Movatterモバイル変換


[0]ホーム

URL:


Skip to main content
                                  NCBI home page
Search in PMCSearch
As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsement of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more:PMC Disclaimer | PMC Copyright Notice
Journal of Lipid Research logo

Calcium-independent phospholipases A2 and their roles in biological processes and diseases

Sasanka Ramanadham*,†,1,Tomader Ali§,Jason W Ashley**,Robert N Bone*,,William D Hancock*,,Xiaoyong Lei*,
*Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
§Undergraduate Research Office, University of Alabama at Birmingham, Birmingham, AL 35294
**Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104
1

To whom correspondence should be addressed. e-mail:sramvem@uab.edu

Series information

Thematic Review Series: Phospholipases: Central Role in Lipid Signaling and Disease

Received 2015 Feb 18; Revised 2015 May 28; Issue date 2015 Sep.

Copyright © 2015 by the American Society for Biochemistry and Molecular Biology, Inc.
PMCID: PMC4548770  PMID:26023050

Abstract

Among the family of phospholipases A2 (PLA2s) are the Ca2+-independent PLA2s (iPLA2s) and they are designated group VI iPLA2s. In relation to secretory and cytosolic PLA2s, the iPLA2s are more recently described and details of their expression and roles in biological functions are rapidly emerging. The iPLA2s or patatin-like phospholipases (PNPLAs) are intracellular enzymes that do not require Ca2+ for activity, and contain lipase (GXSXG) and nucleotide-binding (GXGXXG) consensus sequences. Though nine PNPLAs have been recognized, PNPLA8 (membrane-associated iPLA2γ) and PNPLA9 (cytosol-associated iPLA2β) are the most widely studied and understood. The iPLA2s manifest a variety of activities in addition to phospholipase, are ubiquitously expressed, and participate in a multitude of biological processes, including fat catabolism, cell differentiation, maintenance of mitochondrial integrity, phospholipid remodeling, cell proliferation, signal transduction, and cell death. As might be expected, increased or decreased expression of iPLA2s can have profound effects on the metabolic state, CNS function, cardiovascular performance, and cell survival; therefore, dysregulation of iPLA2s can be a critical factor in the development of many diseases. This review is aimed at providing a general framework of the current understanding of the iPLA2s and discussion of the potential mechanisms of action of the iPLA2s and related involved lipid mediators.

Keywords: signaling, membrane homeostasis, arachidonic acid, eicosanoids, docosahexaenoic acid, inflammation, immune responses, alternate splicing, β-cells, central nervous system disorders, cancers

OVERVIEW OF THE GROUP VIA Ca2+-INDEPENDENT PHOSPHOLIPASES A2

The Ca2+-independent phospholipases A2 (iPLA2s) are part of a diverse family of PLA2s that hydrolyze thesn-2 substituent from membrane phospholipids to release a free fatty acid and a lysolipid (1,2). These enzymes are ubiquitously expressed, and in contrast to secretory PLA2s (sPLA2s) and cytosolic PLA2s (cPLA2s), do not require Ca2+ for either translocation or activity. Some of the first descriptions of iPLA2 activity were in the mid- to late-1980s with the identification of a plasmalogen-selective PLA2 in the cytosol of canine myocardium (3) that migrated with a molecular mass of 40 kDa. Analogous activity was subsequently described in insulinoma cells (4) and renal proximal tubules (5), as well as in a macrophage cell line (6). The Ca2+-independent PLA2s are designated as group VI iPLA2s (7,8) and now include seven members, as described inTable 1: iPLA2β (VIA-1 and -2), iPLA2γ (VIB), iPLA2δ (VIC), iPLA2ε (VID), iPLA2ζ (VIE), and iPLA2η (VIF). Three others (iPLA2φ, iPLA2ι, and iPLA2κ) have been recognized, but very little is known about them and they are not yet assigned to group VI (9,10). Due to their shared homology with patatin, the iPLA2s are included in the patatin-like protein family and are also referred to as PNPLAs. The iPLA2s also share a consensus GXSXG catalytic motif contained within a patatin-like lipase domain. This review discusses the current understanding of the various iPLA2s, starting with the less-described iPLA2δ, iPLA2ε, iPLA2ζ, and iPLA2η, followed by emerging reports relating to iPLA2γ, and ending with the most widely examined, iPLA2β.

TABLE 1.

Listing of the group VI family of iPLA2s

Group VI PLA2sDescribedOther NamesChromosomeAmino AcidskDaAnk RepeatsLocalizationActive Site
VIA-1, iPLA2β1994 (6)PNPLA922q13.1752858CytosolS465
VIA-2, iPLA2β1999 (96)PNPLA922q13.1804887CytosolS519
VIB, iPLA2γ2000 (61,62)PNPLA87q31782900MembraneS483
VIC, iPLA2δ2002 (11)PNPLA6, NTE19p13.3-13.21,3661460Neurons (ER, Golgi)S1005
VID, iPLA2ε2001 (12)PNPLA3, adiponutrin22q13.31481520Liver adipocytesS47
VIE, iPLA2ζ2004 (27,43,44)PNPLA2, desnutrin, ATGL11p15.5504550White and brown adipocytes (lipid droplets)S47
VIF, iPLA2η1994 (60)PNPLA4, gene sequence-2 (GS2)xp22.3253270ubiquitousS43

iPLA2δ

The group VIC iPLA2δ (PNPLA6), also known as neuropathy target esterase (NTE), was recognized for manifesting iPLA2 and lysophospholipase activities in 2002 (11). The gene for iPLA2δ is located at chromosome 19p13.3-13.2, and encodes a protein containing 1,366 amino acids with a molecular mass of 146 kDa and an active site at S1005. Expressed predominantly in neurons, iPLA2δ localizes to the endoplasmic reticulum (ER) and Golgi apparatus (12), and its inhibition or deletion leads to axonal degeneration. It is in this context that NTE was discovered during studies for causes of organophosphorus ester-induced paralysis in the late 1960s (13,14). Whereas conventional KO of NTE is embryonic lethal (15), conditional KO of NTE in the CNS leads to neurodegeneration (16,17), suggesting loss of function as a causative factor in the development of neurological diseases. However, mutations in the catalytic site of NTE lead to hereditary spastic paraplegia (18,19), a symptom of NTE-motor neuron disorder. Clinical manifestations of NTE-motor neuron disorder are only evident when mutations are carried by both alleles, suggesting that the neurodegeneration results from production of abnormal NTE, rather than due to reduction in NTE activity (20,21). Among the syndromes associated with mutations in PNPLA6 are: Gordon Holmes (22,23) and Boucher-Neuhauser (23), characterized by early-onset ataxia and hypogonadism; Oliver-McFarlane (24), characterized by trichomegaly, congenital hypopituitarism, retinal degeneration, and choroidal atrophy; Laurence-Moon (24), characterized by progressive spinocerebellar ataxia and spastic paraplegia; and photoreceptor degeneration and childhood blindness (25). An NTE-related iPLA2 (PNPLA7) awaits further characterization (9,10).

iPLA2ε

The group VID iPLA2ε (PNPLA3), also referred to as adiponutrin, was described in 2001 (12). The gene for iPLA2ε is located at chromosome 22q13.31 and encodes a protein containing 481 amino acids with a molecular mass of 52 kDa and an active site at S47. PNPLA3 is mainly expressed in intracellular membrane fractions in hepatocytes (26) and was originally described as a nutritionally-regulated adipose-specific transcript in 3T3-L1 adipocytes (12). In addition to phospholipase activity, iPLA2ε manifests TG lipase and acylglycerol transacylase activities (27), leading to the suggestion that it facilitates energy/lipid mobilization and storage in adipocytes. In this regard, iPLA2ε correlates highly with the development and progression of nonalcoholic fatty liver disease and has been identified as a genetic determinant of liver fibrosis (28,29). Whereas the WTPNPLA3 exhibits lipolytic activity toward TGs, the rs738409 variantPNPLA3, where isoleucine148 is replaced by methionine (L148M), reduces the access of substrates and activity of PNPLA3 toward glycerolipids. This leads to development of macrovesicular steatosis (26,30), simple steatosis to steatohepatitis and progressive cirrhosis (31), and hepatic fibrinogenesis by a sterol regulatory element-binding protein (SREBP)-1c-PNPLA3 pathway (32). A greater impact of the L148M variant on hepatic lipid content is unmasked in the presence of other risk factors such as obesity (33), visceral adiposity (34), increased intake of sugars (35), omega-6 PUFAs (36), glucokinase regulatory protein gene variant (37), chronic hepatitis B (38), and hepatocellular carcinoma (39). In contrast to these reports, PNPLA3 has been reported to restore lipid homeostasis (40) by mediating acylation of lysophospholipids and hydrolyzing TGs in the liver in a direct manner or by regulation by cofactors (41,42).

iPLA2ζ

The group VIE iPLA2ζ (PNPLA2), also known as TST2.2, desnutrin, and adipose TG lipase (ATGL), was described in 2004 (27,43,44). The gene for iPLA2ζ is located at chromosome 11p15.5 and encodes a protein containing 504 amino acids with a molecular mass of 55 kDa and an active site at S47. Similar to iPLA2ε, iPLA2ζ also exhibits TG lipase and acylglycerol transacylase activities (27). For optimal activity, ATGL requires the cofactor comparative gene identification-58 (CGI-58), which amplifies the hydrolase activity 20-fold (45). Mutations in CGI-58, as in Chanarin-Dorfman syndrome (46), lead to TGs in various tissues and decreases in both CGI-58 and ATGL have been reported to exacerbate myocardial steatosis and oxidative stress to promote cardiac apoptosis in a rodent T2D model (47). Analogously, ATGL deficiency in mice promotes tissue accumulation of lipids and leads to premature death due to cardiomyopathy, as a consequence of reductions in fatty acid oxidative gene expression, mitochondrial fatty acid oxidation, and reduced oxygen consumption (48). Macrophages with ATGL deficiency are more susceptible to ceramide-mediated mitochondrial dysfunction and programmed cell death (49). β-Cell-specific ATGL-deficiency has been demonstrated to lead to hyperglycemia due to impaired insulin secretion, as a consequence of increased islet TG content with lower fatty acid levels. These mice also have decreased expression of PPARδ genes that encode enzymes required in mitochondrial oxidation, and this is reflected by impaired mitochondrial respiration and ATP production needed for glucose-stimulated insulin secretion (50). While polymorphisms inPNPLA2 are reported to highly correlate with T2D (51), the contribution of ATGL to insulin secretion and signaling has been challenged (52,53). In addition to its links to CGI-58 and PPARδ, ATGL has been reported to interact with TNFα in adipocytes (54), estrogen receptor α (ERα) in bone marrow (55), fat-specific protein 27 (FSP27) in human adipocytes (56), sirtuin 1 (SIRT1) during β-adrenergic signaling (57), hepatic PPARα (58), AMPK during thermogenesis (59), and to be a candidate for transcriptional control by PPARγ-mediated signals (54).

iPLA2η

The group VIF iPLA2η (PNPLA4), also known as gene sequence-2 (GS2), was described in 1994 (60). The gene for iPLA2η is located at xp22.3 and encodes a protein containing 253 amino acids with a molecular mass of 27 kDa and an active site at S43. Similar to iPLA2ε and iPLA2ζ, iPLA2η exhibits TG lipase and acylglycerol transacylase activities (27). Though expression of iPLA2η in a variety of tissues (liver, brain, skeletal muscle, lung, placenta, kidney, and pancreas) was identified in 1994, and more recently in adipose tissue (27), to date, very little is known about its biology or its role in metabolic diseases. Similar to iPLA2ε and iPLA2ζ, iPLA2η activation is proposed to contribute to regulation of anabolic and catabolic fluxes of acyl equivalents in tissues. It has been suggested that the TG lipase activity of iPLA2ε, iPLA2ζ, and iPLA2η play roles in serum fatty acid accumulations associated with metabolic syndrome and T2D. A related GS2-like iPLA2 (PNPLA5) has yet to be characterized (9,10).

iPLA2γ

The group VIB iPLA2γ (PNPLA8) genomic organization and mRNA sequence were first described in a variety of tissues (skeletal muscle, heart, placenta, brain, liver, and pancreas) in 2000 (61) and later in the same year in lymphocytes (62). The gene for iPLA2γ is located at 7q31 and encodes a protein containing 782 amino acids with a molecular mass of 90 kDa and an active site at S483. Recognition of the similarity in the catalytic domain between human iPLA2γ, cPLA2, and plant PLA2 patatin and conservation of sequence surrounding Asp627, and noting that substitution of alanine for either Ser483 of Asp627 caused loss of iPLA2γ activity, led to the suggestion that the Ser-Asp dyad constitutes the active site in human iPLA2γ (63). Initially recognized as membrane associated (61,62), dual-competing subcellular localization signals have been identified in discrete isoforms of iPLA2γ (64) that promote its accumulation and expression of activity in the peroxisomes and mitochondria (65), leading to the suggestion that iPLA2γ plays a role in integration of lipid and energy metabolism. Further, iPLA2 activity in the ER of rabbit and rat kidney (66) and ventricular myocyte membranes (67) has been demonstrated to be due to iPLA2γ.

The iPLA2γ protein contains four methionine residues that can act as potential translational initiation sites (60,63) to generate the full-length (∼88 kDa) and three truncated products (77, 74, and 63 kDa). Attempts at expression of the truncated products in HEK293 cells, however, led to the predominant expression of the 63 kDa product (68), the isoform reported earlier to be expressed in peroxisomes (64). Further examination of parental cells revealed that the 63 kDa isoform was much more abundant than the full-length iPLA2γ in HEK293 and human colorectal cancer cell lines, HCA-7 and WiDr, while in human bronchial epithelial (BEAS-2B) and rat fibroblastic (3YI) cells, the full-length iPLA2γ was the predominant isoform (68). These authors suggested that iPLA2γ potentiates arachidonic acid (AA) release from various subclasses of phosphatidylethanolamine (PE) and phosphatidylcholine (PC) to increase prostaglandin E2 (PGE2) production via cyclooxygenase (COX)-1 and -2, and this contributes to cell growth and tumorigenesis. In contrast, comparative substrate preference studies revealed that unlike cPLA2, which generates predominantly 1-palmitoyl lysophosphatidylcholine (LPC) and AA from 1-palmitoyl-2-arachidonoyl-sn-phosphatidylcholine hydrolysis, and iPLA2β, which exhibits mixed PLA1/PLA2 activities and generates 1-palmitoyl LPC at an initial 3-fold rate greater than 2-arachidonoyl LPC, iPLA2γ overexpressed in and purified from Sf9 cells hydrolyzed saturated and monounsaturated fatty acids at equal rates from thesn-1 orsn-2 position in diacyl PC substrates. However, it was less effective in releasing PUFAs from thesn-2 position, as reflected by generation of 2-arachidonoyl LPC at a 10-fold faster rate than 1-palmitoyl LPC (69).

Understanding the role of iPLA2γ-derived lipid signals has substantially advanced following the generation of mice with tissue-specific overexpression or global KO of iPLA2γ. Cardiac-specific overexpression of iPLA2γ presented multiple phenotypes that included reductions in myocardial phospholipid mass in fasted and fed states, accu­mu­lation of TGs with caloric restriction, acute fasting-induced hemodynamic dysfunction (that was accompanied by loosely packed and disorganized mitochondrial cristae), and elevated levels of 2-arachidonoyl-sn-glycero-3-phosphocholine and 2-docosahexaenoyl-sn-glycero-3-phosphocholine (65). These findings were associated with increased expression not of the full-length, but of the 70 and 63 kDa iPLA2γ isoforms. Impairment in mitochondrial function is also evidenced in iPLA2γ-null mice, which exhibit growth retardation, cold intolerance, and increased mortality due to aortic stress that were associated with decreased myocardial function and O2 consumption (70). The iPLA2γ-null mice also become resistant to Western diet-induced increases in body weight, adiposity, circulating levels of cholesterol, glucose, and insulin; and insulin resistance, and glucose intolerance (71). Ability to utilize fat and carbohydrates is also affected in these mice in association with severe impairment in skeletal muscle mitochondrial oxidation of fatty acids. Subsequent studies revealed that marked decreases in cardiolipin molecular species containing 22:6 were an underlying cause for the mitochondrial uncoupling evident in the iPLA2γ-null mice (72). Similarly, hippocampal phospholipid metabolism was found to be severely compromised with iPLA2γ-deficiency leading to a mitochondrial neurodegenerative disorder characterized by degenerating mitochondria, autophagy, and cognitive dysfunction, that was associated with alterations in the compositions and content of PCs, PEs, oxidized PEs, and ceramides and a shift in cardiolipins to shorter chain molecular species (73). Increased lipid peroxidation was also evident in the skeletal muscles of iPLA2γ-null mice, which exhibited abnormal mitochondrial function, oxidative stress, growth retardation, and loss of skeletal muscle structure and function (74). These findings are consistent with a previous report of the protective effects of iPLA2γ, deduced using a selective inhibitor of iPLA2γ against oxidant-induced lipid peroxidation and necrosis of renal proximal tubular cells (75). In contrast to the earlier report in the heart (65), the full-length iPLA2γ was the major isoform detected in the last two studies (74,75).

The identification of iPLA2γ in the mitochondria and ER paved the way for studies that demonstrated a protective role for iPLA2γ against cell death. Utilizing knockdown protocols, global iPLA2γ-null mouse model, or by selective inhibition with bromoenol lactone (BEL) (suicide substrate) of iPLA2γ (R-BEL) versus iPLA2β (S-BEL), several studies reported protective effects of iPLA2γ against oxidant- and cytokine-induced cell death. Human astrocytes exposed to hydrogen peroxide or tert-butyl hydroperoxide exhibited cell death, and pretreatment withS-BEL, but notR-BEL, amplified loss of ATP levels and cell necrosis (76). Similarly, knockdown of iPLA2γ in renal proximal tubular cells resulted in increased susceptibility to oxidant-induced cell death, elevations in lipid peroxidation, and uncoupled oxygen consumption (77,78). Analogous findings were reported in INS-1 insulinoma cells, where knockdown of iPLA2γ promoted increases in cytokine- and oxidant-induced membrane peroxidation and apoptosis (79). Cytoprotective effects of iPLA2γ were also demonstrated in glomerular epithelial cells, and these were attributed to iPLA2γ-mediated upregulation of ER chaperones (80). A general conclusion derived from these studies is that the lack of iPLA2γ decreases substrate availability for reacylation, leading to increases in lipid peroxidation. In apparent contrast to these reports, genetic ablation of iPLA2γ or its inhibition withR-BEL attenuated calcium-, reactive oxygen species (ROS)-, or oxidized lipid-mediated increases in liver mitochondrial swelling, mitochondrial permeability transition pore opening, and cytochromec release from mitochondria, which trigger the intrinsic apoptotic pathway (81,82).

The more recent description of iPLA2γ, to date, has limited wide studies of its role in clinical diseases, but a few reports suggest a role for iPLA2γ in certain clinical-related disorders. Chagas’ disease is caused by protozoan parasiteTrypanosoma cruzi, which infects cardiac myocytes promoting release of inflammatory mediators such as eicosanoids. Inhibition of iPLA2γ attenuated AA and PGE2 release and platelet-activating factor (PAF) production from HL-1 cardiac myocytes infected withT. cruzi (83), and these effects were alleviated by pretreatment withR-BEL. Consistent with a protective role of iPLA2γ in this process, the survival rates were lowered and tissue parasitism amplified inT. cruzi-infected iPLA2γ-null mice, suggesting that iPLA2γ activity affords protection against acute state cardiomyopathy in Chagas’ disease (83). In contrast, iPLA2γ-deficiency has been shown to increase bleeding time and provide resistance to thromboembolism (84), raising the possibility of targeting iPLA2γ for antithrombotic drug development. To date, the only reported clinical manifestation relating to iPLA2γ is a report that its absence is associated with myocardial dysfunction, cognitive defects, and mitochondrial degeneration (85) in a case study that closely parallels the phenotype in iPLA2γ-null mice (73).

iPLA2β

The group VIA iPLA2β (PNPLA9) is the most widely described of the iPLA2s and expression of its activity was first described in P388D1 macrophage-like cells in 1994 (6) and later shown to be the same enzyme (86) as that cloned from Chinese hamster ovary cells in 1997 (8688). Unlike cPLA2, which exhibits preference for hydrolysis of AA from thesn-2 position (89), the iPLA2s demonstrate no substrate specificity and manifest PLA2/PLA1, lysophospholipase (90,91), transacylase (27,91), and thioesterase (92,93) activities. The extensively studied iPLA2β was cloned from hamster (87), mouse (86), and rat (94), and they represent species homologs that are 85 kDa proteins (752 amino acids) with a serine lipase consensus sequence (GTSGT), preceded by eight N-terminal ankyrin (Ank) repeats (87,94).

A homologous 88 kDa iPLA2β was cloned from human lymphocyte lines and testis (95) that contains a 54-amino acid insert interrupting the eighth Ank repeat. Subsequent analyses with human pancreatic islet mRNA identified cDNA species that encoded two distinct 85 kDa (VIA-1) and 88 kDa (VIA-2) human iPLA2β isoforms (96). Analogous transcripts were also identified in human promonocytic U937 cells. The human iPLA2β gene resides on chromosome 22 in region q13.1 and contains 16 exons in the VIA-2 transcript. Exon 8 is not present in the VIA-1 transcript, indicating that it arises by an exon-skipping mechanism of alternative splicing.

Additional alternate splicing events generate iPLA2β variants that differ in their subcellular localization, catalytic activity, and likely cellular function (95). Splice variants (Ank-1 and Ank-2) encode premature stop codons due to alternatively spliced exon 10a. The proteins encoded by these splice variants, VIA Ank-1 (53 kDa, ∼479 amino acids) and VIA Ank-2 (47 kDa, ∼427 amino acids), terminate after the seventh Ank repeat domain and before the active site, whereas VIA-3 (∼70 kDa, 640 amino acids) terminates just after the lipase active site. Two additional active iPLA2β isoforms have been recognized to arise from proteolytic cleavage: a ∼63 kDa isoform (VIA-4, 623 amino acids) arising from caspase-3-catalyzed cleavage at the N terminal (97,98) and a ∼70 kDa (VIA-5, ∼640 amino acids) isoform arising from C-terminal cleavage (99). Proteomic analyses by mass spectrometry further reveal that the iPLA2β is a candidate for numerous truncations at the N-terminal end (100,101), but the activities and biological roles manifested by these products have not yet been discerned.

Basic characteristics of iPLA2β

In addition to the Ank repeats, the iPLA2β protein contains an ATP binding consensus motif (GGGVKG), an N-terminal caspase-3 cleavage site (DVTD), and a putative bipartite nuclear localization sequence (KREFGEHTKMTDVKKPK). Though under basal conditions iPLA2β is predominantly localized in the cytosol (102), translocation of iPLA2β to the Golgi, ER, mitochondria, and nucleus is evident under stimulatory conditions (98,100,103110). The iPLA2β and iPLA2γ share signature ATP binding motif, serine lipase site, and a region of 9 amino acids (627-635 in iPLA2γ), whose biological significance is not known, but otherwise lack any additional homology (61,87).

Modulation of iPLA2β

Oligomerization.

A unique distinction between iPLA2β and other PLA2s is the presence of a variable number of Ank repeats in iPLA2β, which are absent in other PLA2s. Several lines of study suggest that the Ank regions confer iPLA2β protein activity. The active form of iPLA2β appears to be an oligomer of interacting protein subunits, as supported by radiation inactivation and gel filtration chromatography analyses that reveal association of the 85 kDa iPLA2β activity with an apparent molecular mass of 250–350 kDa (6,87,111). This has led to the speculation that the active form of iPLA2β is an oligomer of 85 kDa subunits and that the subunits associate with each other via their Ank repeat regions (87), similar to the involvement of Ank repeats in other protein-protein interactions (112). Consistent with this possibility are the observations that iPLA2β deletion mutants lacking the Ank repeat domain, but retaining the catalytic domain, are catalytically inactive (87) and that activity of the full-length protein is reduced when it is coexpressed with truncated iPLA2β-like proteins that retain the Ank repeat domain, but lack the catalytic domain (95). In the long isoform of human iPLA2, a proline-rich insert interrupts the last iPLA2β Ank repeat with some similarities to the Smad4 domain that mediates interactions with signaling partners (113). This raises the possibility that the proline-rich insert in human iPLA2β allows it to interact with proteins not recognized by the short isoform of iPLA2β.

Oxidation.

It has been suggested that iPLA2β inactivation can occur by a mechanism involving oxidation of sulfhydryl groups within the iPLA2β (78). Subsequently, oligomerization of iPLA2β in INS-1 cells in response to oxidative stress was demonstrated (114). Oxidant-induced oligomerization alters the subcellular localization of iPLA2β and results in reduced release of AA, suggesting inhibition of iPLA2β catalytic activity. These nonproductive oligomers are DTT-sensitive and therefore likely generated through intermolecular disulfide bonds. Like iPLA2β, iPLA2γ activity is suppressed by oxidants, but restored when oxidant-inhibited enzyme is treated with a reducing agent (78,115).

Together, these studies indicate that iPLA2β monomers are capable of assembling into both productive and nonproductive oligomers. The productive oligomerization is mediated through the N-terminal Ank repeat domain, while inactive oligomers are formed through intramolecular disulfide bonds.

Activation.

The iPLA2β protein contains a consensus nucleotide-binding motif (GGGVKG) that is homologous to those of protein kinases (61,116). This feature mediates regulation and stabilization of iPLA2β activity by ATP (6,91,117,118), which is independent of enzyme phosphorylation (6,117,118). Also contained in iPLA2β are a C-terminal 1-9-14 calmodulin-binding motif (IRKGQGNKVKK LSI) and a calmodulin-binding peptide (AWSEMVGIQYFR) (106,116,119). These facilitate formation of a signaling complex between iPLA2β and CaMKIIβ and enhancement of both activities is evident upon their association (120). This has been offered as one explanation of why Ca2+ store depletion activates iPLA2 (121) and may occur in vascular myocytes (90), pancreatic islet β-cells (122), and human granulocytes (123). Ca2+ store depletion also activates hydrolysis of arachidonate from phospholipids in differentiated human U937 promonocytic cells by a mechanism that does not require a rise in cytosolic [Ca2+] (124). The iPLA2β protein also contains a consensus sequence site for caspase-3-mediated cleavage within the first Ank repeat (125). The truncated product manifests higher activity (125) and localizes to the nucleus under high glucose stimulation and prolonged stress (98,107), suggesting that it may amplify hydrolysis of nuclear membrane phospholipids and lead to nuclear membrane lysis, or that the iPLA2β and/or iPLA2β-derived products accumulate in an environment that can potentially participate in transcriptional induction of favorable and nonfavorable genes.

