickel-nitrilotriacetic acid; AA, arachidonic acid. The crystal structure of the 12-LOX catalytic domain possesses a U-shaped substrate-binding site that facilitates the precise positioning of the C10 hydrogens in arachidonic acid for the regioselective and stereospecific oxidization of AA resulting in the production of 12(S)-HpETE [32]. The regiospecificity of iPLA2␥ for the sn-1 position of polyunsaturated phospholipids suggested the existence of a previously unknown signaling pathway initiated by the generation of 2-arachidonoyl-lysophosphatidylcholine, which serves as a signaling node for multiple lipid 2nd messenger cascades [37, 38]. We demonstrated that aging in mice is accompanied by increased levels of 12-HETE-LPC in serum that are attenuated by genetic ablation of iPLA2␥ These results identify previously unknown iPLA2␥-initiated signaling pathways mediated by direct 12-LOX oxidation of 2-arachidonoyl-lysophospholipids to generate previously unrecognized eicosanoid-lysophospholipid metabolites in critical cells of the cardiovascular system that could potentially serve as biomarkers of disease and inflammation during aging

Results
Discussion
Findings
Experimental procedures
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call