Gene induction.

The iPLA2β gene contains a sterol regulatory element (SRE) (126). Under stressful conditions SREBPs are processed to mature forms of SREBPs (127135), which translocate to the nucleus and bind to SRE. This leads to induction of iPLA2β transcription and protein expression, which are suppressed in the presence of a dominant negative form of SREBP-1 (136-138). Intriguingly, the iPLA2β gene exhibits remarkable cross-species homology in the promoter region, which contains putative consensus sequences for a number of stress-related transcriptional factors, suggesting that the iPLA2β gene is a candidate for modulation during periods of stress. Confirmation of iPLA2β induction by these stress-related transcriptional factors will lead to a better understanding of the role of iPLA2β in stress responses and disease manifestation.

iPLA2β inactivation.

Inhibitors of iPLA2β include arachidonyl trifluoromethyl ketone (AACOCF3), methyl arachidonyl fluorophosphonate, and palmitoyl trifluoromethyl ketone (PACOCF3), which are sometimes used for “selective” inhibition of cPLA2 (139141). While siRNAs directed at iPLA2β and, now available, iPLA2β-KO and iPLA2β-Tg mice (142144) have provided insight into biological processes impacted by iPLA2β, the majority of studies to assess the role of the iPLA2β isoform have utilized a selective inhibitor of iPLA2 (145). This inhibitor, (E)-6-(bromo-methylene) tetrahydro-3-(1-naphthalenyl)-2H-pyran-2-one, was synthesized in 1991 and was originally designated as a haloenol lactone substrate (146), but is now referred to as BEL. The BEL compound is an irreversible inhibitor that selectively targets iPLA2 enzymes and has little or no effect on cPLA2 or sPLA2 activity at concentrations that inhibit iPLA2β or iPLA2γ (27,146,147). Over the years, BEL has been used to discern the involvement of iPLA2 in biological processes and, to date, is still considered the only available specific irreversible inhibitor of iPLA2. Because theS- andR-enantiomers of BEL exhibit selective inhibition of iPLA2β and iPLA2γ, respectively (148), comparison of outcomes using racemic and enantiomers of BEL facilitates distinction of effects due to the β versus the γ isoform. Studies of the mechanism of inhibition reveal that the binding of BEL to iPLA2β leads to generation of a diffusible bromoketomethyl acid, which promotes covalent modification of cysteine residues and not the active site serine of iPLA2β (139,146,149,150). In the presence of DTT, C651 was the only cysteine residue that was modified by BEL, leading to the suggestion that DTT protects iPLA2β from inactivation by BEL. However, using different isolation protocols and higher iPLA2β-specific activity, an additional interaction between C651 and active site serine, S465, was suggested to account for substantial BEL-mediated inhibition (151). While the use of BEL continues to enhance our knowledge of iPLA2β-mediated effects, its irreversible inhibitory profile, potential cytotoxicity (152,153), and several examples of inhibition of nonPLA2 enzymes (11,27,152,154,155) render it unfeasible for in vivo iPLA2 inhibition.

To improve selectivity and reduce toxicity, other compounds are being developed and fluoroketone (FK)-based inhibitors are proving to be as potent as BEL, while being more selective for iPLA2β and also exhibiting reversible inhibitory kinetics (1,156,157). Because FK inhibitors target serine active sites, they could potentially also inhibit cPLA2s. However, modification of the FK group along with addition of a hydrophobic terminus, connected by a medium-length carbon chain to mimic the fatty acid chain, conferred selectivity of the FK compounds for iPLA2 versus sPLA2 or cPLA2 (157), and earlier generation FK compounds (FKGK11 and FKGK2) were found to be beneficial in an experimental autoimmune encephalomyelitis animal model of multiple sclerosis (158). Subsequently, the FK-based inhibitor of iPLA2β (FKGK18) was found to be 7-fold more potent than FKGK11 toward iPLA2β 195 and >455 times more potent for iPLA2β than for group IVA cPLA2 and group V sPLA2, respectively (159); and effective in cell-based studies (160) and in countering T1D (161). Recently developed and awaiting characterization is an even more selective inhibitor (GK187) of iPLA2β (162). On-going deuterium exchange mass spectrometry and molecular dynamics analyses suggest that FKGK inhibitor binding to iPLA2β causes changes in the loops surrounding the active site of iPLA2β in the catalytic domain, blocking access to phospholipid substrates and reducing solvent accessibility (163). As the development of chemical inhibitors continues, newer structurally dissimilar and smaller compounds (164) with even greater selectivity for iPLA2β are forthcoming, as described at the 6th International Conference on PLA2s in 2015 (Kokotos et al., unpublished observations).

Proposed roles for iPLA2β

Membrane remodeling.

One of the earliest proposed functions for iPLA2β was a “housekeeping” role that involves generation of lysophospholipid acceptors for incorporation of AA into phospholipids, based on experiments involving inhibition of iPLA2β activity in P388D1 cells with BEL or with an antisense oligonucleotide (165,166). Inhibition of iPLA2β activity in P388D1 cells suppressed (∼60%) incorporation of [3H]AA into phospholipids while reducing (∼60%) [3H]LPC levels in [3H]choline-labeled P388D1 cells. However, [3H]palmitic acid incorporation was only slightly reduced. This is thought to represent the mechanism whereby iPLA2β inhibition reduces incorporation of [3H]AA into P388D1 cell phospholipids. Such incorporation reflects a deacylation/reacylation cycle (167) of phospholipid remodeling rather than de novo synthesis (168), and the level of LPC acceptors is thought to limit the rate of [3H]AA incorporation into P388D1 cell PC (165,166).

A second housekeeping function for iPLA2β is suggested from studies with CTP:PC cytidyltransferase (CT)-overexpressing Chinese hamster ovary cells (169). CT catalyzes the rate-limiting step in PC biosynthesis via the Kennedy pathway, and cells overexpressing CT exhibit increased rates of PC biosynthesis and degradation and little net change in PC accumulation (169). Immunoreactive iPLA2β protein and activity increase in the CT overexpressors and the increased PC degradation is prevented by BEL, suggesting that iPLA2β is upregulated in response to CT overexpression (169). In general, this could represent an important role for iPLA2β in cell biology because PC biosynthesis is involved in regulation of cell cycle and apoptosis (170).

More recently, in a study examining the effects of lipotoxicity in β-cells, the monolysocardiolipin content was reported to correlate with iPLA2β expression level (171). The authors suggested that iPLA2β contributed to cardiolipin remodeling by excising oxidized PUFA residues from cardiolipin to yield monolysocardiolipin species for reacylation with unoxidized C18:2-CoA to regenerate the native cardiolipin structure and function. This facilitated stabilization of association of cytochromec with mitochondrial membranes and decreasing its appearance in the cytosol, thereby reducing ROS-mediated apoptosis. The authors concluded that participation of iPLA2β in such an excision-reacylation mechanism of repair of oxidized phospholipids represents a special case of the originally proposed function of the enzyme in phospholipid remodeling.

Cell proliferation.

Studies utilizing chemical inhibition or genetic modification protocols reveal a positive correlation between maintenance of iPLA2β activity and cell proliferation. In the presence of BEL, human promonocytic U937 (172) and ovarian carcinoma (173) cells exhibit a decreased rate of proliferation and this is rescued in Caco-2 (174) and endothelial (175) cells by addition of AA. Consistently, knockdown of iPLA2β suppresses and overexpression of iPLA2β accelerates proliferation of insulinoma cells (176,177). Further, while proliferation of vascular smooth muscle cells from iPLA2-null mice is severely reduced, it is reversed upon addition of AA or PGE2 (178). Other studies suggest that iPLA2β is required for cell cycle progression, through both p53-dependent and -independent mechanisms (173,175,179181). The molecular mechanism whereby iPLA2β promotes cell cycle progression and proliferation remains unclear, but is likely to be related to bioactive lipid mediators that are generated by the enzyme. For example, the products of iPLA2β activity may activate genes involved in cell division (87,173,182,183). Arachidonate and eicosanoids have been linked to iPLA2β-dependent proliferation (174,175). Ovarian cancer cells produce lysophosphatidic acid (LPA) in an iPLA2β-dependent manner, and this potent mitogen acts in an autocrine fashion to induce proliferation and migration (173,182). These observations imply that regulation of iPLA2β activity may need to be considered in the context of countering tumorigenesis.

Bone formation.

AA and its metabolites are important mediators of bone remodeling. The 5-lipoxygenase (LO) products, leukotrienes and 5-HETE, function as negative modulators of bone formation by inhibiting osteoblast differentiation and bone formation (184). In contrast, PGE2 enhances bone formation and mass by increasing osteoblast replication and differentiation and/or by inhibiting osteoclastic resorption (185188), although high concentrations of PGE2 can stimulate bone resorption (189). Dietary supplementation with AA promotes increases in bone mass and volume (190,191), reflecting a beneficial role of eicosanoids in bone formation. iPLA2β-null female mice exhibit an age-related low bone/high bone adiposity phenotype that is independent of changes in estradiol levels (192). Osteoblasts and adipocytes share a common mesenchymal stem cell origin, and treatment of WT bone marrow stromal cells with BEL recapitulates the in vivo phenotype in promoting differentiation of the cells in favor of adipocytes away from osteoblasts (192). The higher adiposity in the bone marrow would be expected to compromise bone integrity, as in osteoporosis or diabetes, and this indeed is evidenced in the iPLA2β-null mice, as reflected by increased fragility and decreased strength of their bones (192). These findings suggest that iPLA2β-derived lipids play a critical role in deciding the fate of stem cells toward becoming osteoblasts or adipocytes, and raise the possibility that cell differentiation may be modulated by differential iPLA2β activation.

Male fertility.

Disruptions in PLA2 are often associated with impairment in normal reproduction. For instance, deficiency in sPLA2-III, which is expressed in proximal epididymal epithelium, causes defects in sperm maturation and impairment in the ability of the spermatozoa to fertilize intact eggs (193). These were associated with a compromised shift in acyl groups from oleic acid, linoleic acid, and AA to docosapentaenoic and docosahexaenoic acids during epididymal transit. Another sPLA2 (group X) expressed in the acrosome of spermatozoa is released in an active form during capacitation through spontaneous acrosome reaction. Deficiency in sPLA2-X promoted lower rates of spontaneous acrosome reaction and decreased in vitro fertilization efficiency (194). In contrast, only the females are affected by cPLA2 deficiency and they exhibit problems with ovulation, oocyte transport, and oocyte implantation (195). Consistent with this, female patients experiencing decreased implantation rates in in vitro fertilization have reduced cPLA2α expression and PGE2 levels (196). Similar to absences in sPLA2s, male mice with homozygous iPLA2β gene disruption have impaired reproductive ability that is marked by reduced sperm motility and ability to fertilize mouse oocytes (143). Analogous phenotype is exhibited in WT spermatozoa treated with BEL. Based on earlier reports of involvement of LPC in acrosome reaction (197199) and induction of capacitation (200), it was suggested that LPC levels are decreased due to reduction in hydrolysis of spermatozoa membrane phospholipids in the absence of iPLA2β. Females deficient in iPLA2β, however, do not appear to suffer any adverse fertility consequences (143).

iPLA2β and islet β-cells

The continued description of activity and related biochemistry of a Ca2+-iPLA2 in myocardium in the early 1990s coincided with observations that fuel secretagogue-stimulated accumulations in AA in islet β-cells occurred, in part, in the absence of Ca2+. This led to the first description of a similar Ca2+-iPLA2 activity in pancreatic islet β-cells (102) that resided predominantly in islet β-cells, with very little such activity in islet nonβ-cells. This was followed by the first demonstration of a functional role for iPLA2β activity in a biological process, where activation of iPLA2β was a requisite for optimal glucose-stimulated insulin secretion from islet β-cells (201,202), and implicated a signaling role for iPLA2β. In contrast to reports using P338D1 cells, findings utilizing multiple approaches in a variety of β-cell models indicated that the iPLA2β did not serve a membrane remodeling role, but rather, it had a signaling role in β-cells (105,142,144,176,177,203205). Further, the failure of iPLA2β manipulations to modulate membrane remodeling in native macrophages (206) suggested that the housekeeping role of iPLA2β may be cell-specific and is predominant in the macrophage-like cell line P338D1. However, the demonstration of iPLA2β activity protecting β-cells against lipotoxicity by preserving cardiolipin content (171) raises the possibility that iPLA2β may manifest an organelle- and stimuli-specific protective/remodeling role in β-cells.

Glucose-stimulated insulin secretion.

The β-cell is the primary sensor of glucose and when circulating levels of glucose rise, it is transported into the β-cell where it undergoes glycolytic metabolism (207210). During this process, ATP is generated and its binding to plasma membrane ATP-sensitive potassium (KATP) channels leads to inactivation of the channels and membrane depolarization (211214). This activates voltage-operated Ca2+ channels promoting Ca2+ influx and a rise in cytosol [Ca2+] (215219), which is a critical signal of insulin release from secretory granules (215217,220). It has been recognized for a long time that fuel secretagogues, such as glucose, also induce hydrolysis of membrane phospholipids leading to accumulations in inositol 1,4,5-trisphosphate, free AA, and AA metabolites (221225). AA, at concentrations that accumulate following stimulation with glucose, induces a rise in β-cell cytosolic Ca2+ concentrations, due in part to Ca2+ influx and Ca2+ release from intracellular (i.e., ER) stores (170,202,222,226,227). Whereas activation of phosphoinositide-phospholipase C requires Ca2+ (221), a component of the accumulation of AA and its metabolites do not (170). This led to the hypothesis that a Ca2+-independent phospholipase activity may be manifested in β-cells that is activated upon glucose stimulation, and the resulting generation of AA serves to amplify the Ca2+ signal necessary for optimal insulin secretion. Utilization of various insulinoma cell lines, rodent and human islets, molecular biological protocols, and genetically-modified mice, collectively, provided a mechanism by which stimulation of β-cells increases hydrolysis of AA from β-cell membrane phospholipids, in parallel with insulin secretion, in an iPLA2β-dependent manner (4,105,177,201,203,204). Chemical inhibition, siRNA knockdown, or genetic ablation of iPLA2β activity attenuated glucose-stimulated insulin secretion, while the opposite was evident with increased expression of β-cell iPLA2β (99,111,142,144,176,201,203205). As shown earlier in myocardial studies (87,118), the predominant lipid pools that serve as substrates for iPLA2β in β-cells are plasmalogens, and their abundance is decreased within minutes of glucose stimulation (4,203,204). Interestingly, islet β-cell membranes are enriched in AA, in particular, plasmenylethanolamine molecular species that contain AA (4,204). The presence of such a membrane phospholipid composition lends itself to accumulations in AA in the β-cells upon activation of β-cell iPLA2β. Modulation of the islet β-cell-delayed rectifier potassium channel, Kv2.1, by AA amplifies Ca2+ influx into the β-cells and enhances glucose-stimulated insulin secretion (142,228). While AA itself manifests biological activity, the accompanying lysolipids and metabolites of AA, or eicosanoids, significantly impact various cellular processes to promote a multitude of effects (229,230).

Pancreatic islets also express Ca2+-dependent PLA2s that are involved in the insulin secretory process. The cPLA2 is much more abundant in human islets than rodent islets, but very little is expressed in insulinoma cells (147,231235). cPLA2 is activated by influx of Ca2+ into the cell (232) and is subject to glucose-mediated phosphorylation (235). A variety of sPLA2s (1B, IIC, IIF, XIIA/B) have also been described in whole pancreas (236), islets (233,234,237), and in β-cell secretory granules (234,238,239) from which sPLA2-1B is co-released with insulin (236). When observations from these studies are taken together, a sequential participation of PLA2s in the insulin secretory process is suggested. Activation of iPLA2β following glucose stimulation amplifies the Ca2+ signal within the β-cell to promote insulin secretion, which can be returned to basal levels by inhibition of iPLA2β (201,204). Activation of cPLA2 due to accumulations in intracellular Ca2+ may then serve to maintain insulin secretion and inhibition of cPLA2 appears to reduce secretion only 50% (233). The role of sPLA2 may reside in its ability to enhance the ability of plasma membranes to bind secretory granules (238,240242), though a role for sPLA2 in inactivating the KATP channels has also been described (237). In view of expression of membrane receptors for sPLA2 in β-cells (238,243,244), the role of sPLA2s in β-cells may be to serve as feedback modulators to maintain/amplify insulin secretion.

iPLA2β and β-cell death and T1D

Among the roles ascribed to iPLA2β, its contribution to apoptosis was initially recognized in studies performed by Kudo’s group (97,125). Their work with human leukemic monocyte lymphoma U937 cells revealed that activation of iPLA2β, as opposed to cPLA2, enhanced cell death and that iPLA2β undergoes caspase-3 (apoptosis executioner)-catalyzed cleavage to generate a more active shorter isoform of iPLA2β. During the same period, Polonsky’s group reported that ER stress-induced death of insulinoma cells occurs independently of Ca2+ and is mediated by a metabolite of AA (245). Intriguingly, while short-term (minutes) stimulation of iPLA2β in β-cells has a beneficial effect of enhancing glucose-stimulated insulin secretion (4,105,177,201,203,204), long-term (hours) exposures to pro-inflammatory cytokines, hyperglycemia, and ER stress induce iPLA2β at the message, protein, and activity levels in β-cells (98,136138,246) and lead to deleterious consequences that ultimately cause β-cell death (98,103,104,136138,246). β-Cell apoptosis is a critical contributor to the onset and progression of diabetes and it is in this context that our group has focused on iPLA2β biology to understand the underlying molecular mechanisms by which iPLA2β-derived lipids promote β-cell death during T1D development, and our current understanding is illustrated inFig. 1.

Fig. 1.

Fig. 1.

Proposed roles for iPLA2β-derived lipid signals in promoting β-cell death leading to T1D. Our collection of studies reveal that iPLA2β activation is associated with processes that lead to β-cell apoptosis. The bioactive lipids (and their metabolites), derived from iPLA2β-catalyzed hydrolysis of membrane phospholipids, are proposed to trigger:a) generation of pro-apoptotic sphingolipids, pro-apoptotic variants of apoptotic factors, and autophagy dysfunction to promote β-cell death; andb) inflammatory responses, immune cell functionality, and chemotaxis to promote immune responses, which serve to amplify the β-cell death process. We suggest that these effects of iPLA2β-derived lipids, working in concert, contribute to the onset and progression of β-cell death, which eventually leads to the development of T1D.

iPLA2β and sphingolipids.

Stress stimuli induce the intrinsic (mitochondrial) apoptotic pathway, which is mitigated by suppressing iPLA2β expression/activity (103,104,136,138,246). Intriguingly, the primary lipid signals promoting mitochondrial decompensation were ceramides, derived through hydrolysis of sphingomyelins by neutral sphingomyelinase-2 (NSMase-2) (103,104). Ceramides are part of the sphingolipid family and act as lipid messengers that can suppress cell growth and induce apoptosis (247249), and as expected, inhibition or knockdown of NSMase-2-mitigated ceramide accumulation (103,104,137). Unexpectedly, inhibition, knockdown, or iPLA2β deficiency attenuated, and iPLA2β overexpression exacerbated, NSMase-2 induction and ceramide accumulation (103,104,136,138,246). These findings indicated that NSMase-2-catalyzed ceramide accumulations during β-cell apoptosis occur via an iPLA2β-dependent mechanism. A recent report suggests that iPLA2β-derived AA stimulates p38 MAPK and that this may serve as an additional intervening modulator of iPLA2β-mediated increase in ceramide accumulations in β-cells (250).

iPLA2β and alternate splicing.

β-Cell apoptosis due to ER stress and pro-inflammatory cytokines is mediated through the intrinsic pathway, which is dependent on mitochondrial dysfunction and activation of caspase-9 (103,104,136,138,246,251). The intrinsic apoptosis pathway is regulated by members of the Bcl-2 family of proteins that can be pro- or anti-apoptotic, depending on the spectrum of Bcl-2 homology domains that they contain. Among the anti-apoptotic Bcl-2 family members is Bcl-x(L), which associates with mitochondrial membranes and prevents their permeabilization, an early step in the intrinsic apoptosis pathway (252,253). Overexpression of Bcl-x(L) has been correlated with increased survival of a variety of cells and tissues (254), including islet β-cells (255257). Bcl-x(L)-null β-cells are hypersensitive to pro-apoptotic stimuli and reduced expression of Bcl-x(L) protein correlates with β-cell apoptosis in response to immunosuppressive drugs or high glucose (255258). Conversely, overexpression of exogenous Bcl-x(L) protects β-cells from pro-inflammatory cytokine- and thapsigargin-induced apoptosis (258,259).

Modulation of Bcl-x(L) expression is a complex mechanism consisting of both transcriptional and posttranscriptional processes and often leads to generation of both pro- and anti-apoptotic proteins from a single pre-mRNA (254,260). Bcl-x(L) is the most abundant variant of the Bcl-x pre-mRNA, but other species can be generated at the expense of the mature mRNA encoding this anti-apoptotic protein (254). To date, Bcl-x RNA splicing has not been investigated in the β-cell, especially in the context of β-cell apoptosis and diabetes mellitus. We find that increased expression of iPLA2β in β-cells is associated with a lower ratio of Bcl-x(L) to Bcl-x(s), while the opposite is true with iPLA2β inhibition or deficiency (261). Lipidomic analyses by mass spectrometry revealed that the ratio of Bcl-x(L)/x(s) was directly proportional to the ratio of 5-HETE to EPA, suggesting that prolonged iPLA2β activation in β-cells promotes generation of lipids, AA- and nonAA-derived, away from 5-HETE, thus disfavoring generation of the anti-apoptotic Bcl-x(L) variant. These findings raise the possibility that among the mechanisms by which iPLA2β promotes β-cell apoptosis is one in which its activation triggers alternate splicing events favoring apoptotic processes. Of note, β-cell death due to direct activation of the mitochondrial apoptotic pathway by staurosporine is mitigated by iPLA2β activity and this was related to preservation of repair mechanisms to sustain mitochondrial membrane components, such as cardiolipins (262,263).

iPLA2β and immune responses.

β-Cell subcellular membranes are enriched in AA-containing phospholipids (144,203,204) and iPLA2β activation leads to hydrolysis of AA, which can be metabolized by COX and LO enzymes to generate eicosanoids (97,125,245,264). These bioactive lipids act as paracrine and autocrine factors and greatly contribute to inflammation (264266) and autoimmune diseases (267274). PGE2 has been reported to reduce debris clearance (275), and apoptotic clearance defects in non-obese diabetic (NOD) macrophages and dendritic cells are attributed to high PGE2 levels (276,277). It might be speculated that suppression of PGE2 or other COX/LO-derived metabolite (278) generation can prevent the spread of β-cell apoptosis. Further, lysolipids and eicosanoids act as “chemoattractants” (230,279281), and increased generation of these could promote migration of immune cells toward the islet and subsequent infiltration, raising the possibility that in vivo inhibition of iPLA2β could mitigate immune responses leading to T1D. Autoimmune insulitis is marked by degradation of the islet basement membrane, which presents a physical barrier to infiltrating leukocytes (282). Remarkably, the abundance of infiltrating cells is significantly reduced in NOD mice treated with FKGK18 (161).

It is well-established that pancreatic islet β-cells are subject to pro-inflammatory cytokine-induced apoptosis during the pathogenesis of T1D (283). Our group has observed that in addition to inducing iPLA2β, pro-inflammatory cytokines promote ER stress, LPC generation, caspase-3 activation, and β-cell apoptosis and these outcomes were prevented following inhibition of iPLA2β withS-BEL (136). Further, addition of the cytokines to islets overexpressing iPLA2β resulted in exacerbated ER stress and β-cell apoptosis, which were blunted in islets devoid of iPLA2β (136). Taken together, these findings provided evidence of iPLA2β participation in β-cell apoptosis due to pro-inflammatory cytokines. This prompted further investigation of the role of iPLA2β in the pathogenesis in the NOD mouse, a model of spontaneous autoimmune-mediated T1D (161). In this study, we found that following administration of iPLA2β-selective inhibitor FKGK18, NOD mice had significantly reduced incidence of diabetes and reduced insulitis, higher circulating insulin, and preservation of β-cell mass (161). Additionally, FKGK18 reduced TNFα production from CD4+ T-cells and antibodies from B-cells, independent of iPLA2γ activation, suggesting modulation of immune cell responses by iPLA2β or iPLA2β-derived products (161). Furthermore, adoptive transfer of diabetes by CD4+ T-cells to immunodeficient and nondiabetogenic NOD.scid mice was mitigated by FKGK18 pretreatment and TNFα production from CD4+ T-cells was reduced by inhibitors of COX and 12-LO (161). These observations suggest that modulation of immune cell function may be a mechanism by which iPLA2β and iPLA2β-derived lipid signals participate in autoimmune-mediated β-cell death. In support of involvement of iPLA2β-derived lipid signals is the recent observation that 12-LO products contribute to pro-inflammatory cytokine-mediated β-cell dysfunction and apoptosis (284).

iPLA2β and autophagy.

Whereas apoptosis is a well-studied process in β-cells, autophagy in β-cells has not received significant attention. Autophagy is a constitutively active process of cellular degradation in all cell types and is regarded as a generally protective process to prolong cell survival. However, under increased stress, dysregulation of autophagy can lead to cell death (285). In view of evidence linking ceramides with autophagy (286,287) and our collective observations that ceramide generation can occur via an iPLA2β-mediated mechanism, we addressed the possibility that iPLA2β expression modulates β-cell autophagy (246). We found that thapsigargin-induced ER stress promoted a greater conversion of LC3-I to LC3-II, reflecting activation of the autophagic response in islets from RIP-iPLA2β-Tg mice relative to islets from age-matched WT mice. In contrast, LC3-II generation was decreased in islets deficient in iPLA2β. Thapsigargin is thought to specifically block fusion of autophagosomes with lysosomes (288) and in the presence of inhibitors of autolysosomal activity, LC3-II accumulations in the RIP-iPLA2β-Tg and iPLA2β-KO mice are similar to their corresponding WT groups. Interestingly, inhibition of NSMase-2 in the presence of thapsigargin had minimal effect in WT islets, but modestly reduced LC3-II flux in the Tg. These findings suggest that differential iPLA2β activation can impact β-cell autophagy and that the most likely point of effect is beyond the induction step, in part, mediated by NSMase-2.

While this was the first demonstration of a link between iPLA2β and autophagy, it has been reported that mice deficient in iPLA2γ had enlarged hippocampal mitochondria, and that their degeneration led to an increase in autophagy (73). Those authors concluded that iPLA2γ-deficiency decreased mitochondrial membrane remodeling, resulting in loss of membrane potential and subsequent mitochondrial dysfunction leading to cognitive dysfunction and increased autophagy in the hippocampus. However, more detailed studies are needed to identify the precise location of iPLA2β impact on the autophagic responses in β-cells.

iPLA2β and diseases

Neurodegenerative.

PLA2s, including iPLA2β, are widely expressed in different regions of the brain (289291), and iPLA2β immunoreactivity has been reported to be predominant in the cytosol and of less abundance in the nuclear fraction (292), suggesting a role for the enzyme and its products in nuclear signaling. iPLA2β has been purified from rat brain cytosol (293) and identified in dendrites and neurons, microglia, Purkinje cells, and astrocytes (290,294296), suggesting that it also participates in neuronal signaling. Advances in lipidomic analyses, coupled with kinetic studies in rodents and positron emission tomography protocols in humans, reveal that the turnover of AA in the brain is associated with cPLA2 and COX-2 activities, whereas that of DHA (22:6n-3) was related to iPLA2β and COX-1 activities (297). This is consistent with the reports that in contrast to the islets, which are enriched in AA-containing glycerophospholipids, the most prominentsn-2 substituent in brain glycerophospholipids is DHA (297,298), and iPLA2β-deficiency results in reduced DHA metabolism and content in the brain (299). Considering its ubiquitous expression in the brain, dysregulation of iPLA2β, as opposed to or in association with other PLA2s, has been recognized to play critical roles in a variety of neurological disorders.

Schizophrenia.

Magnetic resonance spectroscopy studies reveal a higher than normal turnover of brain membrane phospholipids in patients with schizophrenia (300,301). Whereas serum Ca2+-dependent PLA2 activity appears to be unchanged, iPLA2β is significantly higher in patient sera relative to control subjects (300). Consistent with these findings, iPLA2β activity was increased (nearly 45%) in the temporal cortex of patients and proposed to be a causative factor in abnormal fatty acid metabolism and oxidative stress in schizophrenia (302). The increased breakdown in membrane phospholipids due to higher increases in iPLA2β activity, may be reflected by alterations in neuronal membrane properties leading to hypodopaminergy (303). In contrast, Ca2+-dependent PLA2 activity was decreased in the patients with schizophrenia (302). Another report suggests that increases in serum iPLA2β activity are only evident in first-episode or acute early phase schizophrenia, and not apparent at chronic stages or in patients experiencing multi-episode schizophrenia (304). Brain samples from epilepsy patients with schizophreniform symptoms also express higher iPLA2β activity (305). Genetic studies to identify PLA2 polymorphisms associated with schizophrenia are, however, unclear. While allelic association betweenPLA2G6 gene polymorphism has been reported, in the absence of similar links to PLA2G1A or PLA2GIIA-D (sPLA2s), PLA2GIVA-C (cPLA2), PNPLA3 (iPLA2ε), or PNPLA8 (iPLA2γ) genes (306,307), others have reported potential association between schizophrenia and PLA2GIVA (303) and PLA2GIVC (308).

Alzheimer’s disease.

This disease is characterized by the polymerization of amyloid β and especially tau proteins, leading to the formation of senile plaques and neurofibrillary tangles, and such polymerization is stimulated by AA and other unsaturated fatty acids (309). All groups of PLA2s have been proposed to play a role in the development of Alzheimer’s disease, which is a leading cause of dementia in the elderly. For instance, sPLA2s (IIA, V, IVA) are thought to be involved in neuronal death, sPLA2s (III and X) with neurogenesis, and cPLA2 and iPLA2 in both neuronal death and neurogenesis (304). A frontal variant of Alzheimer’s disease, in which there is a high occurrence of neurofibrillary tangles in the frontal cortex, is accompanied by a decrease in iPLA2, but not cPLA2, activity in the dorsolateral prefrontal cortex (310). This report suggests that this may be a compensatory response to accelerated phospholipid metabolism early in the disorder. Consistently, the predominant PLA2 activity in rodent hippocampal slices was reported to be manifested by iPLA2 and to be essential for synaptic plasticity, as reflected in iPLA2 inhibition studies (311). This was later confirmed in whole animal studies (141), leading to the suggestion that reductions in cPLA2 and iPLA2 activities can have adverse effects on memory in patients with Alzheimer’s (312). These studies, however, did not distinguish which iPLA2 (β or γ) isoform activity was critical for the dementia associated with Alzheimer’s. However, a more recent report suggests that the early-onset, but not the later-onset, fronto-temporal type of dementia may be linked toPLA2G6 mutations (313). Further, sPLA2, cPLA2, and iPLA2 activities were reported to be reduced in serum from patients with bipolar disorders, who have a 5-fold increased risk for developing Alzheimer’s disease (314).

Parkinson’s disease.

Selective degeneration of dopaminergic neurons in the substantia nigra has been suggested to be related to low activity of phospholipid catabolic/anabolic enzymes, which may promote the oxidative membrane damage associated with Parkinson’s disease that begins at 15–30 years of age (315). Many recessive loci have been linked to Parkinson’s disease; however, they all do not associate with the disease. Homozygosity mapping and mutational analyses over the past 5 years reveals a complicated connection with mutations in thePLA2G6 gene at the PARK14 locus. Initially recognized in adult-onset parkinsonism (316),PLA2G6 gene mutations are now linked to levodopa-responsive parkinsonism with severe impairments in swallowing, dystonia, and pyramidal weakness (317,318), and to autosomal early-onset parkinsonism (313,318322). However, studies among Asian populations are conflicting, where a link betweenPLA2G6 mutations and Parkinson’s disease was reported in Singapore (323), but not in China (324,325) or Japan (326).

INAD and NBIA.

Infantile neuroaxonal dystrophy (INAD) and neurodegeneration with brain iron accumulation (NBIA) are two additional autosomal recessive neurodegenerative diseases, which begin within 1–2 years of life and are characterized by widespread neurodegeneration that includes psychomotor regression, spasticity, and optic atrophy. INAD is characterized by the presence of axonal spheroids throughout the central and peripheral systems. NBIA patients are a subset of INAD patients with brain iron accumulation and they feature idiopathic neurodegeneration. A potential link between INAD andPLA2G6 mutations was first reported in two unrelated Bedouin Israeli kindreds (327) and in a 2-year-old boy with psychomotor regression (328), and the pathogenesis is thought to begin prior to birth (329). The accompanying clinical INAD symptoms were found to recapitulate in subsequently generated global iPLA2β-null mice (330,331). Screening of DNA from human patients with INAD and NBIA identified 80% of INAD and 20% of NBIA patients with mutations in thePLA2G6 gene, and neuropathologic changes that were analogous to those associated with Parkinson’s and Alzheimer’s diseases (332,333). Studies using iPLA2β-null mice suggest degeneration of mitochondrial inner membranes and presynaptic membranes (334), reductions in capacitative Ca2+ entry in astrocytes (335), point mutation in the Ank repeat producing an inactive iPLA2β protein (336), and neuro-inflammation and Purkinje cell loss (337) as potential underlying mechanisms that lead to pathogenesis in neurodegenerative disorders associated with brain iron accumulations such as INAD, NBIA (338), Karak syndrome (339), and Parkinson’s and Alzheimer’s diseases accompanied by iron accumulations. In this regard, it should be noted that common mutations and combination of mutations inPLA2G6-associated neurodegeneration may be associated with Parkinson’s and Alzheimer’s diseases in the absence or presence of iron accumulations (316,339347).

Other roles in the CNS.

Additional evidence of iPLA2β involvement in neurodegenerative disorder include: association of a subset of Shindler’s disease in infants withPLA2G6 mutations (348); elevations in iPLA2β in patients with bipolar l disorder and a history of psychosis (349); neuro-inflammation and associated neuropathology with motor dysfunction in later life due to iPLA2β-deficiency (350); and roles for iPLA2β during early beneficial stages of myelin breakdown following peripheral nerve injury (351) and detrimental demyelination due to spinal cord injury (352), brain endothelial cell migration and proliferation (353), antidepressant-like effects of maprotiline (354), and pro-oxidative signaling related to ethanol-induced neurotoxicity (355).

Cancers.

The ability of iPLA2β to promote cell proliferation becomes prominent in the context of tumorigenesis. Several in vitro studies reveal higher expression of iPLA2β in stimulated immortal cell lines and that chemical inhibition or siRNAs targeted against iPLA2β reduces proliferation and promotes apoptosis of the cells (97,108,125,356364). Subsequent studies targeting specific cancers suggest that iPLA2β promotes cancer cell growth via signal transduction pathways involving epidermal growth factor receptors, MAPKs, E3 ubiquitin-protein ligase mdm2, tumor suppressor protein p53, and cell cycle regulator p21 (365367). In addition, there is increasing support for a role of iPLA2β and iPLA2β-derived LPA in promoting cancer cell proliferation and metastasis.

Ovarian.

A significant number of studies have explored the impact of PLA2 activation in the context of ovarian cancer development. A potential involvement of cPLA2 and iPLA2 activities was recognized in women with endometrial dysfunction, who had a 4-fold increase in PLA2 activity (368). Studies with ovarian carcinoma cells demonstrated a role for iPLA2β, but not for cPLA2, in promoting S and G2/M cell cycle phases, that was independent of p53 (173). BEL prevented these effects, but they were rescued with addition of LPA, and knockdown of iPLA2β inhibited cell proliferation in culture and tumorigenicity of cancer cell lines in nude mice. Consistent with these findings, tumorigenesis and ascites formation associated with the epithelial ovarian cancer cell line ID8 administration were reduced by nearly 50% in iPLA2β-null mice, as were the levels of LPA and LPC (370). Such inhibition was elevated to 95% when ID8 cells in which iPLA2β was knocked down were used, suggesting the importance of iPLA2β activity in host and tumor cells for cancer progression. Further, LPA, but not LPC, enhanced in vivo ascites formation and tumorigenesis in the iPLA2β-null mice. Similar inhibition of cell adhesion, migration, and invasion of epithelial ovarian cancer cells was demonstrated with a structurally dissimilar iPLA2β inhibitor, FKGK11 (371), confirming a role for iPLA2β in these processes. In contrast, reports using nonselective (372) and selective PLA2 inhibitors combined with targeted genetic knockdowns (373) suggest that both cPLA2 and iPLA2β activities contribute to elevations in LPA and to ovarian cancer development.

Other cancers.

Presently, there are limited numbers of studies linking iPLA2β activity to development of other cancers; however, this area has experienced growth over the past 5 years. Comparison of LNCaP prostate cancer cells with normal prostate epithelial RWPE-1 cells revealed decreased expression of inhibitory Ank-iPLA2β, but increases in expression (VIA-1 and -2) and activity of iPLA2β in the cancer cells. Both inhibition of iPLA2β with BEL and iPLA2β knockdown reduced prostate-specific antigen (PSA) secretion from and apoptosis of the cancer cells (374). Exogenous PSA rescued inhibition of apoptosis by BEL, suggesting that iPLA2β modulates PSA secretion, which in turn provides an autocrine survival function. Inhibition of iPLA2β, but not iPLA2γ, in p53-positive LNCaP cells was also found to be associated with activation of p38 and induction of reactive species, which leads to cell cycle arrest and cytostasis (375). Lung tumor growth in an in vivo allograft model is promoted by PRDX6, which is a bifunctional protein with glutathione peroxidase (GPx) and iPLA2β activities (376). Nude mice bearing PRDX6-overexpressing lung cancer cells exhibited increases in tumor size and weight and increased expression of both GPx and iPLA2β, and these outcomes were inhibited when a mutant PRDX6 was used, suggesting that PRDX6 promotes lung tumor growth via GPx and iPLA2β. Occurrence of breast cancer is often accompanied by metastasis to the lung and injection of E0771 breast cancer cells in the mouse mammary pads resulted in an 11-fold higher number of breast cancer cells in WT lungs, relative to lungs from iPLA2β-null mice (377). Further, production of thrombin-stimulated platelet-activating factor in lung endothelial cells was increased in the WT lungs and absent in the iPLA2β-null mice. Taken together with the finding that inhibition of iPLA2β, but not iPLA2γ, prevented increases in PAF in WT cells suggests that iPLA2β modulation of PAF production is a factor in cancer cell metastasis. Consistent with these findings, cigarette smoke extract increased PAF accumulations and increased cell motility in MDA-MB-231 breast tumor cells, and these effects were mitigated by iPLA2β inhibition (378). Recently, unicortin, a member of the corticotrophin-releasing factor family, was shown to promote hepatic cancer cell line migration by upregulating cPLA2 and suppress migration by downregulating iPLA2β at the transcriptional level (379). Polymorphisms inPLA2G6 have also been linked to increased risks for colorectal (380) and invasive cutaneous melanoma (381,382).

Cardiovascular.

The myocardial 40 kDa iPLA2, described as a plasmalogen-selective PLA2 (3,118), was inhibited by BEL (146), stabilized by ATP (117), and formed an oligomeric regulatory complex with phosphofructokinase (384). From the collection of related studies, it can be ascertained that iPLA2β activation in the heart may be beneficial and also detrimental (265,385,386). For instance, increases in membrane-iPLA2 (387) and mitochondrial-iPLA2 (388) activities were associated with irreversible cell damage during myocardial ischemia and reperfusion; cardiomyopathy associated with HIV infection may involve an iPLA2 signaling pathway (389); and iPLA2β activation contributing to arrhythmogenic conduction slowing due to ischemia in diabetic hearts (390) have been reported. In contrast, iPLA2 activation or its preference for plasmalogens was evident during global ischemia (391). However, iPLA2 activation facilitated incorporation of 18:2 into (18:2)4-cardiolipin to maintain mitochondrial bioenergetics during heart failure (392), protected against oxidant-induced cardiotoxicity (393), and contributed to cardiac endothelial cell production of PGI2, which manifests protective effects in the heart (394). In Barth syndrome, a metabolic disorder caused by mutations in the mitochondrial transacylase tafazzin that is characterized with disturbances in cardiolipin abundance and molecular species, cardiolipin deacylation by iPLA2β, but not iPLA2γ, is thought to play a role (395,396). There remains some uncertainty about which iPLA2 isoform is expressed in the myocardium, with both iPLA2γ (385) and iPLA2β (265) reported to be the predominant isoform, and their relative contribution to myocardial PLA2 activity.

iPLA2β has also been implicated in activation of store-operated Ca2+-channels and Ca2+ release-activated Ca2+-channels in a variety of cells, and it has been suggested that an endogenous calcium influx factor activates iPLA2β when applied to cell homogenates (397399). In this context, inhibition of iPLA2β reduces endothelial agonist-induced intracellular Ca2+ release and extracellular Ca2+ influx, suggesting that iPLA2β is an important mediator of vascular relaxation (400).

Inflammatory and autoimmune.

iPLA2β activity has been reported in many components of the immune system, including macrophages (6,401404), monocytes (405), neutrophils (406,407), mast cells (408), and T-cells and B-cells (409). Much like different immune cells can have differing roles within the immune system; iPLA2β can have cell-specific roles. Among its proposed roles in macrophages, iPLA2β has been implicated in playing a major role in free fatty acid accumulation in macrophages (165,410412) leading to apoptosis. iPLA2β, but not cPLA2, has also been reported to promote macrophage proliferation (413). Interestingly, this phenomenon may be cell-specific, as transfection of a kidney cell line with iPLA2β reduced cell proliferation (125), whereas, mature T- and B-lymphocytes do not express iPLA2β and inhibition of iPLA2β in immature T- and B-cells reduces cell viability and proliferation (409). Altered proliferation may work in tandem with increased apoptosis due to iPLA2β-derived free fatty acids. Further, iPLA2β-derived LPC during apoptosis has been reported to promote binding of IgM antibodies to dying cells and promote their clearance (414). Thus, iPLA2β-derived free fatty acids may have both proliferative and apoptotic effects dependent on the specific cell system and/or stimuli.

More clearly evidenced, however, is the contribution of iPLA2β in macrophage adhesion during inflammation. The iPLA2β is required for maintenance of macrophage spreading and adhesion (415). Further, adhesion of macrophages is coupled by iPLA2β-mediated AA release and 12/15-LO activity on receptors for macrophages within the extracellular matrix, contributing to inflammation by increasing macrophage retention at inflammation sites (416). Interestingly, the lack of iPLA2β resulted in macrophage buildup late after nerve injury, suggesting iPLA2β is involved in late state macrophage clearance (351). In support of this phenomenon, macrophage phagocytosis was blunted in iPLA2β-depleted cells and increased by overexpression of iPLA2β (417). Furthermore, chemical inhibition of iPLA2β resulted in decreased macrophage IgG-mediated phagocytosis, which was associated with decreased AA release from the macrophages (418,419). Thus, iPLA2β likely influences macrophage activity, translocation, and attachment to the site of inflammation, and clearance of debris and apoptotic cells at the end of inflammation.

As might be expected, products derived from iPLA2β activation can participate in inflammatory responses, which are also triggered during development of autoimmune diseases. Several factors, including ER stress (420,421), ROS (422,423), and NF-κB (424,425), contribute to inflammatory diseases (i.e., autoimmune, metabolic, cancers, rheumatoid arthritis). NADPH oxidases generate ROS and cytokines induce it in a 12-LO product-dependent manner (230). ROS induce iPLA2β (82,426432) and ROS generation is increased in neutrophils by iPLA2β activation (433) and reduced in macrophages with iPLA2β deficiency (434). Various stresses activate NF-κB (435438), and iPLA2β likely modulates NF-κB via ROS generation (439). ROS subsequently induce the chemoattractant, MCP-1 (434), raising the possibility that there is crosstalk between NF-κB and iPLA2β. Further, chemotaxis in response to MCP-1 requires iPLA2β activation (440). It has been suggested that iPLA2β activity drives the initial inflammatory response through synthesis of PGE2, LTB4, and IL-1β, and that sPLA2s and cPLA2 are activated during the resolution phase (441).

The development of FK reversible inhibitors is facilitating testing the impact of PLA2s in autoimmune diseases. Comparisons of earlier generation FKGK11 (iPLA2) and FKGK2 (strong pan) with cPLA2 inhibition in an experimental autoimmune encephalomyelitis animal model of multiple sclerosis revealed that cPLA2 was involved in early onset, iPLA2β-deficiency in the onset and progression, and sPLA2 in later remission stages of experimental autoimmune encephalomyelitis (158).

Metabolic.

Studies in iPLA2β-null mice revealed that while fasting and fed blood glucose are unchanged from WT mice, iPLA2β-null mice develop more severe hyperglycemia than WT mice after administration of multiple low doses of streptozotocin (144). This led to the suggestion that iPLA2β results in an impaired ability to compensate for metabolic stresses. Consistent with this, high-fat diet exacerbates glucose tolerance in the iPLA2β-null mice, relative to WT mice. However, in spite of a global iPLA2β-deficiency, these mice do not develop dyslipidemia in response to high-fat diet (442).

CONCLUDING COMMENTS

In addition to the above-discussed iPLA2s, other smaller mass isoforms with different substrate preferences and susceptibility to BEL have been reported in the kidney (443,444) and rat parotid gland (445). It is likely that continued studies will reveal additional novel iPLA2 activities that are products of different genes or are generated via alternate splicing or proteolytic cleavage of the currently identified isoforms. Regardless, it is clear that the iPLA2s participate in numerous biological processes and that modulation of their cell and/or organelle-specific expression or activity can have profound beneficial or detrimental consequences on membrane integrity and signal transduction. As global and tissue-specific iPLA2-KO or Tg models continue to be developed, so will our understanding of the specific roles of iPLA2s in vivo. Studies incorporating genetic analyses will no doubt aid in identifying further associations between diseases and polymorphisms in the iPLA2 genes. While the focus of this review is on iPLA2s, it is readily apparent that cPLA2s and sPLA2s may act sequentially or in concert with iPLA2s in a cell/organelle-specific manner to produce relevant effects, and this should be a consideration in further exploration of the roles of PLA2 in biological processes. Over the past decade, important links between iPLA2 dysregulation and various diseases have come to the forefront, as illustrated inFig. 2, and with continued studies of this family of PLA2s, a greater understanding of the importance of iPLA2-derived lipid signaling in disease development can be attained and this will facilitate identification of novel pathways that can potentially be targeted for drug therapy. Herein, every attempt was made to provide a comprehensive current understanding of iPLA2s and omission of any relevant citations was unintentional. Readers interested in further elaboration of the areas addressed here are directed to other reviews (1,153,289,291,365367,447451).

Fig. 2.

Fig. 2.

Biological roles and consequences of iPLA2 activation. The different isoforms of iPLA2 (δ, ε, ζ, η, γ, and β) manifest cell/organelle-specific roles by expressing a variety of activities at the plasma membrane, ER, mitochondria, peroxisomes, and nucleus. The outcomes can be homeostatic and beneficial under normal conditions, but when the expression and/or activity are dysregulated (increased or decreased), they can be detrimental and lead to a variety of disorders. While the illustration describes the involvement of iPLA2s, it might be expected that cPLA2s and sPLA2s, in a cell/organelle-specific manner, participate in a sequential manner or in concert with the iPLA2s to produce the various outcomes.

Footnotes

Abbreviations:

AA
arachidonic acid
Ank
ankyrin
ATGL
adipose triglyceride lipase
BEL
bromoenol lactone
CGI-58
comparative gene identification-58
COX
cyclooxygenase
cPLA2
cytosolic phospholipase A2
CT
cytidyltransferase
ER
endoplasmic reticulum
FK
fluoroketone
FKGK18
fluoroketone-based inhibitor of iPLA2β
GPx
glutathione peroxidase
INAD
infantile neuroaxonal dystrophy
iPLA2
independent phospholipase A2
iPLA2β
cytosol-associated Ca2+-independent phospholipase A2
iPLA2β-KO
global iPLA2β-null mice
iPLA2γ
membrane-associated Ca2+-independent phospholipase A2
LO
lipoxygenase
LPA
lysophosphatidic acid
LPC
lysophosphatidylcholine
NBIA
neurodegeneration with brain iron accumulation
NOD
non-obese diabetic
NSMase-2
neutral SMase-2
NTE
neuropathy target esterase
PAF
platelet-activating factor
PC
phosphatidylcholine
PE
phosphatidylethanolamine
PGE2
prostaglandin E2
PLA2
phospholipase A2
PNPLA
patatin-like phospholipase
PSA
prostate-specific antigen
RIP-iPLA2β-Tg
mice which overexpress iPLA2β only in β-cells
R-BEL
enantiomer of BEL selective for iPLA2γ
ROS
reactive oxygen species
S-BEL
enantiomer of BEL selective for iPLA2β
sPLA2
secretory phospholipase A2
SRE
sterol regulatory element
SREBP
sterol regulatory element-binding protein
Tg
transgenic

Our group’s data was supported by lipidomic analyses through the Mass Spectrometry Resource at Washington University School of Medicine and Virginia Commonwealth University; human islets from and Juvenile Diabetes Research Foundation and National Institutes of Health sponsored islet distribution centers; islet morphometry and histology cores at Washington University School of Medicine and University of Alabama at Birmingham; and funding from the National Institutes of Health/National Institute of Diabetes and Digestive and Kidney Diseases (R01-DK69455), American Diabetes Association, Iacocca Family Foundation, National Science Foundation, Seed/Pilot & Feasibility awards from the University of Alabama at Birmingham (Diabetes Research Center, Comprehensive Diabetes Center, Comprehensive Cancer Center, and Center for Metabolic Bone Diseases).

REFERENCES

  • 1.Dennis E. A., Cao J., Hsu Y. H., Magrioti V., Kokotos G.2011. Phospholipase A2 enzymes: physical structure, biological function, disease implication, chemical inhibition, and therapeutic intervention. Chem. Rev.111: 6130–6185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gijón M. A., Leslie C. C.1997. Phospholipases A2. Semin. Cell Dev. Biol.8: 297–303. [DOI] [PubMed] [Google Scholar]
  • 3.Wolf R. A., Gross R. W.1985. Identification of neutral active phospholipase C which hydrolyzes choline glycerophospholipids and plasmalogen selective phospholipase A2 in canine myocardium. J. Biol. Chem.260: 7295–7303. [PubMed] [Google Scholar]
  • 4.Ramanadham S., Wolf M. J., Jett P. A., Gross R. W., Turk J.1994. Characterization of an ATP-stimulatable Ca2+-independent phospholipase A2 from clonal insulin-secreting HIT cells and rat pancreatic islets: a possible molecular component of the beta-cell fuel sensor. Biochemistry.33: 7442–7452. [DOI] [PubMed] [Google Scholar]
  • 5.Portilla D., Shah S. V., Lehman P. A., Creer M. H.1994. Role of cytosolic calcium-independent plasmalogen-selective phospholipase A2 in hypoxic injury to rabbit proximal tubules. J. Clin. Invest.93: 1609–1615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ackermann E. J., Kempner E. S., Dennis E. A.1994. Ca2+-independent cytosolic phospholipase A2 from macrophage-like P388D1 cells. Isolation and characterization. J. Biol. Chem.269: 9227–9233. [PubMed] [Google Scholar]
  • 7.Dennis E. A.1997. The growing phospholipase A2 superfamily of signal transduction enzymes. Trends Biochem. Sci.22: 1–2. [DOI] [PubMed] [Google Scholar]
  • 8.Balsinde J., Dennis E. A.1997. Function and inhibition of intracellular calcium-independent phospholipase A2. J. Biol. Chem.272: 16069–16072. [DOI] [PubMed] [Google Scholar]
  • 9.Kienesberger P. C., Oberer M., Lass A., Zechner R.2009. Mammalian patatin domain containing proteins: a family with diverse lipolytic activities involved in multiple biological functions. J. Lipid Res.50(Suppl): S63–S68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wilson P. A., Gardner S. D., Lambie N. M., Commans S. A., Crowther D. J.2006. Characterization of the human patatin-like phospholipase family. J. Lipid Res.47: 1940–1949. [DOI] [PubMed] [Google Scholar]
  • 11.van Tienhoven M., Atkins J., Li Y., Glynn P.2002. Human neuropathy target esterase catalyzes hydrolysis of membrane lipids. J. Biol. Chem.277: 20942–20948. [DOI] [PubMed] [Google Scholar]
  • 12.Baulande S., Lasnier F., Lucas M., Pairault J.2001. Adiponutrin, a transmembrane protein corresponding to a novel dietary- and obesity-linked mRNA specifically expressed in the adipose lineage. J. Biol. Chem.276: 33336–33344. [DOI] [PubMed] [Google Scholar]
  • 13.Johnson M. K.1969. A phosphorylation site in brain and the delayed neurotoxic effect of some organophosphorus compounds. Biochem. J.111: 487–495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Johnson M. K.1969. The delayed neurotoxic effect of some organophosphorus compounds. Identification of the phosphorylation site as an esterase. Biochem. J.114: 711–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Winrow C. J., Hemming M. L., Allen D. M., Quistad G. B., Casida J. E., Barlow C.2003. Loss of neuropathy target esterase in mice links organophosphate exposure to hyperactivity. Nat. Genet.33: 477–485. [DOI] [PubMed] [Google Scholar]
  • 16.Akassoglou K., Malester B., Xu J., Tessarollo L., Rosenbluth J., Chao M. V.2004. Brain-specific deletion of neuropathy target esterase/swisscheese results in neurodegeneration. Proc. Natl. Acad. Sci. USA.101: 5075–5080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Read D. J., Li Y., Chao M. V., Cavanagh J. B., Glynn P.2009. Neuropathy target esterase is required for adult vertebrate axon maintenance. J. Neurosci.29: 11594–11600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Rainier S., Bui M., Mark E., Thomas D., Tokarz D., Ming L., Delaney C., Richardson R. J., Albers J. W., Matsunami N., et al. 2008. Neuropathy target esterase gene mutations cause motor neuron disease. Am. J. Hum. Genet.82: 780–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Rainier S., Albers J. W., Dyck P. J., Eldevik O. P., Wilcock S., Richardson R. J., Fink J. K.2011. Motor neuron disease due to neuropathy target esterase gene mutation: clinical features of the index families. Muscle Nerve.43: 19–25. [DOI] [PubMed] [Google Scholar]
  • 20.Hein N. D., Stuckey J. A., Rainier S. R., Fink J. K., Richardson R. J.2010. Constructs of human neuropathy target esterase catalytic domain containing mutations related to motor neuron disease have altered enzymatic properties. Toxicol. Lett.196: 67–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hein N. D., Rainier S. R., Richardson R. J., Fink J. K.2010. Motor neuron disease due to neuropathy target esterase mutation: enzyme analysis of fibroblasts from human subjects yields insights into pathogenesis. Toxicol. Lett.199: 1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Topaloglu A. K., Lomniczi A., Kretzschmar D., Dissen G. A., Kotan L. D., McArdle C. A., Koc A. F., Hamel B. C., Guclu M., Papatya E. D., et al. 2014. Loss-of-function mutations in PNPLA6 encoding neuropathy target esterase underlie pubertal failure and neurological deficits in Gordon Holmes syndrome. J. Clin. Endocrinol. Metab.99: E2067–E2075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Synofzik M., Gonzalez M. A., Lourenco C. M., Coutelier M., Haack T. B., Rebelo A., Hannequin D., Strom T. M., Prokisch H., Kernstock C., et al. 2014. PNPLA6 mutations cause Boucher-Neuhauser and Gordon Holmes syndromes as part of a broad neurodegenerative spectrum. Brain.137: 69–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hufnagel R. B., Arno G., Hein N. D., Hersheson J., Prasad M., Anderson Y., Krueger L. A., Gregory L. C., Stoetzel C., Jaworek T. J., et al. 2015. Neuropathy target esterase impairments cause Oliver-McFarlane and Laurence-Moon syndromes. J. Med. Genet.52: 85–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kmoch S., Majewski J., Ramamurthy V., Cao S., Fahiminiya S., Ren H., MacDonald I. M., Lopez I., Sun V., Keser V., et al. 2015. Mutations in PNPLA6 are linked to photoreceptor degeneration and various forms of childhood blindness. Nat. Commun.6: 5614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.He S., McPhaul C., Li J. Z., Garuti R., Kinch L., Grishin N. V., Cohen J. C., Hobbs H. H.2010. A sequence variation (I148M) in PNPLA3 associated with nonalcoholic fatty liver disease disrupts triglyceride hydrolysis. J. Biol. Chem.285: 6706–6715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Jenkins C. M., Mancuso D. J., Yan W., Sims H. F., Gibson B., Gross R. W.2004. Identification, cloning, expression, and purification of three novel human calcium-independent phospholipase A2 family members possessing triacylglycerol lipase and acylglycerol transacylase activities. J. Biol. Chem.279: 48968–48975. [DOI] [PubMed] [Google Scholar]
  • 28.Romeo S., Kozlitina J., Xing C., Pertsemlidis A., Cox D., Pennacchio L. A., Boerwinkle E., Cohen J. C., Hobbs H. H.2008. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat. Genet.40: 1461–1465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Yuan X., Waterworth D., Perry J. R., Lim N., Song K., Chambers J. C., Zhang W., Vollenweider P., Stirnadel H., Johnson T., et al. 2008. Population-based genome-wide association studies reveal six loci influencing plasma levels of liver enzymes. Am. J. Hum. Genet.83: 520–528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Pirazzi C., Adiels M., Burza M. A., Mancina R. M., Levin M., Stahlman M., Taskinen M. R., Orho-Melander M., Perman J., Pujia A., et al. 2012. Patatin-like phospholipase domain-containing 3 (PNPLA3) I148M (rs738409) affects hepatic VLDL secretion in humans and in vitro. J. Hepatol.57: 1276–1282. [DOI] [PubMed] [Google Scholar]
  • 31.Valenti L., Alisi A., Nobili V.2012. I148M PNPLA3 variant and progressive liver disease: a new paradigm in hepatology. Hepatology.56: 1883–1889. [PubMed] [Google Scholar]
  • 32.Krawczyk M., Grunhage F., Lammert F.2013. Identification of combined genetic determinants of liver stiffness within the SREBP1c-PNPLA3 pathway. Int. J. Mol. Sci.14: 21153–21166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Romeo S., Sentinelli F., Dash S., Yeo G. S., Savage D. B., Leonetti F., Capoccia D., Incani M., Maglio C., Iacovino M., et al. 2010. Morbid obesity exposes the association between PNPLA3 I148M (rs738409) and indices of hepatic injury in individuals of European descent. Int. J. Obes. (Lond).34: 190–194. [DOI] [PubMed] [Google Scholar]
  • 34.Giudice E. M., Grandone A., Cirillo G., Santoro N., Amato A., Brienza C., Savarese P., Marzuillo P., Perrone L.2011. The association of PNPLA3 variants with liver enzymes in childhood obesity is driven by the interaction with abdominal fat. PLoS ONE.6: e27933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Davis J. N., Le K. A., Walker R. W., Vikman S., Spruijt-Metz D., Weigensberg M. J., Allayee H., Goran M. I.2010. Increased hepatic fat in overweight Hispanic youth influenced by interaction between genetic variation in PNPLA3 and high dietary carbohydrate and sugar consumption. Am. J. Clin. Nutr.92: 1522–1527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Santoro N., Savoye M., Kim G., Marotto K., Shaw M. M., Pierpont B., Caprio S.2012. Hepatic fat accumulation is modulated by the interaction between the rs738409 variant in the PNPLA3 gene and the dietary omega6/omega3 PUFA intake. PLoS ONE.7: e37827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Santoro N., Zhang C. K., Zhao H., Pakstis A. J., Kim G., Kursawe R., Dykas D. J., Bale A. E., Giannini C., Pierpont B., et al. 2012. Variant in the glucokinase regulatory protein (GCKR) gene is associated with fatty liver in obese children and adolescents. Hepatology.55: 781–789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Viganò M., Valenti L., Lampertico P., Facchetti F., Motta B. M., D’Ambrosio R., Romagnoli S., Dongiovanni P., Donati B., Fargion S., et al. 2013. Patatin-like phospholipase domain-containing 3 I148M affects liver steatosis in patients with chronic hepatitis B. Hepatology.58: 1245–1252. [DOI] [PubMed] [Google Scholar]
  • 39.Friedrich K., Wannhoff A., Kattner S., Brune M., Hov J. R., Weiss K. H., Antoni C., Dollinger M., Neumann-Haefelin C., Seufferlein T., et al. 2014. PNPLA3 in end-stage liver disease: alcohol consumption, hepatocellular carcinoma development, and transplantation-free survival. J. Gastroenterol. Hepatol.29: 1477–1484. [DOI] [PubMed] [Google Scholar]
  • 40.Chamoun Z., Vacca F., Parton R. G., Gruenberg J.2013. PNPLA3/adiponutrin functions in lipid droplet formation. Biol. Cell.105: 219–233. [DOI] [PubMed] [Google Scholar]
  • 41.Huang Y., Cohen J. C., Hobbs H. H.2011. Expression and characterization of a PNPLA3 protein isoform (I148M) associated with nonalcoholic fatty liver disease. J. Biol. Chem.286: 37085–37093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kumari M., Schoiswohl G., Chitraju C., Paar M., Cornaciu I., Rangrez A. Y., Wongsiriroj N., Nagy H. M., Ivanova P. T., Scott S. A., et al. 2012. Adiponutrin functions as a nutritionally regulated lysophosphatidic acid acyltransferase. Cell Metab.15: 691–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Villena J. A., Roy S., Sarkadi-Nagy E., Kim K. H., Sul H. S.2004. Desnutrin, an adipocyte gene encoding a novel patatin domain-containing protein, is induced by fasting and glucocorticoids: ectopic expression of desnutrin increases triglyceride hydrolysis. J. Biol. Chem.279: 47066–47075. [DOI] [PubMed] [Google Scholar]
  • 44.Zimmermann R., Strauss J. G., Haemmerle G., Schoiswohl G., Birner-Gruenberger R., Riederer M., Lass A., Neuberger G., Eisenhaber F., Hermetter A., et al. 2004. Fat mobilization in adipose tissue is promoted by adipose triglyceride lipase. Science.306: 1383–1386. [DOI] [PubMed] [Google Scholar]
  • 45.Lass A., Zimmermann R., Haemmerle G., Riederer M., Schoiswohl G., Schweiger M., Kienesberger P., Strauss J. G., Gorkiewicz G., Zechner R.2006. Adipose triglyceride lipase-mediated lipolysis of cellular fat stores is activated by CGI-58 and defective in Chanarin-Dorfman Syndrome. Cell Metab.3: 309–319. [DOI] [PubMed] [Google Scholar]
  • 46.Yamaguchi T., Osumi T.2009. Chanarin-Dorfman syndrome: deficiency in CGI-58, a lipid droplet-bound coactivator of lipase. Biochim. Biophys. Acta.1791: 519–523. [DOI] [PubMed] [Google Scholar]
  • 47.Inoue T., Kobayashi K., Inoguchi T., Sonoda N., Maeda Y., Hirata E., Fujimura Y., Miura D., Hirano K., Takayanagi R.2013. Downregulation of adipose triglyceride lipase in the heart aggravates diabetic cardiomyopathy in db/db mice. Biochem. Biophys. Res. Commun.438: 224–229. [DOI] [PubMed] [Google Scholar]
  • 48.Haemmerle G., Lass A., Zimmermann R., Gorkiewicz G., Meyer C., Rozman J., Heldmaier G., Maier R., Theussl C., Eder S., et al. 2006. Defective lipolysis and altered energy metabolism in mice lacking adipose triglyceride lipase. Science.312: 734–737. [DOI] [PubMed] [Google Scholar]
  • 49.Aflaki E., Doddapattar P., Radovic B., Povoden S., Kolb D., Vujic N., Wegscheider M., Koefeler H., Hornemann T., Graier W. F., et al. 2012. C16 ceramide is crucial for triacylglycerol-induced apoptosis in macrophages. Cell Death Dis.3: e280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Tang T., Abbott M. J., Ahmadian M., Lopes A. B., Wang Y., Sul H. S.2013. Desnutrin/ATGL activates PPARdelta to promote mitochondrial function for insulin secretion in islet beta cells. Cell Metab.18: 883–895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Schoenborn V., Heid I. M., Vollmert C., Lingenhel A., Adams T. D., Hopkins P. N., Illig T., Zimmermann R., Zechner R., Hunt S. C., et al. 2006. The ATGL gene is associated with free fatty acids, triglycerides, and type 2 diabetes. Diabetes.55: 1270–1275. [DOI] [PubMed] [Google Scholar]
  • 52.Kienesberger P. C., Lee D., Pulinilkunnil T., Brenner D. S., Cai L., Magnes C., Koefeler H. C., Streith I. E., Rechberger G. N., Haemmerle G., et al. 2009. Adipose triglyceride lipase deficiency causes tissue-specific changes in insulin signaling. J. Biol. Chem.284: 30218–30229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Peyot M. L., Guay C., Latour M. G., Lamontagne J., Lussier R., Pineda M., Ruderman N. B., Haemmerle G., Zechner R., Joly E., et al. 2009. Adipose triglyceride lipase is implicated in fuel- and non-fuel-stimulated insulin secretion. J. Biol. Chem.284: 16848–16859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Kim J. Y., Tillison K., Lee J. H., Rearick D. A., Smas C. M.2006. The adipose tissue triglyceride lipase ATGL/PNPLA2 is downregulated by insulin and TNF-alpha in 3T3–L1 adipocytes and is a target for transactivation by PPARgamma. Am. J. Physiol. Endocrinol. Metab.291: E115–E127. [DOI] [PubMed] [Google Scholar]
  • 55.Wend K., Wend P., Drew B. G., Hevener A. L., Miranda-Carboni G. A., Krum S. A.2013. ERα regulates lipid metabolism in bone through ATGL and perilipin. J. Cell. Biochem.114: 1306–1314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Grahn T. H., Kaur R., Yin J., Schweiger M., Sharma V. M., Lee M. J., Ido Y., Smas C. M., Zechner R., Lass A., et al. 2014. Fat-specific protein 27 (FSP27) interacts with adipose triglyceride lipase (ATGL) to regulate lipolysis and insulin sensitivity in human adipocytes. J. Biol. Chem.289: 12029–12039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Khan S. A., Sathyanarayan A., Mashek M. T., Ong K. T., Wollaston-Hayden E. E., Mashek D. G.2015. ATGL-catalyzed lipolysis regulates SIRT1 to control PGC-1α/PPAR-α signaling. Diabetes.64: 418–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ong K. T., Mashek M. T., Davidson N. O., Mashek D. G.2014. Hepatic ATGL mediates PPAR-alpha signaling and fatty acid channeling through an L-FABP independent mechanism. J. Lipid Res.55: 808–815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ahmadian M., Abbott M. J., Tang T., Hudak C. S., Kim Y., Bruss M., Hellerstein M. K., Lee H. Y., Samuel V. T., Shulman G. I., et al. 2011. Desnutrin/ATGL is regulated by AMPK and is required for a brown adipose phenotype. Cell Metab.13: 739–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lee W. C., Salido E., Yen P. H.1994. Isolation of a new gene GS2 (DXS1283E) from a CpG island between STS and KAL1 on Xp22.3. Genomics.22: 372–376. [DOI] [PubMed] [Google Scholar]
  • 61.Mancuso D. J., Jenkins C. M., Gross R. W.2000. The genomic organization, complete mRNA sequence, cloning, and expression of a novel human intracellular membrane-associated calcium-independent phospholipase A2. J. Biol. Chem.275: 9937–9945. [DOI] [PubMed] [Google Scholar]
  • 62.Tanaka H., Takeya R., Sumimoto H.2000. A novel intracellular membrane-bound calcium-independent phospholipase A2. Biochem. Biophys. Res. Commun.272: 320–326. [DOI] [PubMed] [Google Scholar]
  • 63.Tanaka H., Minakami R., Kanaya H., Sumimoto H.2004. Catalytic residues of group VIB calcium-independent phospholipase A2 (iPLA2gamma). Biochem. Biophys. Res. Commun.320: 1284–1290. [DOI] [PubMed] [Google Scholar]
  • 64.Mancuso D. J., Jenkins C. M., Sims H. F., Cohen J. M., Yang J., Gross R. W.2004. Complex transcriptional and translational regulation of iPLA2gamma resulting in multiple gene products containing dual competing sites for mitochondrial or peroxisomal localization. Eur. J. Biochem.271: 4709–4724. [DOI] [PubMed] [Google Scholar]
  • 65.Mancuso D. J., Han X., Jenkins C. M., Lehman J. J., Sambandam N., Sims H. F., Yang J., Yan W., Yang K., Green K., et al. 2007. Dramatic accumulation of triglycerides and precipitation of cardiac hemodynamic dysfunction during brief caloric restriction in transgenic myocardium expressing human calcium-independent phospholipase A2gamma. J. Biol. Chem.282: 9216–9227. [DOI] [PubMed] [Google Scholar]
  • 66.Kinsey G. R., Cummings B. S., Beckett C. S., Saavedra G., Zhang W., McHowat J., Schnellmann R. G.2005. Identification and distribution of endoplasmic reticulum iPLA2. Biochem. Biophys. Res. Commun.327: 287–293. [DOI] [PubMed] [Google Scholar]
  • 67.Beckett C. S., McHowat J.2008. Calcium-independent phospholipase A2 in rabbit ventricular myocytes. Lipids.43: 775–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Murakami M., Masuda S., Ueda-Semmyo K., Yoda E., Kuwata H., Takanezawa Y., Aoki J., Arai H., Sumimoto H., Ishikawa Y., et al. 2005. Group VIB Ca2+-independent phospholipase A2gamma promotes cellular membrane hydrolysis and prostaglandin production in a manner distinct from other intracellular phospholipases A2. J. Biol. Chem.280: 14028–14041. [DOI] [PubMed] [Google Scholar]
  • 69.Yan W., Jenkins C. M., Han X., Mancuso D. J., Sims H. F., Yang K., Gross R. W.2005. The highly selective production of 2-arachidonoyl lysophosphatidylcholine catalyzed by purified calcium-independent phospholipase A2gamma: identification of a novel enzymatic mediator for the generation of a key branch point intermediate in eicosanoid signaling. J. Biol. Chem.280: 26669–26679. [DOI] [PubMed] [Google Scholar]
  • 70.Mancuso D. J., Sims H. F., Han X., Jenkins C. M., Guan S. P., Yang K., Moon S. H., Pietka T., Abumrad N. A., Schlesinger P. H., et al. 2007. Genetic ablation of calcium-independent phospholipase A2gamma leads to alterations in mitochondrial lipid metabolism and function resulting in a deficient mitochondrial bioenergetic phenotype. J. Biol. Chem.282: 34611–34622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Song H., Wohltmann M., Bao S., Ladenson J. H., Semenkovich C. F., Turk J.2010. Mice deficient in group VIB phospholipase A2 (iPLA2gamma) exhibit relative resistance to obesity and metabolic abnormalities induced by a Western diet. Am. J. Physiol. Endocrinol. Metab.298: E1097–E1114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Mancuso D. J., Sims H. F., Yang K., Kiebish M. A., Su X., Jenkins C. M., Guan S., Moon S. H., Pietka T., Nassir F., et al. 2010. Genetic ablation of calcium-independent phospholipase A2gamma prevents obesity and insulin resistance during high fat feeding by mitochondrial uncoupling and increased adipocyte fatty acid oxidation. J. Biol. Chem.285: 36495–36510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Mancuso D. J., Kotzbauer P., Wozniak D. F., Sims H. F., Jenkins C. M., Guan S., Han X., Yang K., Sun G., Malik I., et al. 2009. Genetic ablation of calcium-independent phospholipase A2{gamma} leads to alterations in hippocampal cardiolipin content and molecular species distribution, mitochondrial degeneration, autophagy, and cognitive dysfunction. J. Biol. Chem.284: 35632–35644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yoda E., Hachisu K., Taketomi Y., Yoshida K., Nakamura M., Ikeda K., Taguchi R., Nakatani Y., Kuwata H., Murakami M., et al. 2010. Mitochondrial dysfunction and reduced prostaglandin synthesis in skeletal muscle of Group VIB Ca2+-independent phospholipase A2gamma-deficient mice. J. Lipid Res.51: 3003–3015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Kinsey G. R., McHowat J., Beckett C. S., Schnellmann R. G.2007. Identification of calcium-independent phospholipase A2gamma in mitochondria and its role in mitochondrial oxidative stress. Am. J. Physiol. Renal Physiol.292: F853–F860. [DOI] [PubMed] [Google Scholar]
  • 76.Peterson B., Knotts T., Cummings B. S.2007. Involvement of Ca2+-independent phospholipase A2 isoforms in oxidant-induced neural cell death. Neurotoxicology.28: 150–160. [DOI] [PubMed] [Google Scholar]
  • 77.Kinsey G. R., Blum J. L., Covington M. D., Cummings B. S., McHowat J., Schnellmann R. G.2008. Decreased iPLA2gamma expression induces lipid peroxidation and cell death and sensitizes cells to oxidant-induced apoptosis. J. Lipid Res.49: 1477–1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Cummings B. S., Gelasco A. K., Kinsey G. R., McHowat J., Schnellmann R. G.2004. Inactivation of endoplasmic reticulum bound Ca2+-independent phospholipase A2 in renal cells during oxidative stress. J. Am. Soc. Nephrol.15: 1441–1451. [DOI] [PubMed] [Google Scholar]
  • 79.Bao S., Song H., Tan M., Wohltmann M., Ladenson J. H., Turk J.2012. Group VIB phospholipase A2 promotes proliferation of INS-1 insulinoma cells and attenuates lipid peroxidation and apoptosis induced by inflammatory cytokines and oxidant agents. Oxid. Med. Cell. Longev.2012: 989372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Elimam H., Papillon J., Takano T., Cybulsky A. V.2015. Calcium-independent phospholipase A2gamma enhances activation of the ATF6 transcription factor during endoplasmic reticulum stress. J. Biol. Chem.290: 3009–3020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Moon S. H., Jenkins C. M., Kiebish M. A., Sims H. F., Mancuso D. J., Gross R. W.2012. Genetic ablation of calcium-independent phospholipase A2gamma (iPLA2gamma) attenuates calcium-induced opening of the mitochondrial permeability transition pore and resultant cytochrome c release. J. Biol. Chem.287: 29837–29850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Brustovetsky T., Antonsson B., Jemmerson R., Dubinsky J. M., Brustovetsky N.2005. Activation of calcium-independent phospholipase A2 (iPLA2) in brain mitochondria and release of apoptogenic factors by BAX and truncated BID. J. Neurochem.94: 980–994. [DOI] [PubMed] [Google Scholar]
  • 83.Sharma J., Eickhoff C. S., Hoft D. F., Ford D. A., Gross R. W., McHowat J.2013. The absence of myocardial calcium-independent phospholipase A2gamma results in impaired prostaglandin E2 production and decreased survival in mice with acute Trypanosoma cruzi infection. Infect. Immun.81: 2278–2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Yoda E., Rai K., Ogawa M., Takakura Y., Kuwata H., Suzuki H., Nakatani Y., Murakami M., Hara S.2014. Group VIB calcium-independent phospholipase A2 (iPLA2gamma) regulates platelet activation, hemostasis and thrombosis in mice. PLoS ONE.9: e109409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Saunders C. J., Moon S. H., Liu X., Thiffault I., Coffman K., LePichon J. B., Taboada E., Smith L. D., Farrow E. G., Miller N., et al. 2015. Loss of function variants in human PNPLA8 encoding calcium-independent phospholipase A2 gamma recapitulate the mitochondriopathy of the homologous null mouse. Hum. Mutat.36: 301–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Balboa M. A., Balsinde J., Jones S. S., Dennis E. A.1997. Identity between the Ca2+-independent phospholipase A2 enzymes from P388D1 macrophages and Chinese hamster ovary cells. J. Biol. Chem.272: 8576–8580. [DOI] [PubMed] [Google Scholar]
  • 87.Tang J., Kriz R. W., Wolfman N., Shaffer M., Seehra J., Jones S. S.1997. A novel cytosolic calcium-independent phospholipase A2 contains eight ankyrin motifs. J. Biol. Chem.272: 8567–8575. [DOI] [PubMed] [Google Scholar]
  • 88.Jones S. S., Tang J., Kriz R., Shaffer M., Knopf J., Seehra J.1996. Isolation, molecular cloning and expression of a novel calcium-independent phospholipase A2FASEB J.10: A977. [Google Scholar]
  • 89.Ghosh M., Tucker D. E., Burchett S. A., Leslie C. C.2006. Properties of the group IV phospholipase A2 family. Prog. Lipid Res.45: 487–510. [DOI] [PubMed] [Google Scholar]
  • 90.Wolf M. J., Gross R. W.1996. Expression, purification, and kinetic characterization of a recombinant 80-kDa intracellular calcium-independent phospholipase A2. J. Biol. Chem.271: 30879–30885. [DOI] [PubMed] [Google Scholar]
  • 91.Lio Y. C., Dennis E. A.1998. Interfacial activation, lysophospholipase and transacylase activity of group VI Ca2+-independent phospholipase A2. Biochim. Biophys. Acta.1392: 320–332. [DOI] [PubMed] [Google Scholar]
  • 92.Jenkins C. M., Yan W., Mancuso D. J., Gross R. W.2006. Highly selective hydrolysis of fatty acyl-CoAs by calcium-independent phospholipase A2beta. Enzyme autoacylation and acyl-CoA-mediated reversal of calmodulin inhibition of phospholipase A2 activity. J. Biol. Chem.281: 15615–15624. [DOI] [PubMed] [Google Scholar]
  • 93.Carper M. J., Zhang S., Turk J., Ramanadham S.2008. Skeletal muscle group VIA phospholipase A2 (iPLA2β): expression and role in fatty acid oxidation. Biochemistry.47: 12241–12249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Ma Z., Ramanadham S., Kempe K., Chi X. S., Ladenson J., Turk J.1997. Pancreatic islets express a Ca2+-independent phospholipase A2 enzyme that contains a repeated structural motif homologous to the integral membrane protein binding domain of ankyrin. J. Biol. Chem.272: 11118–11127. [PubMed] [Google Scholar]
  • 95.Larsson P. K. A., Claesson H-E., Kennedy B. P.1998. Multiple splice variants of the human calcium-independent phospholipase A2 and their effect on enzyme activity. J. Biol. Chem.273: 207–214. [DOI] [PubMed] [Google Scholar]
  • 96.Ma Z., Wang X., Nowatzke W., Ramanadham S., Turk J.1999. Human pancreatic islets express mRNA species encoding two distinct catalytically active isoforms of group VI phospholipase A2 (iPLA2) that arise from an exon-skipping mechanism of alternative splicing of the transcript from the iPLA2 gene on chromosome 22q13.1. J. Biol. Chem.274: 9607–9616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Atsumi G., Tajima M., Hadano A., Nakatani Y., Murakami M., Kudo I.1998. Fas-induced arachidonic acid release is mediated by Ca2+-independent phospholipase A2 but not cytosolic phospholipase A2, which undergoes proteolytic inactivation. J. Biol. Chem.273: 13870–13877. [DOI] [PubMed] [Google Scholar]
  • 98.Ramanadham S., Hsu F. F., Zhang S., Jin C., Bohrer A., Song H., Bao S., Ma Z., Turk J.2004. Apoptosis of insulin-secreting cells induced by endoplasmic reticulum stress is amplified by overexpression of group VIA calcium-independent phospholipase A2 (iPLA2β) and suppressed by inhibition of iPLA2β. Biochemistry.43: 918–930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ramanadham S., Song H., Hsu F. F., Zhang S., Crankshaw M., Grant G. A., Newgard C. B., Bao S., Ma Z., Turk J.2003. Pancreatic islets and insulinoma cells express a novel isoform of group VIA phospholipase A2 (iPLA2β) that participates in glucose-stimulated insulin secretion and is not produced by alternate splicing of the iPLA2β transcript. Biochemistry.42: 13929–13940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Song H., Bao S., Lei X., Jin C., Zhang S., Turk J., Ramanadham S.2010. Evidence for proteolytic processing and stimulated organelle redistribution of iPLA2β. Biochim. Biophys. Acta.1801: 547–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Song H., Hecimovic S., Goate A., Hsu F. F., Bao S., Vidavsky I., Ramanadham S., Turk J.2004. Characterization of N-terminal processing of group VIA phospholipase A2 and of potential cleavage sites of amyloid precursor protein constructs by automated identification of signature peptides in LC/MS/MS analyses of proteolytic digests. J. Am. Soc. Mass Spectrom.15: 1780–1793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Gross R. W., Ramanadham S., Kruszka K. K., Han X., Turk J.1993. Rat and human pancreatic islet cells contain a calcium ion independent phospholipase A2 activity selective for hydrolysis of arachidonate which is stimulated by adenosine triphosphate and is specifically localized to islet beta-cells. Biochemistry.32: 327–336. [DOI] [PubMed] [Google Scholar]
  • 103.Lei X., Zhang S., Bohrer A., Bao S., Song H., Ramanadham S.2007. The group VIA calcium-independent phospholipase A2 participates in ER stress-induced INS-1 insulinoma cell apoptosis by promoting ceramide generation via hydrolysis of sphingomyelins by neutral sphingomyelinase. Biochemistry.46: 10170–10185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Lei X., Zhang S., Bohrer A., Ramanadham S.2008. Calcium-independent phospholipase A2 (iPLA2β)-mediated ceramide generation plays a key role in the cross-talk between the endoplasmic reticulum (ER) and mitochondria during ER stress-induced insulin-secreting cell apoptosis. J. Biol. Chem.283: 34819–34832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Ma Z., Ramanadham S., Wohltmann M., Bohrer A., Hsu F. F., Turk J.2001. Studies of insulin secretory responses and of arachidonic acid incorporation into phospholipids of stably transfected insulinoma cells that overexpress group VIA phospholipase A2 (iPLA2β) indicate a signaling rather than a housekeeping role for iPLA2β. J. Biol. Chem.276: 13198–13208. [DOI] [PubMed] [Google Scholar]
  • 106.Turk J., Ramanadham S.2004. The expression and function of a group VIA calcium-independent phospholipase A2 (iPLA2β) in beta-cells. Can. J. Physiol. Pharmacol.82: 824–832. [DOI] [PubMed] [Google Scholar]
  • 107.Ma Z., Zhang S., Turk J., Ramanadham S.2002. Stimulation of insulin secretion and associated nuclear accumulation of iPLA2β in INS-1 insulinoma cells. Am. J. Physiol. Endocrinol. Metab.282: E820–E833. [DOI] [PubMed] [Google Scholar]
  • 108.Shinzawa K., Tsujimoto Y.2003. PLA2 activity is required for nuclear shrinkage in caspase-independent cell death. J. Cell Biol.163: 1219–1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Bao S., Jin C., Zhang S., Turk J., Ma Z., Ramanadham S.2004. Beta-cell calcium-independent group VIA phospholipase A2 (iPLA2β): tracking iPLA2β movements in response to stimulation with insulin secretagogues in INS-1 cells. Diabetes.53(Suppl 1): S186–S189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Ramanadham S., Song H., Bao S., Hsu F-F., Zhang S., Ma Z., Jin C., Turk J.2004. Islet complex lipids: Involvement in the actions of group VIA calcium-independent phospholipase A2 in β-cells. Diabetes.53: S179–S185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ramanadham S., Wolf M. J., Ma Z., Li B., Wang J., Gross R. W., Turk J.1996. Evidence for association of an ATP-stimulatable Ca2+-independent phospholipase A2 from pancreatic islets and HIT insulinoma cells with a phosphofructokinase-like protein. Biochemistry.35: 5464–5471. [DOI] [PubMed] [Google Scholar]
  • 112.Bennett V.1992. Ankyrins. Adaptors between diverse plasma membrane proteins and the cytoplasm. J. Biol. Chem.267: 8703–8706. [PubMed] [Google Scholar]
  • 113.de Caestecker M. P., Hemmati P., Larisch-Bloch S., Ajmera R., Roberts A. B., Lechleider R. J.1997. Characterization of functional domains within Smad4/DPC4. J. Biol. Chem.272: 13690–13696. [DOI] [PubMed] [Google Scholar]
  • 114.Song H., Bao S., Ramanadham S., Turk J.2006. Effects of biological oxidants on the catalytic activity and structure of group VIA phospholipase A2. Biochemistry.45: 6392–6406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.McHowat J., Swift L. M., Arutunyan A., Sarvazyan N.2001. Clinical concentrations of doxorubicin inhibit activity of myocardial membrane-associated, calcium-independent phospholipase A2. Cancer Res.61: 4024–4029. [PubMed] [Google Scholar]
  • 116.Ma Z., Turk J.2001. The molecular biology of the group VIA Ca2+-independent phospholipase A2. Prog. Nucleic Acid Res. Mol. Biol.67: 1–33. [DOI] [PubMed] [Google Scholar]
  • 117.Hazen S. L., Gross R. W.1991. ATP-dependent regulation of rabbit myocardial cytosolic calcium- independent phospholipase A2. J. Biol. Chem.266: 14526–14534. [PubMed] [Google Scholar]
  • 118.Hazen S. L., Ford D. A., Gross R. W.1991. Activation of a membrane-associated phospholipase A2 during rabbit myocardial ischemia which is highly selective for plasmalogen substrate. J. Biol. Chem.266: 5629–5633. [PubMed] [Google Scholar]
  • 119.Jenkins C. M., Wolf M. J., Mancuso D. J., Gross R. W.2001. Identification of the calmodulin-binding domain of recombinant calcium-independent phospholipase A2beta. Implications for structure and function. J. Biol. Chem.276: 7129–7135. [DOI] [PubMed] [Google Scholar]
  • 120.Wang Z., Ramanadham S., Ma Z. A., Bao S., Mancuso D. J., Gross R. W., Turk J.2005. Group VIA phospholipase A2 forms a signaling complex with the calcium/calmodulin-dependent protein kinase IIbeta expressed in pancreatic islet beta-cells. J. Biol. Chem.280: 6840–6849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Wolf M. J., Wang J., Turk J., Gross R. W.1997. Depletion of intracellular calcium stores activates smooth muscle cell calcium-independent phospholipase A2. A novel mechanism underlying arachidonic acid mobilization. J. Biol. Chem.272: 1522–1526. [DOI] [PubMed] [Google Scholar]
  • 122.Nowatzke W., Ramanadham S., Ma Z., Hsu F. F., Bohrer A., Turk J.1998. Mass spectrometric evidence that agents that cause loss of Ca2+ from intracellular compartments induce hydrolysis of arachidonic acid from pancreatic islet membrane phospholipids by a mechanism that does not require a rise in cytosolic Ca2+ concentration. Endocrinology.139: 4073–4085. [DOI] [PubMed] [Google Scholar]
  • 123.Larsson Forsell P. K., Runarsson G., Ibrahim M., Bjorkholm M., Claesson H. E.1998. On the expression of cytosolic calcium-independent phospholipase A2 (88 kDa) in immature and mature myeloid cells and its role in leukotriene synthesis in human granulocytes. FEBS Lett.434: 295–299. [DOI] [PubMed] [Google Scholar]
  • 124.Rzigalinski B. A., Blackmore P. F., Rosenthal M. D.1996. Arachidonate mobilization is coupled to depletion of intracellular calcium stores and influx of extracellular calcium in differentiated U937 cells. Biochim. Biophys. Acta.1299: 342–352. [DOI] [PubMed] [Google Scholar]
  • 125.Atsumi G., Murakami M., Kojima K., Hadano A., Tajima M., Kudo I.2000. Distinct roles of two intracellular phospholipase A2s in fatty acid release in the cell death pathway. Proteolytic fragment of type IVA cytosolic phospholipase A2alpha inhibits stimulus-induced arachidonate release, whereas that of type VI Ca2+-independent phospholipase A2 augments spontaneous fatty acid release. J. Biol. Chem.275: 18248–18258. [DOI] [PubMed] [Google Scholar]
  • 126.Seashols S. J., del Castillo Olivares A., Gil G., Barbour S. E.2004. Regulation of group VIA phospholipase A2 expression by sterol availability. Biochim. Biophys. Acta.1684: 29–37. [DOI] [PubMed] [Google Scholar]
  • 127.Akarte A. S., Srinivasan B. P., Gandhi S.2012. Vildagliptin selectively ameliorates GLP-1, GLUT4, SREBP-1c mRNA levels and stimulates β-cell proliferation resulting in improved glucose homeostasis in rats with streptozotocin-induced diabetes. J. Diabetes Complications.26: 266–274. [DOI] [PubMed] [Google Scholar]
  • 128.Boslem E., MacIntosh G., Preston A. M., Bartley C., Busch A. K., Fuller M., Laybutt D. R., Meikle P. J., Biden T. J.2011. A lipidomic screen of palmitate-treated MIN6 beta-cells links sphingolipid metabolites with endoplasmic reticulum (ER) stress and impaired protein trafficking. Biochem. J.435: 267–276. [DOI] [PubMed] [Google Scholar]
  • 129.Chan J. Y., Cooney G. J., Biden T. J., Laybutt D. R.2011. Differential regulation of adaptive and apoptotic unfolded protein response signalling by cytokine-induced nitric oxide production in mouse pancreatic beta cells. Diabetologia.54: 1766–1776. [DOI] [PubMed] [Google Scholar]
  • 130.Chin H. J., Fu Y. Y., Ahn J. M., Na K. Y., Kim Y. S., Kim S., Chae D. W.2010. Omacor, n-3 polyunsaturated fatty acid, attenuated albuminuria and renal dysfunction with decrease of SREBP-1 expression and triglyceride amount in the kidney of type II diabetic animals. Nephrol. Dial. Transplant.25: 1450–1457. [DOI] [PubMed] [Google Scholar]
  • 131.Heller J. J., Qiu J., Zhou L.2011. Nuclear receptors take center stage in Th17 cell-mediated autoimmunity. J. Clin. Invest.121: 519–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Kaplan M., Aviram M., Hayek T.2012. Oxidative stress and macrophage foam cell formation during diabetes mellitus-induced atherogenesis: Role of insulin therapy. Pharmacol. Ther.136: 175–185. [DOI] [PubMed] [Google Scholar]
  • 133.Véret J., Coant N., Berdyshev E. V., Skobeleva A., Therville N., Bailbe D., Gorshkova I., Natarajan V., Portha B., Le Stunff H.2011. Ceramide synthase 4 and de novo production of ceramides with specific N-acyl chain lengths are involved in glucolipotoxicity-induced apoptosis of INS-1 β-cells. Biochem. J.438: 177–189. [DOI] [PubMed] [Google Scholar]
  • 134.Wang H., Kouri G., Wollheim C. B.2005. ER stress and SREBP-1 activation are implicated in beta-cell glucolipotoxicity. J. Cell Sci.118: 3905–3915. [DOI] [PubMed] [Google Scholar]
  • 135.Yano M., Watanabe K., Yamamoto T., Ikeda K., Senokuchi T., Lu M., Kadomatsu T., Tsukano H., Ikawa M., Okabe M., et al. 2011. Mitochondrial dysfunction and increased reactive oxygen species impair insulin secretion in sphingomyelin synthase 1-null mice. J. Biol. Chem.286: 3992–4002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Lei X., Bone R. N., Ali T., Zhang S., Bohrer A., Tse H. M., Bidasee K. R., Ramanadham S.2014. Evidence of contribution of iPLA2β-mediated events during islet beta-cell apoptosis due to proinflammatory cytokines suggests a role for iPLA2β in T1D development. Endocrinology.155: 3352–3364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Lei X., Zhang S., Barbour S. E., Bohrer A., Ford E. L., Koizumi A., Papa F. R., Ramanadham S.2010. Spontaneous development of endoplasmic reticulum stress that can lead to diabetes mellitus is associated with higher calcium-independent phospholipase A2 expression: a role for regulation by SREBP-1. J. Biol. Chem.285: 6693–6705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Lei X., Zhang S., Bohrer A., Barbour S. E., Ramanadham S.2012. Role of calcium-independent phospholipase A2beta in human pancreatic islet beta-cell apoptosis. Am. J. Physiol. Endocrinol. Metab.303: E1386–E1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Ackermann E. J., Conde-Frieboes K., Dennis E. A.1995. Inhibition of macrophage Ca2+-independent phospholipase A2 by bromoenol lactone and trifluoromethyl ketones. J. Biol. Chem.270: 445–450. [DOI] [PubMed] [Google Scholar]
  • 140.Magrioti V., Kokotos G.2010. Phospholipase A2 inhibitors as potential therapeutic agents for the treatment of inflammatory diseases. Expert Opin. Ther. Pat.20: 1–18. [DOI] [PubMed] [Google Scholar]
  • 141.Schaeffer E. L., Gattaz W. F.2005. Inhibition of calcium-independent phospholipase A2 activity in rat hippocampus impairs acquisition of short- and long-term memory. Psychopharmacology (Berl.).181: 392–400. [DOI] [PubMed] [Google Scholar]
  • 142.Bao S., Jacobson D. A., Wohltmann M., Bohrer A., Jin W., Philipson L. H., Turk J.2008. Glucose homeostasis, insulin secretion, and islet phospholipids in mice that overexpress iPLA2β in pancreatic β-cells and in iPLA2β-null mice. Am. J. Physiol. Endocrinol. Metab.294: E217–E229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Bao S., Miller D. J., Ma Z., Wohltmann M., Eng G., Ramanadham S., Moley K., Turk J.2004. Male mice that do not express group VIA phospholipase A2 produce spermatozoa with impaired motility and have greatly reduced fertility. J. Biol. Chem.279: 38194–38200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Bao S., Song H., Wohltmann M., Ramanadham S., Jin W., Bohrer A., Turk J.2006. Insulin secretory responses and phospholipid composition of pancreatic islets from mice that do not express group VIA phospholipase A2 and effects of metabolic stress on glucose homeostasis. J. Biol. Chem.281: 20958–20973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Lei X., Barbour S. E., Ramanadham S.2010. Group VIA Ca2+-independent phospholipase A2 (iPLA2β) and its role in beta-cell programmed cell death. Biochimie.92: 627–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Hazen S. L., Zupan L. A., Weiss R. H., Getman D. P., Gross R. W.1991. Suicide inhibition of canine myocardial cytosolic calcium-independent phospholipase A2. Mechanism-based discrimination between calcium- dependent and -independent phospholipases A2. J. Biol. Chem.266: 7227–7232. [PubMed] [Google Scholar]
  • 147.Ma Z., Ramanadham S., Hu Z., Turk J.1998. Cloning and expression of a group IV cytosolic Ca2+-dependent phospholipase A2 from rat pancreatic islets. Comparison of the expressed activity with that of an islet group VI cytosolic Ca2+-independent phospholipase A2. Biochim. Biophys. Acta.1391: 384–400. [DOI] [PubMed] [Google Scholar]
  • 148.Jenkins C. M., Han X., Mancuso D. J., Gross R. W.2002. Identification of calcium-independent phospholipase A2 (iPLA2)beta, and not iPLA2gamma, as the mediator of arginine vasopressin-induced arachidonic acid release in A-10 smooth muscle cells. Enantioselective mechanism-based discrimination of mammalian iPLA2s. J. Biol. Chem.277: 32807–32814. [DOI] [PubMed] [Google Scholar]
  • 149.Song H., Ramanadham S., Bao S., Hsu F. F., Turk J.2006. A bromoenol lactone suicide substrate inactivates group VIA phospholipase A2 by generating a diffusible bromomethyl keto acid that alkylates cysteine thiols. Biochemistry.45: 1061–1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Song H., Rohrs H., Tan M., Wohltmann M., Ladenson J. H., Turk J.2010. Effects of endoplasmic reticulum stress on group VIA phospholipase A2 in beta cells include tyrosine phosphorylation and increased association with calnexin. J. Biol. Chem.285: 33843–33857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Jenkins C. M., Yang J., Gross R. W.2013. Mechanism-based inhibition of iPLA2β demonstrates a highly reactive cysteine residue (C651) that interacts with the active site: mass spectrometric elucidation of the mechanisms underlying inhibition. Biochemistry.52: 4250–4263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Fuentes L., Perez R., Nieto M. L., Balsinde J., Balboa M. A.2003. Bromoenol lactone promotes cell death by a mechanism involving phosphatidate phosphohydrolase-1 rather than calcium-independent phospholipase A2. J. Biol. Chem.278: 44683–44690. [DOI] [PubMed] [Google Scholar]
  • 153.Wilkins W. P., 3rd, Barbour S. E.2008. Group VI phospholipases A2: homeostatic phospholipases with significant potential as targets for novel therapeutics. Curr. Drug Targets.9: 683–697. [DOI] [PubMed] [Google Scholar]
  • 154.Balsinde J., Dennis E. A.1996. Bromoenol lactone inhibits magnesium-dependent phosphatidate phosphohydrolase and blocks triacylglycerol biosynthesis in mouse P388D1 macrophages. J. Biol. Chem.271: 31937–31941. [DOI] [PubMed] [Google Scholar]
  • 155.Daniels S. B., Cooney E., Sofia M. J., Chakravarty P. K., Katzenellenbogen J. A.1983. Haloenol lactones. Potent enzyme-activated irreversible inhibitors for alpha-chymotrypsin. J. Biol. Chem.258: 15046–15053. [PubMed] [Google Scholar]
  • 156.Ali T., Kokotos G., Magrioti V., Bone R. N., Mobley J. A., Hancock W., Ramanadham S.2013. Characterization of FKGK18 as inhibitor of group VIA Ca2+-independent phospholipase A2 (iPLA2β): candidate drug for preventing beta-cell apoptosis and diabetes. PLoS ONE.8: e71748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Kokotos G., Hsu Y. H., Burke J. E., Baskakis C., Kokotos C. G., Magrioti V., Dennis E. A.2010. Potent and selective fluoroketone inhibitors of group VIA calcium-independent phospholipase A2. J. Med. Chem.53: 3602–3610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Kalyvas A., Baskakis C., Magrioti V., Constantinou-Kokotou V., Stephens D., Lopez-Vales R., Lu J. Q., Yong V. W., Dennis E. A., Kokotos G., et al. 2009. Differing roles for members of the phospholipase A2 superfamily in experimental autoimmune encephalomyelitis. Brain.132: 1221–1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Stephens D., Barbayianni E., Constantinou-Kokotou V., Peristeraki A., Six D. A., Cooper J., Harkewicz R., Deems R. A., Dennis E. A., Kokotos G.2006. Differential inhibition of group IVA and group VIA phospholipases A2 by 2-oxoamides. J. Med. Chem.49: 2821–2828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Gil-de-Gómez L., Astudillo A. M., Guijas C., Magrioti V., Kokotos G., Balboa M. A., Balsinde J.2014. Cytosolic group IVA and calcium-independent group VIA phospholipase A2s act on distinct phospholipid pools in zymosan-stimulated mouse peritoneal macrophages. J. Immunol.192: 752–762. [DOI] [PubMed] [Google Scholar]
  • 161.Bone R. N., Gai Y., Magrioti V., Kokotou M. G., Ali T., Lei X., Tse H. M., Kokotos G., Ramanadham S.2015. Inhibition of Ca2+-independent phospholipase A2beta (iPLA2β) ameliorates islet infiltration and incidence of diabetes in NOD mice. Diabetes.64: 541–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Magrioti V., Nikolaou A., Smyrniotou A., Shah I., Constantinou-Kokotou V., Dennis E. A., Kokotos G.2013. New potent and selective polyfluoroalkyl ketone inhibitors of GVIA calcium-independent phospholipase A2. Bioorg. Med. Chem.21: 5823–5829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Hsu Y. H., Bucher D., Cao J., Li S., Yang S. W., Kokotos G., Woods V. L., Jr, McCammon J. A., Dennis E. A.2013. Fluoroketone inhibition of Ca2+-independent phospholipase A2 through binding pocket association defined by hydrogen/deuterium exchange and molecular dynamics. J. Am. Chem. Soc.135: 1330–1337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Barbayianni E., Stephens D., Grkovich A., Magrioti V., Hsu Y. H., Dolatzas P., Kalogiannidis D., Dennis E. A., Kokotos G.2009. 2-Oxoamide inhibitors of phospholipase A2 activity and cellular arachidonate release based on dipeptides and pseudodipeptides. Bioorg. Med. Chem.17: 4833–4843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Balsinde J., Balboa M. A., Dennis E. A.1997. Antisense inhibition of group VI Ca2+-independent phospholipase A2 blocks phospholipid fatty acid remodeling in murine P388D1 macrophages. J. Biol. Chem.272: 29317–29321. [DOI] [PubMed] [Google Scholar]
  • 166.Balsinde J., Bianco I. D., Ackermann E. J., Conde-Frieboes K., Dennis E. A.1995. Inhibition of calcium-independent phospholipase A2 prevents arachidonic acid incorporation and phospholipid remodeling in P388D1 macrophages. Proc. Natl. Acad. Sci. USA.92: 8527–8531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Chilton F. H., Fonteh A. N., Surette M. E., Triggiani M., Winkler J. D.1996. Control of arachidonate levels within inflammatory cells. Biochim. Biophys. Acta.1299: 1–15. [DOI] [PubMed] [Google Scholar]
  • 168.Dennis E. A.1992. The biosynthesis of phospholipids. Methods Enzymol.209: 1–4. [DOI] [PubMed] [Google Scholar]
  • 169.Barbour S. E., Kapur A., Deal C. L.1999. Regulation of phosphatidylcholine homeostasis by calcium-independent phospholipase A2. Biochim. Biophys. Acta.1439: 77–88. [DOI] [PubMed] [Google Scholar]
  • 170.Wolf B. A., Pasquale S. M., Turk J.1991. Free fatty acid accumulation in secretagogue-stimulated pancreatic islets and effects of arachidonate on depolarization-induced insulin secretion. Biochemistry.30: 6372–6379. [DOI] [PubMed] [Google Scholar]
  • 171.Song H., Wohltmann M., Tan M., Ladenson J. H., Turk J.2014. Group VIA phospholipase A2 mitigates palmitate-induced beta-cell mitochondrial injury and apoptosis. J. Biol. Chem.289: 14194–14210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Balboa M. A., Sáez Y., Balsinde J.2003. Calcium-independent phospholipase A2 is required for lysozyme secretion in U937 promonocytes. J. Immunol.170: 5276–5280. [DOI] [PubMed] [Google Scholar]
  • 173.Song Y., Wilkins P., Hu W., Murthy K. S., Chen J., Lee Z., Oyesanya R., Wu J., Barbour S. E., Fang X.2007. Inhibition of calcium-independent phospholipase A2 suppresses proliferation and tumorigenicity of ovarian carcinoma cells. Biochem. J.406: 427–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Sanchez T., Moreno J. J.2002. Calcium-independent phospholipase A2 through arachidonic acid mobilization is involved in Caco-2 cell growth. J. Cell. Physiol.193: 293–298. [DOI] [PubMed] [Google Scholar]
  • 175.Herbert S. P., Walker J. H.2006. Group VIA calcium-independent phospholipase A2 mediates endothelial cell S phase progression. J. Biol. Chem.281: 35709–35716. [DOI] [PubMed] [Google Scholar]
  • 176.Bao S., Bohrer A., Ramanadham S., Jin W., Zhang S., Turk J.2006. Effects of stable suppression of group VIA phospholipase A2 expression on phospholipid content and composition, insulin secretion, and proliferation of INS-1 insulinoma cells. J. Biol. Chem.281: 187–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Ma Z., Bohrer A., Wohltmann M., Ramanadham S., Hsu F-F., Turk J.2001. Studies of phospholipid metabolism, proliferation, and secretion of stably transfected insulinoma cells that overexpress group VIA phospholipase A2. Lipids.36: 689–700. [DOI] [PubMed] [Google Scholar]
  • 178.Moon S. H., Jenkins C. M., Mancuso D. J., Turk J., Gross R. W.2008. Smooth muscle cell arachidonic acid release, migration, and proliferation are markedly attenuated in mice null for calcium-independent phospholipase A2beta. J. Biol. Chem.283: 33975–33987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Berti-Mattera L. N., Harwalkar S., Hughes B., Wilkins P. L., Almhanna K.2001. Proliferative and morphological effects of endothelins in Schwann cells: roles of p38 mitogen-activated protein kinase and Ca2+-independent phospholipase A2. J. Neurochem.79: 1136–1148. [DOI] [PubMed] [Google Scholar]
  • 180.Zhang X. H., Zhao C., Seleznev K., Song K., Manfredi J. J., Ma Z. A.2006. Disruption of G1-phase phospholipid turnover by inhibition of Ca2+-independent phospholipase A2 induces a p53-dependent cell-cycle arrest in G1 phase. J. Cell Sci.119: 1005–1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Sanchez T., Moreno J. J.2001. The effect of high molecular phospholipase A2 inhibitors on 3T6 fibroblast proliferation. Biochem. Pharmacol.61: 811–816. [DOI] [PubMed] [Google Scholar]
  • 182.Sengupta S., Xiao Y. J., Xu Y.2003. A novel laminin-induced LPA autocrine loop in the migration of ovarian cancer cells. FASEB J.17: 1570–1572. [DOI] [PubMed] [Google Scholar]
  • 183.Turk J., Gross R. W., Ramanadham S.1993. Amplification of insulin secretion by lipid messengers. Diabetes.42: 367–374. [DOI] [PubMed] [Google Scholar]
  • 184.Traianedes K., Dallas M. R., Garrett I. R., Mundy G. R., Bonewald L. F.1998. 5-Lipoxygenase metabolites inhibit bone formation in vitro. Endocrinology.139: 3178–3184. [DOI] [PubMed] [Google Scholar]
  • 185.Akamine T., Jee W. S., Ke H. Z., Li X. J., Lin B. Y.1992. Prostaglandin E2 prevents bone loss and adds extra bone to immobilized distal femoral metaphysis in female rats. Bone.13: 11–22. [DOI] [PubMed] [Google Scholar]
  • 186.Weinreb M., Shamir D., Machwate M., Rodan G. A., Harada S., Keila S.2006. Prostaglandin E2 (PGE2) increases the num­ber of rat bone marrow osteogenic stromal cells (BMSC) via binding the EP4 receptor, activating sphingosine kinase and inhibiting caspase activity. Prostaglandins Leukot. Essent. Fatty Acids.75: 81–90. [DOI] [PubMed] [Google Scholar]
  • 187.Weinreb M., Suponitzky I., Keila S.1997. Systemic administration of an anabolic dose of PGE2 in young rats increases the osteogenic capacity of bone marrow. Bone.20: 521–526. [DOI] [PubMed] [Google Scholar]
  • 188.Yang R. S., Liu T. K., Lin-Shiau S. Y.1993. Increased bone growth by local prostaglandin E2 in rats. Calcif. Tissue Int.52: 57–61. [DOI] [PubMed] [Google Scholar]
  • 189.Miyaura C., Inada M., Matsumoto C., Ohshiba T., Uozumi N., Shimizu T., Ito A.2003. An essential role of cytosolic phospholipase A2alpha in prostaglandin E2-mediated bone resorption associated with inflammation. J. Exp. Med.197: 1303–1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Weiler H. A.2000. Dietary supplementation of arachidonic acid is associated with higher whole body weight and bone mineral density in growing pigs. Pediatr. Res.47: 692–697. [DOI] [PubMed] [Google Scholar]
  • 191.Sato Y., Arai N., Negishi A., Ohya K.1997. Expression of cyclooxygenase genes and involvement of endogenous prostaglandin during osteogenesis in the rat tibial bone marrow cavity. J. Med. Dent. Sci.44: 81–92. [PubMed] [Google Scholar]
  • 192.Ramanadham S., Yarasheski K. E., Silva M. J., Wohltmann M., Novack D. V., Christiansen B., Tu X., Zhang S., Lei X., Turk J.2008. Age-related changes in bone morphology are accelerated in group VIA phospholipase A2 (iPLA2β)-null mice. Am. J. Pathol.172: 868–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Sato H., Taketomi Y., Isogai Y., Miki Y., Yamamoto K., Masuda S., Hosono T., Arata S., Ishikawa Y., Ishii T., et al. 2010. Group III secreted phospholipase A2 regulates epididymal sperm maturation and fertility in mice. J. Clin. Invest.120: 1400–1414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Escoffier J., Jemel I., Tanemoto A., Taketomi Y., Payre C., Coatrieux C., Sato H., Yamamoto K., Masuda S., Pernet-Gallay K., et al. 2010. Group X phospholipase A2 is released during sperm acrosome reaction and controls fertility outcome in mice. J. Clin. Invest.120: 1415–1428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Bonventre J. V., Huang Z., Taheri M. R., O’Leary E., Li E., Moskowitz M. A., Sapirstein A.1997. Reduced fertility and postischaemic brain injury in mice deficient in cytosolic phospholipase A2. Nature.390: 622–625. [DOI] [PubMed] [Google Scholar]
  • 196.Achache H., Tsafrir A., Prus D., Reich R., Revel A.2010. Defective endometrial prostaglandin synthesis identified in patients with repeated implantation failure undergoing in vitro fertilization. Fertil. Steril.94: 1271–1278. [DOI] [PubMed] [Google Scholar]
  • 197.Ho H. C., Suarez S. S.2001. An inositol 1,4,5-trisphosphate receptor-gated intracellular Ca2+ store is involved in regulating sperm hyperactivated motility. Biol. Reprod.65: 1606–1615. [DOI] [PubMed] [Google Scholar]
  • 198.Marquez B., Suarez S. S.2004. Different signaling pathways in bovine sperm regulate capacitation and hyperactivation. Biol. Reprod.70: 1626–1633. [DOI] [PubMed] [Google Scholar]
  • 199.Yuan Y. Y., Chen W. Y., Shi Q. X., Mao L. Z., Yu S. Q., Fang X., Roldan E. R.2003. Zona pellucida induces activation of phospholipase A2 during acrosomal exocytosis in guinea pig spermatozoa. Biol. Reprod.68: 904–913. [DOI] [PubMed] [Google Scholar]
  • 200.Wu C., Stojanov T., Chami O., Ishii S., Shimizu T., Li A., O’Neill C.2001. Evidence for the autocrine induction of capacitation of mammalian spermatozoa. J. Biol. Chem.276: 26962–26968. [Erratum. 2001.J. Biol. Chem276: 47746.] [DOI] [PubMed] [Google Scholar]
  • 201.Ramanadham S., Gross R. W., Han X., Turk J.1993. Inhibition of arachidonate release by secretagogue-stimulated pancreatic islets suppresses both insulin secretion and the rise in beta-cell cytosolic calcium ion concentration. Biochemistry.32: 337–346. [DOI] [PubMed] [Google Scholar]
  • 202.Ramanadham S., Gross R., Turk J.1992. Arachidonic acid induces an increase in the cytosolic calcium concentration in single pancreatic islet beta cells. Biochem. Biophys. Res. Commun.184: 647–653. [DOI] [PubMed] [Google Scholar]
  • 203.Ramanadham S., Bohrer A., Gross R. W., Turk J.1993. Mass spectrometric characterization of arachidonate-containing plasmalogens in human pancreatic islets and in rat islet beta-cells and subcellular membranes. Biochemistry.32: 13499–13509. [DOI] [PubMed] [Google Scholar]
  • 204.Ramanadham S., Bohrer A., Mueller M., Jett P., Gross R. W., Turk J.1993. Mass spectrometric identification and quantitation of arachidonate-containing phospholipids in pancreatic islets: prominence of plasmenylethanolamine molecular species. Biochemistry.32: 5339–5351. [DOI] [PubMed] [Google Scholar]
  • 205.Ramanadham S., Hsu F. F., Bohrer A., Ma Z., Turk J.1999. Studies of the role of group VI phospholipase A2 in fatty acid incorporation, phospholipid remodeling, lysophosphatidylcholine generation, and secretagogue-induced arachidonic acid release in pancreatic islets and insulinoma cells. J. Biol. Chem.274: 13915–13927. [DOI] [PubMed] [Google Scholar]
  • 206.Bao S., Li Y., Lei X., Wohltmann M., Jin W., Bohrer A., Semenkovich C. F., Ramanadham S., Tabas I., Turk J.2007. Attenuated free cholesterol loading-induced apoptosis but preserved phospholipid composition of peritoneal macrophages from mice that do not express group VIA phospholipase A2. J. Biol. Chem.282: 27100–27114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Ashcroft S. J.1980. Glucoreceptor mechanisms and the control of insulin release and biosynthesis. Diabetologia.18: 5–15. [DOI] [PubMed] [Google Scholar]
  • 208.Hedeskov C. J.1980. Mechanism of glucose-induced insulin secretion. Physiol. Rev.60: 442–509. [DOI] [PubMed] [Google Scholar]
  • 209.Malaisse W. J., Sener A., Herchuelz A., Hutton J. C.1979. Insulin release: the fuel hypothesis. Metabolism.28: 373–386. [DOI] [PubMed] [Google Scholar]
  • 210.Meglasson M. D., Matschinsky F. M.1986. Pancreatic islet glucose metabolism and regulation of insulin secretion. Diabetes Metab. Rev.2: 163–214. [DOI] [PubMed] [Google Scholar]
  • 211.Ashcroft F. M., Harrison D. E., Ashcroft S. J.1984. Glucose induces closure of single potassium channels in isolated rat pancreatic beta-cells. Nature.312: 446–448. [DOI] [PubMed] [Google Scholar]
  • 212.Cook D. L., Hales C. N.1984. Intracellular ATP directly blocks K+ channels in pancreatic B-cells. Nature.311: 271–273. [DOI] [PubMed] [Google Scholar]
  • 213.Rorsman P., Trube G.1985. Glucose dependent K+-channels in pancreatic beta-cells are regulated by intracellular ATP. Pflugers Arch.405: 305–309. [DOI] [PubMed] [Google Scholar]
  • 214.Sturgess N. C., Ashford M. L., Cook D. L., Hales C. N.1985. The sulphonylurea receptor may be an ATP-sensitive potassium channel. Lancet.2: 474–475. [DOI] [PubMed] [Google Scholar]
  • 215.Arkhammar P., Nilsson T., Rorsman P., Berggren P. O.1987. Inhibition of ATP-regulated K+ channels precedes depolarization-induced increase in cytoplasmic free Ca2+ concentration in pancreatic beta-cells. J. Biol. Chem.262: 5448–5454. [PubMed] [Google Scholar]
  • 216.Gylfe E.1988. Nutrient secretagogues induce bimodal early changes in cytoplasmic calcium of insulin-releasing ob/ob mouse beta-cells. J. Biol. Chem.263: 13750–13754. [PubMed] [Google Scholar]
  • 217.Gylfe E.1988. Glucose-induced early changes in cytoplasmic calcium of pancreatic beta-cells studied with time-sharing dual-wavelength fluorometry. J. Biol. Chem.263: 5044–5048. [PubMed] [Google Scholar]
  • 218.Keahey H. H., Rajan A. S., Boyd A. E., 3rd, Kunze D. L.1989. Characterization of voltage-dependent Ca2+ channels in beta-cell line. Diabetes.38: 188–193. [DOI] [PubMed] [Google Scholar]
  • 219.Vacher P., McKenzie J., Dufy B.1989. Arachidonic acid affects membrane ionic conductances of GH3 pituitary cells. Am. J. Physiol.257: E203–E211. [DOI] [PubMed] [Google Scholar]
  • 220.Wollheim C. B., Sharp G. W.1981. Regulation of insulin release by calcium. Physiol. Rev.61: 914–973. [DOI] [PubMed] [Google Scholar]
  • 221.Biden T. J., Peter-Riesch B., Schlegel W., Wollheim C. B.1987. Ca2+-mediated generation of inositol 1,4,5-triphosphate and inositol 1,3,4,5-tetrakisphosphate in pancreatic islets. Studies with K+, glucose, and carbamylcholine. J. Biol. Chem.262: 3567–3571. [PubMed] [Google Scholar]
  • 222.Metz S. A.1988. Membrane phospholipid turnover as an intermediary step in insulin secretion. Putative roles of phospholipases in cell signaling. Am. J. Med.85: 9–21. [DOI] [PubMed] [Google Scholar]
  • 223.Prentki M., Matschinsky F. M.1987. Ca2+, cAMP, and phospholipid-derived messengers in coupling mechanisms of insulin secretion. Physiol. Rev.67: 1185–1248. [DOI] [PubMed] [Google Scholar]
  • 224.Robertson R. P.1988. Eicosanoids as pluripotential modulators of pancreatic islet function. Diabetes.37: 367–370. [DOI] [PubMed] [Google Scholar]
  • 225.Turk J., Wolf B. A., McDaniel M. L.1987. The role of phospholipid-derived mediators including arachidonic acid, its metabolites, and inositoltrisphosphate and of intracellular Ca2+ in glucose-induced insulin secretion by pancreatic islets. Prog. Lipid Res.26: 125–181. [DOI] [PubMed] [Google Scholar]
  • 226.Turk J., Mueller M., Bohrer A., Ramanadham S.1992. Arachidonic acid metabolism in isolated pancreatic islets. VI. Carbohydrate insulin secretagogues must be metabolized to induce eicosanoid release. Biochim. Biophys. Acta.1125: 280–291. [DOI] [PubMed] [Google Scholar]
  • 227.Metz S. A., Draznin B., Sussman K. E., Leitner J. W.1987. Unmasking of arachidonate-induced insulin release by removal of extracellular calcium. Arachidonic acid mobilizes cellular calcium in rat islets of Langerhans. Biochem. Biophys. Res. Commun.142: 251–258. [DOI] [PubMed] [Google Scholar]
  • 228.Jacobson D. A., Weber C. R., Bao S., Turk J., Philipson L. H.2007. Modulation of the pancreatic islet beta-cell-delayed rectifier potassium channel Kv2.1 by the polyunsaturated fatty acid arachidonate. J. Biol. Chem.282: 7442–7449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Nanda B. L., Nataraju A., Rajesh R., Rangappa K. S., Shekar M. A., Vishwanath B. S.2007. PLA2 mediated arachidonate free radicals: PLA2 inhibition and neutralization of free radicals by anti-oxidants–a new role as anti-inflammatory molecule. Curr. Top. Med. Chem.7: 765–777. [DOI] [PubMed] [Google Scholar]
  • 230.Weaver J. R., Holman T. R., Imai Y., Jadhav A., Kenyon V., Maloney D. J., Nadler J. L., Rai G., Simeonov A., Taylor-Fishwick D. A.2012. Integration of pro-inflammatory cytokines, 12-lipoxygenase and NOX-1 in pancreatic islet beta cell dysfunction. Mol. Cell. Endocrinol.358: 88–95. [DOI] [PubMed] [Google Scholar]
  • 231.Loweth A. C., Scarpello J. H., Morgan N. G.1994. A comparison of cytosolic phospholipase A2 expression in human islets of Langerhans and rodent insulin-secreting cells. Biochem. Soc. Trans.22: 430S. [DOI] [PubMed] [Google Scholar]
  • 232.Jolly Y. C., Major C., Wolf B. A.1993. Transient activation of calcium-dependent phospholipase A2 by insulin secretagogues in isolated pancreatic islets. Biochemistry.32: 12209–12217. [DOI] [PubMed] [Google Scholar]
  • 233.Loweth A. C., Scarpello J. H., Morgan N. G.1995. Phospholipase A2 expression in human and rodent insulin-secreting cells. Mol. Cell. Endocrinol.112: 177–183. [DOI] [PubMed] [Google Scholar]
  • 234.Chen M., Yang Z., Naji A., Wolf B. A.1996. Identification of calcium-dependent phospholipase A2 isoforms in human and rat pancreatic islets and insulin secreting beta-cell lines. Endocrinology.137: 2901–2909. [DOI] [PubMed] [Google Scholar]
  • 235.Dunlop M., Clark S.1995. Glucose-induced phosphorylation and activation of a high molecular weight cytosolic phospholipase A2 in neonatal rat pancreatic islets. Int. J. Biochem. Cell Biol.27: 1191–1199. [DOI] [PubMed] [Google Scholar]
  • 236.Eerola L. I., Surrel F., Nevalainen T. J., Gelb M. H., Lambeau G., Laine V. J.2006. Analysis of expression of secreted phospholipases A2 in mouse tissues at protein and mRNA levels. Biochim. Biophys. Acta.1761: 745–756. [DOI] [PubMed] [Google Scholar]
  • 237.Juhl K., Efanov A. M., Olsen H. L., Gromada J.2003. Secretory phospholipase A2 is released from pancreatic beta-cells and stimulates insulin secretion via inhibition of ATP-dependent K+ channels. Biochem. Biophys. Res. Commun.310: 274–279. [DOI] [PubMed] [Google Scholar]
  • 238.Ramanadham S., Ma Z., Arita H., Zhang S., Turk J.1998. Type IB secretory phospholipase A2 is contained in insulin secretory granules of pancreatic islet beta-cells and is co-secreted with insulin from glucose-stimulated islets. Biochim. Biophys. Acta.1390: 301–312. [DOI] [PubMed] [Google Scholar]
  • 239.Metz S., Holmes D., Robertson R. P., Leitner W., Draznin B.1991. Gene expression of type I phospholipase A2 in pancreatic beta cells. Regulation of mRNA levels by starvation or glucose excess. FEBS Lett.295: 110–112. [DOI] [PubMed] [Google Scholar]
  • 240.Karli U. O., Schafer T., Burger M. M.1990. Fusion of neurotransmitter vesicles with target membrane is calcium independent in a cell-free system. Proc. Natl. Acad. Sci. USA.87: 5912–5915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Nagao T., Kubo T., Fujimoto R., Nishio H., Takeuchi T., Hata F.1995. Ca2+-independent fusion of secretory granules with phospholipase A2-treated plasma membranes in vitro. Biochem. J.307: 563–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Nishio H., Takeuchi T., Hata F., Yagasaki O.1996. Ca(2+)-independent fusion of synaptic vesicles with phospholipase A2-treated presynaptic membranes in vitro. Biochem. J.318: 981–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Ishizaki J., Hanasaki K., Higashino K., Kishino J., Kikuchi N., Ohara O., Arita H.1994. Molecular cloning of pancreatic group I phospholipase A2 receptor. J. Biol. Chem.269: 5897–5904. [PubMed] [Google Scholar]
  • 244.Lambeau G., Ancian P., Barhanin J., Lazdunski M.1994. Cloning and expression of a membrane receptor for secretory phospholipases A2. J. Biol. Chem.269: 1575–1578. [PubMed] [Google Scholar]
  • 245.Zhou Y. P., Teng D., Dralyuk F., Ostrega D., Roe M. W., Philipson L., Polonsky K. S.1998. Apoptosis in insulin-secreting cells. Evidence for the role of intracellular Ca2+ stores and arachidonic acid metabolism. J. Clin. Invest.101: 1623–1632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Lei X., Bone R. N., Ali T., Wohltmann M., Gai Y., Goodwin K. J., Bohrer A. E., Turk J., Ramanadham S.2013. Genetic modulation of islet beta-cell iPLA2β expression provides evidence for its impact on beta-cell apoptosis and autophagy. Islets.5: 29–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Jayadev S., Liu B., Bielawska A. E., Lee J. Y., Nazaire F., Pushkareva M. Y., Obeid L. M., Hannun Y. A.1995. Role for ceramide in cell cycle arrest. J. Biol. Chem.270: 2047–2052. [DOI] [PubMed] [Google Scholar]
  • 248.Obeid L. M., Hannun Y. A.1995. Ceramide: a stress signal and mediator of growth suppression and apoptosis. J. Cell. Biochem.58: 191–198. [DOI] [PubMed] [Google Scholar]
  • 249.Venable M. E., Lee J. Y., Smyth M. J., Bielawska A., Obeid L. M.1995. Role of ceramide in cellular senescence. J. Biol. Chem.270: 30701–30708. [DOI] [PubMed] [Google Scholar]
  • 250.Song H., Wohltmann M., Tan M., Bao S., Ladenson J. H., Turk J.2012. Group VIA PLA2 (iPLA2beta) is activated upstream of p38 mitogen-activated protein kinase (MAPK) in pancreatic islet beta-cell signaling. J. Biol. Chem.287: 5528–5541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Vannuvel K., Renard P., Raes M., Arnould T.2013. Functional and morphological impact of ER stress on mitochondria. J. Cell. Physiol.228: 1802–1818. [DOI] [PubMed] [Google Scholar]
  • 252.Adams J. M., Cory S.2007. The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene.26: 1324–1337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Michels J., Kepp O., Senovilla L., Lissa D., Castedo M., Kroemer G., Galluzzi L.2013. Functions of Bcl-xL at the interface between cell death and metabolism. Int. J. Cell Biol.2013: 705294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Yip K. W., Reed J. C.2008. Bcl-2 family proteins and cancer. Oncogene.27: 6398–6406. [DOI] [PubMed] [Google Scholar]
  • 255.Carrington E. M., McKenzie M. D., Jansen E., Myers M., Fynch S., Kos C., Strasser A., Kay T. W., Scott C. L., Allison J.2009. Islet beta-cells deficient in Bcl-xL develop but are abnormally sensitive to apoptotic stimuli. Diabetes.58: 2316–2323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Federici M., Hribal M., Perego L., Ranalli M., Caradonna Z., Perego C., Usellini L., Nano R., Bonini P., Bertuzzi F., et al. 2001. High glucose causes apoptosis in cultured human pancreatic islets of Langerhans: a potential role for regulation of specific Bcl family genes toward an apoptotic cell death program. Diabetes.50: 1290–1301. [DOI] [PubMed] [Google Scholar]
  • 257.Hui H., Khoury N., Zhao X., Balkir L., D’Amico E., Bullotta A., Nguyen E. D., Gambotto A., Perfetti R.2005. Adenovirus-mediated XIAP gene transfer reverses the negative effects of immunosuppressive drugs on insulin secretion and cell viability of isolated human islets. Diabetes.54: 424–433. [DOI] [PubMed] [Google Scholar]
  • 258.Klein D., Ribeiro M. M., Mendoza V., Jayaraman S., Kenyon N. S., Pileggi A., Molano R. D., Inverardi L., Ricordi C., Pastori R. L.2004. Delivery of Bcl-XL or its BH4 domain by protein transduction inhibits apoptosis in human islets. Biochem. Biophys. Res. Commun.323: 473–478. [DOI] [PubMed] [Google Scholar]
  • 259.Zhou Y-P., Pena J. C., Roe M. W., Mittal A., Levisetti M., Baldwin A. C., Pugh W., Ostrega D., Ahmed N., Bindokas V. P., et al. 2000. Overexpression of Bcl-xL in beta -cells prevents cell death but impairs mitochondrial signal for insulin secretion. Am. J. Physiol. Endocrinol. Metab.278: E340–E351. [DOI] [PubMed] [Google Scholar]
  • 260.Schwerk C., Schulze-Osthoff K.2005. Regulation of apoptosis by alternative pre-mRNA splicing. Mol. Cell.19: 1–13. [DOI] [PubMed] [Google Scholar]
  • 261.Barbour S. E., Nguyen P. T., Park M., Emani B., Lei X., Kambalapalli M., Shultz J. C., Wijesinghe D., Chalfant C. E., Ramanadham S.2015. Group VIA phospholipase A2 (iPLA2β) modulates Bcl-x 5′-splice site selection and suppresses anti-apoptotic Bcl-x(L) in β-cells. J. Biol. Chem.290: 11021–11031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Seleznev K., Zhao C., Zhang X. H., Song K., Ma Z. A.2006. Calcium-independent phospholipase A2 localizes in and protects mitochondria during apoptotic induction by staurosporine. J. Biol. Chem.281: 22275–22288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Zhao Z., Zhang X., Zhao C., Choi J., Shi J., Song K., Turk J., Ma Z. A.2010. Protection of pancreatic beta-cells by group VIA phospholipase A2-mediated repair of mitochondrial membrane peroxidation. Endocrinology.151: 3038–3048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Luo P., Wang M. H.2011. Eicosanoids, beta-cell function, and diabetes. Prostaglandins Other Lipid Mediat.95: 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Jenkins C. M., Cedars A., Gross R. W.2009. Eicosanoid signalling pathways in the heart. Cardiovasc. Res.82: 240–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Khanapure S. P., Garvey D. S., Janero D. R., Letts L. G.2007. Eicosanoids in inflammation: biosynthesis, pharmacology, and therapeutic frontiers. Curr. Top. Med. Chem.7: 311–340. [DOI] [PubMed] [Google Scholar]
  • 267.Adibhatla R. M., Hatcher J. F.2008. Phospholipase A(2), reactive oxygen species, and lipid peroxidation in CNS pathologies. BMB Rep.41: 560–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Bending D., Zaccone P., Cooke A.2012. Inflammation and type one diabetes. Int. Immunol.24: 339–346. [DOI] [PubMed] [Google Scholar]
  • 269.Kim N., Luster A. D.2007. Regulation of immune cells by eicosanoid receptors. ScientificWorldJournal.7: 1307–1328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Korotkova M., Jakobsson P. J.2014. Persisting eicosanoid pathways in rheumatic diseases. Nat. Rev. Rheumatol.10: 229–241. [DOI] [PubMed] [Google Scholar]
  • 271.Li H., Edin M. L., Gruzdev A., Cheng J., Bradbury J. A., Graves J. P., DeGraff L. M., Zeldin D. C.2013. Regulation of T helper cell subsets by cyclooxygenases and their metabolites. Prostaglandins Other Lipid Mediat.104–105: 74–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Mirshafiey A., Jadidi-Niaragh F.2010. Prostaglandins in pathogenesis and treatment of multiple sclerosis. Immunophar­macol. Immunotoxicol.32: 543–554. [DOI] [PubMed] [Google Scholar]
  • 273.Willcox A., Richardson S. J., Bone A. J., Foulis A. K., Morgan N. G.2009. Analysis of islet inflammation in human type 1 diabetes. Clin. Exp. Immunol.155: 173–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Yousefi B., Jadidi-Niaragh F., Azizi G., Hajighasemi F., Mirshafiey A.2014. The role of leukotrienes in immunopathogenesis of rheumatoid arthritis. Mod. Rheumatol.24: 225–235. [DOI] [PubMed] [Google Scholar]
  • 275.Kim R., Emi M., Tanabe K.2005. Cancer cell immune escape and tumor progression by exploitation of anti-inflammatory and pro-inflammatory responses. Cancer Biol. Ther.4: 924–933. [DOI] [PubMed] [Google Scholar]
  • 276.Atkinson M. A.1997. Mechanisms underlying the loss of self tolerance in NOD mice. Res. Immunol.148: 301–306. [DOI] [PubMed] [Google Scholar]
  • 277.Trudeau J. D., Dutz J. P., Arany E., Hill D. J., Fieldus W. E., Finegood D. T.2000. Neonatal beta-cell apoptosis: a trigger for autoimmune diabetes?Diabetes.49: 1–7. [DOI] [PubMed] [Google Scholar]
  • 278.Basu S., Larsson A., Vessby J., Vessby B., Berne C.2005. Type 1 diabetes is associated with increased cyclooxygenase- and cytokine-mediated inflammation. Diabetes Care.28: 1371–1375. [DOI] [PubMed] [Google Scholar]
  • 279.Lauber K., Bohn E., Krober S. M., Xiao Y. J., Blumenthal S. G., Lindemann R. K., Marini P., Wiedig C., Zobywalski A., Baksh S., et al. 2003. Apoptotic cells induce migration of phagocytes via caspase-3-mediated release of a lipid attraction signal. Cell.113: 717–730. [DOI] [PubMed] [Google Scholar]
  • 280.Peter C., Waibel M., Keppeler H., Lehmann R., Xu G., Halama A., Adamski J., Schulze-Osthoff K., Wesselborg S., Lauber K.2012. Release of lysophospholipid ‘find-me’ signals during apoptosis requires the ATP-binding cassette transporter A1. Autoimmunity.45: 568–573. [DOI] [PubMed] [Google Scholar]
  • 281.Peter C., Waibel M., Radu C. G., Yang L. V., Witte O. N., Schulze-Osthoff K., Wesselborg S., Lauber K.2008. Migration to apoptotic “find-me” signals is mediated via the phagocyte receptor G2A. J. Biol. Chem.283: 5296–5305. [DOI] [PubMed] [Google Scholar]
  • 282.Irving-Rodgers H. F., Ziolkowski A. F., Parish C. R., Sado Y., Ninomiya Y., Simeonovic C. J., Rodgers R. J.2008. Molecu­lar composition of the peri-islet basement membrane in NOD mice: a barrier against destructive insulitis. Diabetologia.51: 1680–1688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Eizirik D. L., Mandrup-Poulsen T.2001. A choice of death-the signal-transduction of immune-mediated beta-cell apoptosis. Diabetologia.44: 2115–2133. [DOI] [PubMed] [Google Scholar]
  • 284.Taylor-Fishwick D. A., Weaver J., Glenn L., Kuhn N., Rai G., Jadhav A., Simeonov A., Dudda A., Schmoll D., Holman T. R., et al. 2015. Selective inhibition of 12-lipoxygenase protects islets and beta cells from inflammatory cytokine-mediated beta cell dysfunction. Diabetologia.58: 549–557. [DOI] [PubMed] [Google Scholar]
  • 285.Choi K. S.2012. Autophagy and cancer. Exp. Mol. Med.44: 109–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Scarlatti F., Bauvy C., Ventruti A., Sala G., Cluzeaud F., Vandewalle A., Ghidoni R., Codogno P.2004. Ceramide-mediated macroautophagy involves inhibition of protein kinase B and up-regulation of beclin 1. J. Biol. Chem.279: 18384–18391. [DOI] [PubMed] [Google Scholar]
  • 287.Tanida I.2011. Autophagosome formation and molecular mechanism of autophagy. Antioxid. Redox Signal.14: 2201–2214. [DOI] [PubMed] [Google Scholar]
  • 288.Ganley I. G., Wong P. M., Jiang X.2011. Thapsigargin distinguishes membrane fusion in the late stages of endocytosis and autophagy. Autophagy.7: 1397–1399. [DOI] [PubMed] [Google Scholar]
  • 289.Farooqui A. A., Ong W. Y., Horrocks L. A.2006. Inhibitors of brain phospholipase A2 activity: their neuropharmacological effects and therapeutic importance for the treatment of neurologic disorders. Pharmacol. Rev.58: 591–620. [DOI] [PubMed] [Google Scholar]
  • 290.Molloy G. Y., Rattray M., Williams R. J.1998. Genes encoding multiple forms of phospholipase A2 are expressed in rat brain. Neurosci. Lett.258: 139–142. [DOI] [PubMed] [Google Scholar]
  • 291.Farooqui A. A., Horrocks L. A.2004. Brain phospholipases A2: a perspective on the history. Prostaglandins Leukot. Essent. Fatty Acids.71: 161–169. [DOI] [PubMed] [Google Scholar]
  • 292.Ong W. Y., Yeo J. F., Ling S. F., Farooqui A. A.2005. Distribution of calcium-independent phospholipase A2 (iPLA2) in monkey brain. J. Neurocytol.34: 447–458. [DOI] [PubMed] [Google Scholar]
  • 293.Yang H. C., Mosior M., Ni B., Dennis E. A.1999. Regional distribution, ontogeny, purification, and characterization of the Ca2+-independent phospholipase A2 from rat brain. J. Neurochem.73: 1278–1287. [DOI] [PubMed] [Google Scholar]
  • 294.Zanassi P., Paolillo M., Schinelli S.1998. Coexpression of phospholipase A2 isoforms in rat striatal astrocytes. Neurosci. Lett.247: 83–86. [DOI] [PubMed] [Google Scholar]
  • 295.Balboa M. A., Varela-Nieto I., Killermann Lucas K., Dennis E. A.2002. Expression and function of phospholipase A2 in brain. FEBS Lett.531: 12–17. [DOI] [PubMed] [Google Scholar]
  • 296.Shirai Y., Ito M.2004. Specific differential expression of phospholipase A2 subtypes in rat cerebellum. J. Neurocytol.33: 297–307. [DOI] [PubMed] [Google Scholar]
  • 297.Rapoport S. I.2008. Arachidonic acid and the brain. J. Nutr.138: 2515–2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Ramanadham S., Hsu F. F., Bohrer A., Nowatzke W., Ma Z., Turk J.1998. Electrospray ionization mass spectrometric analyses of phospholipids from rat and human pancreatic islets and subcellular membranes: comparison to other tissues and implications for membrane fusion in insulin exocytosis. Biochemistry.37: 4553–4567. [DOI] [PubMed] [Google Scholar]
  • 299.Cheon Y., Kim H. W., Igarashi M., Modi H. R., Chang L., Ma K., Greenstein D., Wohltmann M., Turk J., Rapoport S. I., et al. 2012. Disturbed brain phospholipid and docosahexaenoic acid metabolism in calcium-independent phospholipase A2-VIA (iPLA2β)-knockout mice. Biochim. Biophys. Acta.1821: 1278–1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Ross B. M., Hudson C., Erlich J., Warsh J. J., Kish S. J.1997. Increased phospholipid breakdown in schizophrenia. Evidence for the involvement of a calcium-independent phospholipase A2. Arch. Gen. Psychiatry.54: 487–494. [DOI] [PubMed] [Google Scholar]
  • 301.Smalheiser N. R., Swanson D. R.1998. Calcium-independent phospholipase A2 and schizophrenia. Arch. Gen. Psychiatry.55: 752–753. [DOI] [PubMed] [Google Scholar]
  • 302.Ross B. M., Turenne S., Moszczynska A., Warsh J. J., Kish S. J.1999. Differential alteration of phospholipase A2 activities in brain of patients with schizophrenia. Brain Res.821: 407–413. [DOI] [PubMed] [Google Scholar]
  • 303.Schaeffer E. L., Gattaz W. F., Eckert G. P.2012. Alterations of brain membranes in schizophrenia: impact of phospholipase A2. Curr. Top. Med. Chem.12: 2314–2323. [DOI] [PubMed] [Google Scholar]
  • 304.Smesny S., Kinder D., Willhardt I., Rosburg T., Lasch J., Berger G., Sauer H.2005. Increased calcium-independent phospholipase A2 activity in first but not in multiepisode chronic schizophrenia. Biol. Psychiatry.57: 399–405. [DOI] [PubMed] [Google Scholar]
  • 305.Gattaz W. F., Valente K. D., Raposo N. R., Vincentiis S., Talib L. L.2011. Increased PLA2 activity in the hippocampus of patients with temporal lobe epilepsy and psychosis. J. Psychiatr. Res.45: 1617–1620. [DOI] [PubMed] [Google Scholar]
  • 306.Junqueira R., Cordeiro Q., Meira-Lima I., Gattaz W. F., Vallada H.2004. Allelic association analysis of phospholipase A2 genes with schizophrenia. Psychiatr. Genet.14: 157–160. [DOI] [PubMed] [Google Scholar]
  • 307.Kerr D. S., Talib L. L., Yamamoto V. J., Ferreira A. S., Zanetti M. V., Serpa M. H., Busatto G. F., Van de Bilt M. T., Gattaz W. F.2013. Antipsychotic drugs decrease iPLA2 gene expression in schizophrenia. Schizophr. Res.147: 203–204. [DOI] [PubMed] [Google Scholar]
  • 308.Yu Y., Tao R., Shi J., Zhang X., Kou C., Guo Y., Zhang X., Lin X., Liu S., Ju G., et al. 2005. A genetic study of two calcium-independent cytosolic PLA2 genes in schizophrenia. Prostaglandins Leukot. Essent. Fatty Acids.73: 351–354. [DOI] [PubMed] [Google Scholar]
  • 309.Wilson D. M., Binder L. I.1997. Free fatty acids stimulate the polymerization of tau and amyloid beta peptides. In vitro evidence for a common effector of pathogenesis in Alzheimer’s disease. Am. J. Pathol.150: 2181–2195. [PMC free article] [PubMed] [Google Scholar]
  • 310.Talbot K., Young R. A., Jolly-Tornetta C., Lee V. M., Trojanowski J. Q., Wolf B. A.2000. A frontal variant of Alzheimer’s disease exhibits decreased calcium-independent phospholipase A2 activity in the prefrontal cortex. Neurochem. Int.37: 17–31. [DOI] [PubMed] [Google Scholar]
  • 311.Wolf M. J., Izumi Y., Zorumski C. F., Gross R. W.1995. Long-term potentiation requires activation of calcium-independent phospholipase A2. FEBS Lett.377: 358–362. [DOI] [PubMed] [Google Scholar]
  • 312.Schaeffer E. L., Forlenza O. V., Gattaz W. F.2009. Phos­pholipase A2 activation as a therapeutic approach for cognitive enhancement in early-stage Alzheimer disease. Psycho­pharmacology (Berl.).202: 37–51. [DOI] [PubMed] [Google Scholar]
  • 313.Tomiyama H., Yoshino H., Hattori N.2011. Analysis of PLA2G6 in patients with frontotemporal type of dementia. Parkinsonism Relat. Disord.17: 493–494. [DOI] [PubMed] [Google Scholar]
  • 314.Ikenaga E. H., Talib L. L., Ferreira A. S., Machado-Vieira R., Forlenza O. V., Gattaz W. F.2015. Reduced activities of phospholipases A2 in platelets of drug-naive bipolar disorder patients. Bipolar Disord.17: 97–101. [DOI] [PubMed] [Google Scholar]
  • 315.Ross B. M., Moszczynska A., Erlich J., Kish S. J.1998. Low activity of key phospholipid catabolic and anabolic enzymes in human substantia nigra: possible implications for Parkinson’s disease. Neuroscience.83: 791–798. [DOI] [PubMed] [Google Scholar]
  • 316.Paisan-Ruiz C., Bhatia K. P., Li A., Hernandez D., Davis M., Wood N. W., Hardy J., Houlden H., Singleton A., Schneider S. A.2009. Characterization of PLA2G6 as a locus for dystonia-parkinsonism. Ann. Neurol.65: 19–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317.Paisán-Ruiz C., Guevara R., Federoff M., Hanagasi H., Sina F., Elahi E., Schneider S. A., Schwingenschuh P., Bajaj N., Emre M., et al. 2010. Early-onset L-dopa-responsive parkinsonism with pyramidal signs due to ATP13A2, PLA2G6, FBXO7 and spatacsin mutations. Mov. Disord.25: 1791–1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Yoshino H., Tomiyama H., Tachibana N., Ogaki K., Li Y., Funayama M., Hashimoto T., Takashima S., Hattori N.2010. Phenotypic spectrum of patients with PLA2G6 mutation and PARK14-linked parkinsonism. Neurology.75: 1356–1361. [DOI] [PubMed] [Google Scholar]
  • 319.Shi C. H., Tang B. S., Wang L., Lv Z. Y., Wang J., Luo L. Z., Shen L., Jiang H., Yan X. X., Pan Q., et al. 2011. PLA2G6 gene mutation in autosomal recessive early-onset parkinsonism in a Chinese cohort. Neurology.77: 75–81. [DOI] [PubMed] [Google Scholar]
  • 320.Lu C. S., Lai S. C., Wu R. M., Weng Y. H., Huang C. L., Chen R. S., Chang H. C., Wu-Chou Y. H., Yeh T. H.2012. PLA2G6 mutations in PARK14-linked young-onset parkinsonism and sporadic Parkinson’s disease. Am. J. Med. Genet. B Neuropsychiatr. Genet.159B: 183–191. [DOI] [PubMed] [Google Scholar]
  • 321.Paisán-Ruiz C., Li A., Schneider S. A., Holton J. L., Johnson R., Kidd D., Chataway J., Bhatia K. P., Lees A. J., Hardy J., et al. 2012. Widespread Lewy body and tau accumulation in childhood and adult onset dystonia-parkinsonism cases with PLA2G6 mutations. Neurobiol. Aging.33: 814–823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Gui Y. X., Xu Z. P., Wen L., Liu H. M., Zhao J. J., Hu X. Y.2013. Four novel rare mutations of PLA2G6 in Chinese population with Parkinson’s disease. Parkinsonism Relat. Disord.19: 21–26. [DOI] [PubMed] [Google Scholar]
  • 323.Tan E. K., Ho P., Tan L., Prakash K. M., Zhao Y.2010. PLA2G6 mutations and Parkinson’s disease. Ann. Neurol.67: 148. [DOI] [PubMed] [Google Scholar]
  • 324.Tian J. Y., Tang B. S., Shi C. H., Lv Z. Y., Li K., Yu R. L., Shen L., Yan X. X., Guo J. F.2012. Analysis of PLA2G6 gene mutation in sporadic early-onset parkinsonism patients from Chinese population. Neurosci. Lett.514: 156–158. [DOI] [PubMed] [Google Scholar]
  • 325.Lv Z., Guo J., Sun Q., Li K., Yu R., Tian J., Yan X., Tang B.2012. Association between PLA2G6 gene polymorphisms and Parkinson’s disease in the Chinese Han population. Parkinsonism Relat. Disord.18: 641–644. [DOI] [PubMed] [Google Scholar]
  • 326.Tomiyama H., Yoshino H., Ogaki K., Li L., Yamashita C., Li Y., Funayama M., Sasaki R., Kokubo Y., Kuzuhara S., et al. 2011. PLA2G6 variant in Parkinson’s disease. J. Hum. Genet.56: 401–403. [DOI] [PubMed] [Google Scholar]
  • 327.Khateeb S., Flusser H., Ofir R., Shelef I., Narkis G., Vardi G., Shorer Z., Levy R., Galil A., Elbedour K., et al. 2006. PLA2G6 mutation underlies infantile neuroaxonal dystrophy. Am. J. Hum. Genet.79: 942–948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328.Biancheri R., Rossi A., Alpigiani G., Filocamo M., Gandolfo C., Lorini R., Minetti C.2007. Cerebellar atrophy without cerebellar cortex hyperintensity in infantile neuroaxonal dystrophy (INAD) due to PLA2G6 mutation. Eur. J. Paediatr. Neurol.11: 175–177. [DOI] [PubMed] [Google Scholar]
  • 329.Polster B., Crosier M., Lindsay S., Hayflick S.2010. Expression of PLA2G6 in human fetal development: implications for infantile neuroaxonal dystrophy. Brain Res. Bull.83: 374–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Malik I., Turk J., Mancuso D. J., Montier L., Wohltmann M., Wozniak D. F., Schmidt R. E., Gross R. W., Kotzbauer P. T.2008. Disrupted membrane homeostasis and accumulation of ubiquitinated proteins in a mouse model of infantile neuroaxonal dystrophy caused by PLA2G6 mutations. Am. J. Pathol.172: 406–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331.Wada H., Yasuda T., Miura I., Watabe K., Sawa C., Kamijuku H., Kojo S., Taniguchi M., Nishino I., Wakana S., et al. 2009. Establishment of an improved mouse model for infantile neuroaxonal dystrophy that shows early disease onset and bears a point mutation in PLA2G6. Am. J. Pathol.175: 2257–2263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.Gregory A., Westaway S. K., Holm I. E., Kotzbauer P. T., Hogarth P., Sonek S., Coryell J. C., Nguyen T. M., Nardocci N., Zorzi G., et al. 2008. Neurodegeneration associated with genetic defects in phospholipase A2. Neurology.71: 1402–1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 333.Carrilho I., Santos M., Guimaraes A., Teixeira J., Chorao R., Martins M., Dias C., Gregory A., Westaway S., Nguyen T., et al. 2008. Infantile neuroaxonal dystrophy: what’s most important for the diagnosis?Eur. J. Paediatr. Neurol.12: 491–500. [DOI] [PubMed] [Google Scholar]
  • 334.Beck G., Sugiura Y., Shinzawa K., Kato S., Setou M., Tsujimoto Y., Sakoda S., Sumi-Akamaru H.2011. Neuroaxonal dystrophy in calcium-independent phospholipase A2beta deficiency results from insufficient remodeling and degeneration of mitochondrial and presynaptic membranes. J. Neurosci.31: 11411–11420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335.Strokin M., Seburn K. L., Cox G. A., Martens K. A., Reiser G.2012. Severe disturbance in the Ca2+ signaling in astrocytes from mouse models of human infantile neuroaxonal dystrophy with mutated PLA2G6. Hum. Mol. Genet.21: 2807–2814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336.Wada H., Kojo S., Seino K.2013. Mouse models of human INAD by PLA2G6 deficiency. Histol. Histopathol.28: 965–969. [DOI] [PubMed] [Google Scholar]
  • 337.Zhao Z., Wang J., Zhao C., Bi W., Yue Z., Ma Z. A.2011. Genetic ablation of PLA2G6 in mice leads to cerebellar atrophy characterized by Purkinje cell loss and glial cell activation. PLoS ONE.6: e26991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 338.Kruer M. C., Paudel R., Wagoner W., Sanford L., Kara E., Gregory A., Foltynie T., Lees A., Bhatia K., Hardy J., et al. 2012. Analysis of ATP13A2 in large neurodegeneration with brain iron accumulation (NBIA) and dystonia-parkinsonism cohorts. Neurosci. Lett.523: 35–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Morgan N. V., Westaway S. K., Morton J. E., Gregory A., Gissen P., Sonek S., Cangul H., Coryell J., Canham N., Nardocci N., et al. 2006. PLA2G6, encoding a phospholipase A2, is mutated in neurodegenerative disorders with high brain iron. Nat. Genet.38: 752–754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340.Gregory A., Hayflick S. J.2011. Genetics of neurodegeneration with brain iron accumulation. Curr. Neurol. Neurosci. Rep.11: 254–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341.Hayflick S. J.2006. Neurodegeneration with brain iron accumulation: from genes to pathogenesis. Semin. Pediatr. Neurol.13: 182–185. [DOI] [PubMed] [Google Scholar]
  • 342.Engel L. A., Jing Z., O’Brien D. E., Sun M., Kotzbauer P. T.2010. Catalytic function of PLA2G6 is impaired by mutations associated with infantile neuroaxonal dystrophy but not dystonia-parkinsonism. PLoS ONE.5: e12897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343.Sina F., Shojaee S., Elahi E., Paisan-Ruiz C.2009. R632W mutation in PLA2G6 segregates with dystonia-parkinsonism in a consanguineous Iranian family. Eur. J. Neurol.16: 101–104. [DOI] [PubMed] [Google Scholar]
  • 344.Kurian M. A., Morgan N. V., MacPherson L., Foster K., Peake D., Gupta R., Philip S. G., Hendriksz C., Morton J. E., Kingston H. M., et al. 2008. Phenotypic spectrum of neurodegeneration associated with mutations in the PLA2G6 gene (PLAN). Neurology.70: 1623–1629. [DOI] [PubMed] [Google Scholar]
  • 345.Bower M. A., Bushara K., Dempsey M. A., Das S., Tuite P. J.2011. Novel mutations in siblings with later-onset PLA2G6-associated neurodegeneration (PLAN). Mov. Disord.26: 1768–1769. [DOI] [PubMed] [Google Scholar]
  • 346.Salih M. A., Mundwiller E., Khan A. O., AlDrees A., Elmalik S. A., Hassan H. H., Al-Owain M., Alkhalidi H. M., Katona I., Kabiraj M. M., et al. 2013. New findings in a global approach to dissect the whole phenotype of PLA2G6 gene mutations. PLoS ONE.8: e76831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347.Crompton D., Rehal P. K., MacPherson L., Foster K., Lunt P., Hughes I., Brady A. F., Pike M. G., De Gressi S., Morgan N. V., et al. 2010. Multiplex ligation-dependent probe amplification (MLPA) analysis is an effective tool for the detection of novel intragenic PLA2G6 mutations: implications for molecular diagnosis. Mol. Genet. Metab.100: 207–212. [DOI] [PubMed] [Google Scholar]
  • 348.Westaway S. K., Gregory A., Hayflick S. J.2007. Mutations in PLA2G6 and the riddle of Schindler disease. J. Med. Genet.44: e64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.Ross B. M., Hughes B., Kish S. J., Warsh J. J.2006. Serum calcium-independent phospholipase A2 activity in bipolar affective disorder. Bipolar Disord.8: 265–270. [DOI] [PubMed] [Google Scholar]
  • 350.Blanchard H., Taha A. Y., Cheon Y., Kim H. W., Turk J., Rapoport S. I.2014. iPLA2beta knockout mouse, a genetic model for progressive human motor disorders, develops age-related neuropathology. Neurochem. Res.39: 1522–1532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351.Lopez-Vales R., Navarro X., Shimizu T., Baskakis C., Kokotos G., Constantinou-Kokotou V., Stephens D., Dennis E. A., David S.2008. Intracellular phospholipase A2 group IVA and group VIA play important roles in Wallerian degeneration and axon regeneration after peripheral nerve injury. Brain.131: 2620–2631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Lopez-Vales R., Ghasemlou N., Redensek A., Kerr B. J., Barbayianni E., Antonopoulou G., Baskakis C., Rathore K. I., Constantinou-Kokotou V., Stephens D., et al. 2011. Phospholipase A2 superfamily members play divergent roles after spinal cord injury. FASEB J.25: 4240–4252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Giurdanella G., Motta C., Muriana S., Arena V., Anfuso C. D., Lupo G., Alberghina M.2011. Cytosolic and calcium-independent phospholipase A2 mediate glioma-enhanced proangiogenic activity of brain endothelial cells. Microvasc. Res.81: 1–17. [DOI] [PubMed] [Google Scholar]
  • 354.Lee L. H., Tan C. H., Shui G., Wenk M. R., Ong W. Y.2012. Role of prefrontal cortical calcium independent phospholipase A2 in antidepressant-like effect of maprotiline. Int. J. Neuropsychopharmacol.15: 1087–1098. [DOI] [PubMed] [Google Scholar]
  • 355.Moon K. H., Tajuddin N., Brown J., 3rd, Neafsey E. J., Kim H. Y., Collins M. A.2014. Phospholipase A2, oxidative stress, and neurodegeneration in binge ethanol-treated organotypic slice cultures of developing rat brain. Alcohol. Clin. Exp. Res.38: 161–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 356.Liou J. Y., Aleksic N., Chen S. F., Han T. J., Shyue S. K., Wu K. K.2005. Mitochondrial localization of cyclooxygenase-2 and calcium-independent phospholipase A2 in human cancer cells: implication in apoptosis resistance. Exp. Cell Res.306: 75–84. [DOI] [PubMed] [Google Scholar]
  • 357.Pérez R., Matabosch X., Llebaria A., Balboa M. A., Balsinde J.2006. Blockade of arachidonic acid incorporation into phospholipids induces apoptosis in U937 promonocytic cells. J. Lipid Res.47: 484–491. [DOI] [PubMed] [Google Scholar]
  • 358.Pérez R., Melero R., Balboa M. A., Balsinde J.2004. Role of group VIA calcium-independent phospholipase A2 in arachidonic acid release, phospholipid fatty acid incorporation, and apoptosis in U937 cells responding to hydrogen peroxide. J. Biol. Chem.279: 40385–40391. [DOI] [PubMed] [Google Scholar]
  • 359.Saavedra G., Zhang W., Peterson B., Cummings B. S.2006. Differential roles for cytosolic and microsomal Ca2+-independent phospholipase A2 in cell growth and maintenance of phospholipids. J. Pharmacol. Exp. Ther.318: 1211–1219. [DOI] [PubMed] [Google Scholar]
  • 360.Shin K. J., Chung C., Hwang Y. A., Kim S. H., Han M. S., Ryu S. H., Suh P. G.2002. Phospholipase A2-mediated Ca2+ influx by 2,2’,4,6-tetrachlorobiphenyl in PC12 cells. Toxicol. Appl. Pharmacol.178: 37–43. [DOI] [PubMed] [Google Scholar]
  • 361.Wilson H. A., Allred D. V., O’Neill K., Bell J. D.2000. Activities and interactions among phospholipases A2 during thapsigargin-induced S49 cell death. Apoptosis.5: 389–396. [DOI] [PubMed] [Google Scholar]
  • 362.Kim S. Y., Jo H. Y., Kim M. H., Cha Y. Y., Choi S. W., Shim J. H., Kim T. J., Lee K. Y.2008. H2O2-dependent hyperoxidation of peroxiredoxin 6 (Prdx6) plays a role in cellular toxicity via up-regulation of iPLA2 activity. J. Biol. Chem.283: 33563–33568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 363.Lallemand C., Blanchard B., Palmieri M., Lebon P., May E., Tovey M. G.2007. Single-stranded RNA viruses inactivate the transcriptional activity of p53 but induce NOXA-dependent apoptosis via post-translational modifications of IRF-1, IRF-3 and CREB. Oncogene.26: 328–338. [DOI] [PubMed] [Google Scholar]
  • 364.Zhang X. H., Zhao C., Ma Z. A.2007. The increase of cell-membranous phosphatidylcholines containing polyunsaturated fatty acid residues induces phosphorylation of p53 through activation of ATR. J. Cell Sci.120: 4134–4143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365.Hooks S. B., Cummings B. S.2008. Role of Ca2+-independent phospholipase A2 in cell growth and signaling. Biochem. Pharmacol.76: 1059–1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Scott K. F., Sajinovic M., Hein J., Nixdorf S., Galettis P., Liauw W., de Souza P., Dong Q., Graham G. G., Russell P. J.2010. Emerging roles for phospholipase A2 enzymes in cancer. Biochimie.92: 601–610. [DOI] [PubMed] [Google Scholar]
  • 367.Laye J. P., Gill J. H.2003. Phospholipase A2 expression in tumours: a target for therapeutic intervention?Drug Discov. Today.8: 710–716. [DOI] [PubMed] [Google Scholar]
  • 368.Bonney R. C., Franks S.1988. The activity of calcium dependent and calcium independent phospholipase A2 in normal endometrium and in endometrium from women suffering from menorrhagia and polycystic ovary syndrome. Gynecol. Endocrinol.2: 131–138. [DOI] [PubMed] [Google Scholar]
  • 369.Deleted in proof.
  • 370.Li H., Zhao Z., Wei G., Yan L., Wang D., Zhang H., Sandusky G. E., Turk J., Xu Y.2010. Group VIA phospholipase A2 in both host and tumor cells is involved in ovarian cancer development. FASEB J.24: 4103–4116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 371.Li H., Zhao Z., Antalis C., Zhao Z., Emerson R., Wei G., Zhang S., Zhang Z. Y., Xu Y.2011. Combination therapy of an inhibitor of group VIA phospholipase A2 with paclitaxel is highly effective in blocking ovarian cancer development. Am. J. Pathol.179: 452–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Shen Z., Belinson J., Morton R. E., Xu Y., Xu Y.1998. Phorbol 12-myristate 13-acetate stimulates lysophosphatidic acid secretion from ovarian and cervical cancer cells but not from breast or leukemia cells. Gynecol. Oncol.71: 364–368. [DOI] [PubMed] [Google Scholar]
  • 373.Cai Q., Zhao Z., Antalis C., Yan L., Del Priore G., Hamed A. H., Stehman F. B., Schilder J. M., Xu Y.2012. Elevated and secreted phospholipase A2 activities as new potential therapeutic targets in human epithelial ovarian cancer. FASEB J.26: 3306–3320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374.Nicotera T. M., Schuster D. P., Bourhim M., Chadha K., Klaich G., Corral D. A.2009. Regulation of PSA secretion and survival signaling by calcium-independent phopholipase A2beta in prostate cancer cells. Prostate.69: 1270–1280. [DOI] [PubMed] [Google Scholar]
  • 375.Sun B., Zhang X., Yonz C., Cummings B. S.2010. Inhibition of calcium-independent phospholipase A2 activates p38 MAPK signaling pathways during cytostasis in prostate cancer cells. Biochem. Pharmacol.79: 1727–1735. [DOI] [PubMed] [Google Scholar]
  • 376.Yun H. M., Park K. R., Lee H. P., Lee D. H., Jo M., Shin D. H., Yoon D. Y., Han S. B., Hong J. T.2014. PRDX6 promotes lung tumor progression via its GPx and iPLA2 activities. Free Radic. Biol. Med.69: 367–376. [DOI] [PubMed] [Google Scholar]
  • 377.McHowat J., Gullickson G., Hoover R. G., Sharma J., Turk J., Kornbluth J.2011. Platelet-activating factor and metastasis: calcium-independent phospholipase A2beta deficiency protects against breast cancer metastasis to the lung. Am. J. Physiol. Cell Physiol.300: C825–C832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 378.Kispert S., Marentette J., McHowat J.2015. Cigarette smoke induces cell motility via platelet-activating factor accumulation in breast cancer cells: a potential mechanism for metastatic disease. Physiol. Rep.3: e12318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 379.Zhu C., Sun Z., Li C., Guo R., Li L., Jin L., Wan R., Li S.2014. Urocortin affects migration of hepatic cancer cell lines via differential regulation of cPLA2 and iPLA2. Cell. Signal.26: 1125–1134. [DOI] [PubMed] [Google Scholar]
  • 380.Hoeft B., Linseisen J., Beckmann L., Muller-Decker K., Canzian F., Husing A., Kaaks R., Vogel U., Jakobsen M. U., Overvad K., et al. 2010. Polymorphisms in fatty-acid-metabolism-related genes are associated with colorectal cancer risk. Carcinogenesis.31: 466–472. [DOI] [PubMed] [Google Scholar]
  • 381.Kvaskoff M., Whiteman D. C., Zhao Z. Z., Montgomery G. W., Martin N. G., Hayward N. K., Duffy D. L.2011. Polymorphisms in nevus-associated genes MTAP, PLA2G6, and IRF4 and the risk of invasive cutaneous melanoma. Twin Res. Hum. Genet.14: 422–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 382.Scuderi M. R., Anfuso C. D., Lupo G., Motta C., Romeo L., Guerra L., Cappellani A., Ragusa N., Cantarella G., Alberghina M.2008. Expression of Ca2+-independent and Ca2+-dependent phospholipases A2 and cyclooxygenases in human melanocytes and malignant melanoma cell lines. Biochim. Biophys. Acta.1781: 635–642. [DOI] [PubMed] [Google Scholar]
  • 383.Deleted in proof.
  • 384.Hazen S. L., Gross R. W.1993. The specific association of a phosphofructokinase isoform with myocardial calcium-independent phospholipase A2. Implications for the coordinated regulation of phospholipolysis and glycolysis. J. Biol. Chem.268: 9892–9900. [PubMed] [Google Scholar]
  • 385.McHowat J., Creer M. H.2004. Catalytic features, regulation and function of myocardial phospholipase A2. Curr. Med. Chem. Cardiovasc. Hematol. Agents.2: 209–218. [DOI] [PubMed] [Google Scholar]
  • 386.White M. C., McHowat J.2007. The therapeutic potential of phospholipase A2 inhibitors in cardiovascular disease. Cardiovasc. Hematol. Agents Med. Chem.5: 91–95. [DOI] [PubMed] [Google Scholar]
  • 387.Ford D. A., Hazen S. L., Saffitz J. E., Gross R. W.1991. The rapid and reversible activation of a calcium-independent plasmalogen-selective phospholipase A2 during myocardial ischemia. J. Clin. Invest.88: 331–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 388.Williams S. D., Gottlieb R. A.2002. Inhibition of mitochondrial calcium-independent phospholipase A2 (iPLA2) attenuates mitochondrial phospholipid loss and is cardioprotective. Biochem. J.362: 23–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 389.Kan H., Xie Z., Finkel M. S.2006. iPLA2 inhibitor blocks negative inotropic effect of HIV gp120 on cardiac myocytes. J. Mol. Cell. Cardiol.40: 131–137. [DOI] [PubMed] [Google Scholar]
  • 390.Rahnema P., Shimoni Y., Nygren A.2011. Reduced conduction reserve in the diabetic rat heart: role of iPLA2 activation in the response to ischemia. Am. J. Physiol. Heart Circ. Physiol.300: H326–H334. [DOI] [PubMed] [Google Scholar]
  • 391.Vesterqvist O., Sargent C. A., Grover G. J., Ogletree M. L.1996. Myocardial calcium-independent phospholipase A2 activity during global ischemia in isolated rabbit hearts. Cardiovasc. Res.31: 932–940. [PubMed] [Google Scholar]
  • 392.Zachman D. K., Chicco A. J., McCune S. A., Murphy R. C., Moore R. L., Sparagna G. C.2010. The role of calcium-independent phospholipase A2 in cardiolipin remodeling in the spontaneously hypertensive heart failure rat heart. J. Lipid Res.51: 525–534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 393.McHowat J., Swift L. M., Sarvazyan N.2001. Oxidant-induced inhibition of myocardial calcium-independent phospholipase A2. Cardiovasc. Toxicol.1: 309–316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 394.Sharma J., Turk J., Mancuso D. J., Sims H. F., Gross R. W., McHowat J.2011. Activation of group VI phospholipase A2 isoforms in cardiac endothelial cells. Am. J. Physiol. Cell Physiol.300: C872–C879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 395.Malhotra A., Edelman-Novemsky I., Xu Y., Plesken H., Ma J., Schlame M., Ren M.2009. Role of calcium-independent phospholipase A2 in the pathogenesis of Barth syndrome. Proc. Natl. Acad. Sci. USA.106: 2337–2341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 396.Kiebish M. A., Yang K., Liu X., Mancuso D. J., Guan S., Zhao Z., Sims H. F., Cerqua R., Cade W. T., Han X., et al. 2013. Dysfunctional cardiac mitochondrial bioenergetic, lipidomic, and signaling in a murine model of Barth syndrome. J. Lipid Res.54: 1312–1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 397.Smani T., Zakharov S. I., Leno E., Csutora P., Trepakova E. S., Bolotina V. M.2003. Ca2+-independent phospholipase A2 is a novel determinant of store-operated Ca2+ entry. J. Biol. Chem.278: 11909–11915. [DOI] [PubMed] [Google Scholar]
  • 398.Csutora P., Zarayskiy V., Peter K., Monje F., Smani T., Zakharov S. I., Litvinov D., Bolotina V. M.2006. Activation mechanism for CRAC current and store-operated Ca2+ entry: calcium influx factor and Ca2+-independent phospholipase A2beta-mediated pathway. J. Biol. Chem.281: 34926–34935. [DOI] [PubMed] [Google Scholar]
  • 399.Bolotina V. M.2004. Store-operated channels: diversity and activation mechanisms. Sci. STKE.2004: pe34. [DOI] [PubMed] [Google Scholar]
  • 400.Dietrich H. H., Abendschein D. R., Moon S. H., Nayeb-Hashemi N., Mancuso D. J., Jenkins C. M., Kaltenbronn K. M., Blumer K. J., Turk J., Gross R. W.2010. Genetic ablation of calcium-independent phospholipase A2beta causes hypercontractility and markedly attenuates endothelium-dependent relaxation to acetylcholine. Am. J. Physiol. Heart Circ. Physiol.298: H2208–H2220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 401.Balsinde J.2002. Roles of various phospholipases A2 in providing lysophospholipid acceptors for fatty acid phospholipid incorporation and remodelling. Biochem. J.364: 695–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 402.Balsinde J., Balboa M. A., Dennis E. A.2000. Identification of a third pathway for arachidonic acid mobilization and prostaglandin production in activated P388D1 macrophage-like cells. J. Biol. Chem.275: 22544–22549. [DOI] [PubMed] [Google Scholar]
  • 403.Hsu F. F., Ma Z., Wohltmann M., Bohrer A., Nowatzke W., Ramanadham S., Turk J.2000. Electrospray ionization/mass spectrometric analyses of human promonocytic U937 cell glycerolipids and evidence that differentiation is associated with membrane lipid composition changes that facilitate phospholipase A2 activation. J. Biol. Chem.275: 16579–16589. [DOI] [PubMed] [Google Scholar]
  • 404.Pindado J., Balsinde J., Balboa M. A.2007. TLR3-dependent induction of nitric oxide synthase in RAW 264.7 macrophage-like cells via a cytosolic phospholipase A2/cyclooxygenase-2 pathway. J. Immunol.179: 4821–4828. [DOI] [PubMed] [Google Scholar]
  • 405.Pérez-Chacón G., Astudillo A. M., Ruiperez V., Balboa M. A., Balsinde J.2010. Signaling role for lysophosphatidylcholine acyltransferase 3 in receptor-regulated arachidonic acid reacylation reactions in human monocytes. J. Immunol.184: 1071–1078. [DOI] [PubMed] [Google Scholar]
  • 406.Degousee N., Ghomashchi F., Stefanski E., Singer A., Smart B. P., Borregaard N., Reithmeier R., Lindsay T. F., Lichtenberger C., Reinisch W., et al. 2002. Groups IV, V, and X phospholipases A2s in human neutrophils: role in eicosanoid production and gram-negative bacterial phospholipid hydrolysis. J. Biol. Chem.277: 5061–5073. [DOI] [PubMed] [Google Scholar]
  • 407.Marshall J., Krump E., Lindsay T., Downey G., Ford D. A., Zhu P., Walker P., Rubin B.2000. Involvement of cytosolic phospholipase A2 and secretory phospholipase A2 in arachidonic acid release from human neutrophils. J. Immunol.164: 2084–2091. [DOI] [PubMed] [Google Scholar]
  • 408.Ueno N., Taketomi Y., Yamamoto K., Hirabayashi T., Kamei D., Kita Y., Shimizu T., Shinzawa K., Tsujimoto Y., Ikeda K., et al. 2011. Analysis of two major intracellular phospholipases A2 (PLA2) in mast cells reveals crucial contribution of cytosolic PLA2alpha, not Ca2+-independent PLA2beta, to lipid mobilization in proximal mast cells and distal fibroblasts. J. Biol. Chem.286: 37249–37263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 409.Roshak A. K., Capper E. A., Stevenson C., Eichman C., Marshall L. A.2000. Human calcium-independent phospholipase A2 mediates lymphocyte proliferation. J. Biol. Chem.275: 35692–35698. [DOI] [PubMed] [Google Scholar]
  • 410.Balboa M. A., Balsinde J.2002. Involvement of calcium-independent phospholipase A2 in hydrogen peroxide-induced accumulation of free fatty acids in human U937 cells. J. Biol. Chem.277: 40384–40389. [DOI] [PubMed] [Google Scholar]
  • 411.Deleted in proof.
  • 412.Martínez J., Moreno J. J.2001. Role of Ca2+-independent phospholipase A2 on arachidonic acid release induced by reactive oxygen species. Arch. Biochem. Biophys.392: 257–262. [DOI] [PubMed] [Google Scholar]
  • 413.Balboa M. A., Perez R., Balsinde J.2008. Calcium-independent phospholipase A2 mediates proliferation of human promonocytic U937 cells. FEBS J.275: 1915–1924. [DOI] [PubMed] [Google Scholar]
  • 414.Kim S. J., Gershov D., Ma X., Brot N., Elkon K. B.2002. I-PLA2 activation during apoptosis promotes the exposure of membrane lysophosphatidylcholine leading to binding by natural immunoglobulin M antibodies and complement activation. J. Exp. Med.196: 655–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 415.Teslenko V., Rogers M., Lefkowith J. B.1997. Macrophage arachidonate release via both the cytosolic Ca2+-dependent and -independent phospholipases is necessary for cell spreading. Biochim. Biophys. Acta.1344: 189–199. [DOI] [PubMed] [Google Scholar]
  • 416.Nikolic D. M., Gong M. C., Turk J., Post S. R.2007. Class A scavenger receptor-mediated macrophage adhesion requires coupling of calcium-independent phospholipase A2 and 12/15-lipoxygenase to Rac and Cdc42 activation. J. Biol. Chem.282: 33405–33411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 417.Pérez R., Balboa M. A., Balsinde J.2006. Involvement of group VIA calcium-independent phospholipase A2 in macrophage engulfment of hydrogen peroxide-treated U937 cells. J. Immunol.176: 2555–2561. [DOI] [PubMed] [Google Scholar]
  • 418.Karimi K., Gemmill T. R., Lennartz M. R.1999. Protein kinase C and a calcium-independent phospholipase are required for IgG-mediated phagocytosis by Mono-Mac-6 cells. J. Leukoc. Biol.65: 854–862. [DOI] [PubMed] [Google Scholar]
  • 419.Lennartz M. R., Lefkowith J. B., Bromley F. A., Brown E. J.1993. Immunoglobulin G-mediated phagocytosis activates a calcium-independent, phosphatidylethanolamine-specific phospholipase. J. Leukoc. Biol.54: 389–398. [DOI] [PubMed] [Google Scholar]
  • 420.Atkinson M. A., Bluestone J. A., Eisenbarth G. S., Hebrok M., Herold K. C., Accili D., Pietropaolo M., Arvan P. R., Von Herrath M., Markel D. S., et al. 2011. How does type 1 diabetes develop?: the notion of homicide or beta-cell suicide revisited. Diabetes.60: 1370–1379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 421.Satoh T., Abiru N., Kobayashi M., Zhou H., Nakamura K., Kuriya G., Nakamura H., Nagayama Y., Kawasaki E., Yamasaki H., et al. 2011. CHOP deletion does not impact the development of diabetes but suppresses the early production of insulin autoantibody in the NOD mouse. Apoptosis.16: 438–448. [DOI] [PubMed] [Google Scholar]
  • 422.Padgett L. E., Broniowska K. A., Hansen P. A., Corbett J. A., Tse H. M.2013. The role of reactive oxygen species and proinflammatory cytokines in type 1 diabetes pathogenesis. Ann. N. Y. Acad. Sci.1281: 16–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 423.Thayer T. C., Delano M., Liu C., Chen J., Padgett L. E., Tse H. M., Annamali M., Piganelli J. D., Moldawer L. L., Mathews C. E.2011. Superoxide production by macrophages and T cells is critical for the induction of autoreactivity and type 1 diabetes. Diabetes.60: 2144–2151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 424.Baumann B., Salem H. H., Boehm B. O.2012. Anti-inflammatory therapy in type 1 diabetes. Curr. Diab. Rep.12: 499–509. [DOI] [PubMed] [Google Scholar]
  • 425.Chan J. Y., Biden T. J., Laybutt D. R.2012. Cross-talk between the unfolded protein response and nuclear factor-κB signalling pathways regulates cytokine-mediated beta cell death in MIN6 cells and isolated mouse islets. Diabetologia.55: 2999–3009. [DOI] [PubMed] [Google Scholar]
  • 426.Blanc L., Barres C., Bette-Bobillo P., Vidal M.2007. Reticulocyte-secreted exosomes bind natural IgM antibodies: involvement of a ROS-activatable endosomal phospholipase iPLA2. Blood.110: 3407–3416. [DOI] [PubMed] [Google Scholar]
  • 427.Deleted in proof.
  • 428.Deleted in proof.
  • 429.Muralikrishna Adibhatla R., Hatcher J. F.2006. Phospholipase A2, reactive oxygen species, and lipid peroxidation in cerebral ischemia. Free Radic. Biol. Med.40: 376–387. [DOI] [PubMed] [Google Scholar]
  • 430.Osada-Oka M., Takahashi M., Akiba S., Sato T.2006. Involvement of Ca2+-independent phospholipase A2 in the translocation of hypoxia-inducible factor-1α to the nucleus under hypoxic conditions. Eur. J. Pharmacol.549: 58–62. [DOI] [PubMed] [Google Scholar]
  • 431.Deleted in proof.
  • 432.Sun G. Y., Xu J., Jensen M. D., Yu S., Wood W. G., Gonzalez F. A., Simonyi A., Sun A. Y., Weisman G. A.2005. Phospholipase A2 in astrocytes: responses to oxidative stress, inflammation, and G protein-coupled receptor agonists. Mol. Neurobiol.31: 27–41. [DOI] [PubMed] [Google Scholar]
  • 433.Tithof P. K., Peters-Golden M., Ganey P. E.1998. Distinct phospholipases A2 regulate the release of arachidonic acid for eicosanoid production and superoxide anion generation in neutrophils. J. Immunol.160: 953–960. [PubMed] [Google Scholar]
  • 434.Tan C., Day R., Bao S., Turk J., Zhao Q. D.2014. Group VIA phospholipase A2 mediates enhanced macrophage migration in diabetes mellitus by increasing expression of nicotinamide adenine dinucleotide phosphate oxidase 4. Arterioscler. Thromb. Vasc. Biol.34: 768–778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 435.Mollah Z. U., Pai S., Moore C., O’Sullivan B. J., Harrison M. J., Peng J., Phillips K., Prins J. B., Cardinal J., Thomas R.2008. Abnormal NF-kappa B function characterizes human type 1 diabetes dendritic cells and monocytes. J. Immunol.180: 3166–3175. [DOI] [PubMed] [Google Scholar]
  • 436.Devaraj S., Dasu M. R., Rockwood J., Winter W., Griffen S. C., Jialal I.2008. Increased toll-like receptor (TLR) 2 and TLR4 expression in monocytes from patients with type 1 diabetes: further evidence of a proinflammatory state. J. Clin. Endocrinol. Metab.93: 578–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 437.Deng J., Lu P. D., Zhang Y., Scheuner D., Kaufman R. J., Sonenberg N., Harding H. P., Ron D.2004. Translational repression mediates activation of nuclear factor kappa B by phosphorylated translation initiation factor 2. Mol. Cell. Biol.24: 10161–10168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 438.Jiang H. Y., Wek S. A., McGrath B. C., Scheuner D., Kaufman R. J., Cavener D. R., Wek R. C.2003. Phosphorylation of the alpha subunit of eukaryotic initiation factor 2 is required for activation of NF-kappaB in response to diverse cellular stresses. Mol. Cell. Biol.23: 5651–5663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 439.Morgan M. J., Liu Z. G.2011. Crosstalk of reactive oxygen species and NF-kappaB signaling. Cell Res.21: 103–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 440.Carnevale K. A., Cathcart M. K.2001. Calcium-independent phospholipase A2 is required for human monocyte chemotaxis to monocyte chemoattractant protein 1. J. Immunol.167: 3414–3421. [DOI] [PubMed] [Google Scholar]
  • 441.Gilroy D. W., Newson J., Sawmynaden P., Willoughby D. A., Croxtall J. D.2004. A novel role for phospholipase A2 isoforms in the checkpoint control of acute inflammation. FASEB J.18: 489–498. [DOI] [PubMed] [Google Scholar]
  • 442.Zhang L., Zhong S., Li Y., Ji G., Sundaram M., Yao Z.2013. Global inactivation of the Pla2g6 gene in mice does not cause dyslipidemia under chow or high-fat diet conditions. J. Cancer Prev.18: 235–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 443.Portilla D., Crew M. D., Grant D., Serrero G., Bates L. M., Dai G., Sasner M., Cheng J., Buonanno A.1998. cDNA cloning and expression of a novel family of enzymes with calcium-independent phospholipase A2 and lysophospholipase activities. J. Am. Soc. Nephrol.9: 1178–1186. [DOI] [PubMed] [Google Scholar]
  • 444.Portilla D., Dai G.1996. Purification of a novel calcium-independent phospholipase A2 from rabbit kidney. J. Biol. Chem.271: 15451–15457. [DOI] [PubMed] [Google Scholar]
  • 445.Mizuno-Kamiya M., Inokuchi H., Kameyama Y., Yashiro K., Fujita A.2001. Ca2+-independent phospholipase A2 activity in apical plasma membranes from the rat parotid gland. Arch. Oral Biol.46: 789–799. [DOI] [PubMed] [Google Scholar]
  • 446.Deleted in proof.
  • 447.Akiba S., Sato T.2004. Cellular function of calcium-independent phospholipase A2. Biol. Pharm. Bull.27: 1174–1178. [DOI] [PubMed] [Google Scholar]
  • 448.Hui D. Y.2012. Phospholipase A2 enzymes in metabolic and cardiovascular diseases. Curr. Opin. Lipidol.23: 235–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 449.Richardson R. J., Hein N. D., Wijeyesakere S. J., Fink J. K., Makhaeva G. F.2013. Neuropathy target esterase (NTE): overview and future. Chem. Biol. Interact.203: 238–244. [DOI] [PubMed] [Google Scholar]
  • 450.Dongiovanni P., Donati B., Fares R., Lombardi R., Mancina R. M., Romeo S., Valenti L.2013. PNPLA3 I148M polymorphism and progressive liver disease. World J. Gastroenterol.19: 6969–6978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 451.Dubuquoy C., Burnol A. F., Moldes M.2013. PNPLA3, a genetic marker of progressive liver disease, still hiding its metabolic function?Clin. Res. Hepatol. Gastroenterol.37: 30–35. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Lipid Research are provided here courtesy ofAmerican Society for Biochemistry and Molecular Biology

ACTIONS

RESOURCES


[8]ページ先頭

©2009-2025 Movatter.jp