Keywords

1 Introduction

Phospholipase A2 (PLA2) catalyzes the hydrolysis of the sn-2 position of membrane glycerophospholipids to liberate free fatty acids and lysophospholipids. To date, more than 30 enzymes that possess PLA2 or related activities have been identified in mammals, and these have been subdivided into several groups based on their structures, catalytic mechanisms, localizations. and evolutionary relationships. The cPLA2 family contains 6 enzymes (cPLA2α−ζ), which (except for cPLA2γ) contain an N-terminal C2 domain for Ca2+-dependent association with the membrane. The iPLA2 or patatin-like phospholipase domain-containing lipase (PNPLA) family includes 9 enzymes, some of which act principally on phospholipids and others on neutral lipids such as triglyceride (TG). The sPLA2 family, in which 10 catalytically active enzymes have been identified, are low molecular weight, extracellular enzymes that require Ca2+ of the mM order for optimal enzymatic activity. Because of this diversity, PLA2 enzymes have been implicated in various biological processes such as lipid mediator production, membrane remodeling, and energy metabolism.

During the past few decades, studies of various PLA2 transgenic and/or knockout mice as well as human diseases with PLA2 gene mutations have provided new insights into the emerging biological roles of individual PLA2s. The functions of individual PLA2s may not simply reflect changes in lipid mediator signaling, or more particularly eicosanoid signaling, but may also be attributable to hydrolysis of one or a combination of various target membrane lipids. Herein, we focus on the pathophysiology of various PLA2s as revealed by information from transgenic or knockout mice, as well as human diseases.

2 The cPLA2 Family

2.1 General Aspects of cPLA2s

Enzymes belonging to the cPLA2 family are characterized by the presence of a C2 domain in their N-terminal region, with the exception of cPLA2γ, which lacks this domain. Evolutionarily, the cPLA2 family emerged from the ancestral iPLA2 family at the branching point of vertebrates, correlating with the development of eicosanoid signaling cascades. cPLA2α is no doubt the best-known PLA2, with a major role in releasing arachidonic acid (AA), a precursor of eicosanoids (prostaglandins, PGs, and leukotrienes, LTs), from cellular membrane phospholipids.

2.2 cPLA2α

cPLA2α, also known as group IVA PLA2, is localized in the cytosol of resting cells, and in response to an increase in cytosolic Ca2+ levels after cell activation, it translocates to the perinuclear or, more specifically, the Golgi membranes to encounter its preferred substrate, AA-containing phosphatidylcholine (PC). Ceramide-1-phosphate or phosphoinositide-4,5-bisphosphate (PIP2) enhances the membrane interaction of cPLA2α. Mitogen-activated protein kinases phosphorylate Ser505 on cPLA2α, leading to its activation. The AA released by cPLA2α is then converted to PGs and LTs by cyclooxygenases and 5-lipoxygenase, respectively. As an example, the cPLA2α activation mechanism in mast cells (MCs) is shown in Fig. 2.1.

Fig. 2.1
figure 1

Activation of cytosolic PLA2 (cPLA2)α in mast cells (MCs). In response to Ca2+ influx following FcεRI activation with IgE and cognate antigen, cPLA2α translocates from the cytosol to the perinuclear membrane and is phosphorylated by mitogen-activated protein kinase (MAPK) for optimal activation. The arachidonic acid (AA) released from membrane phospholipids by cPLA2α is then converted to PGD2 by the sequential action of cyclooxygenase (COX)-1 (or COX-2 when the cells are primed by particular stimuli) and hematopoietic PGD2 synthase (H-PGDS) to PGD2 or by the sequential action of 5-lipoxygenase (5-LOX) incorporation with 5-LOX-activating protein (FLAP) and LTC4 synthase (LTC4S) to LTC4

Mice lacking cPLA2α (Pla2g4a −/−) exhibit a number of striking phenotypes that can be explained by defects in pathways involving PGs, LTs, or platelet-activating factor (PAF). For instance, Pla2g4a −/− mice are protected from asthma, acute respiratory distress syndrome, and pulmonary fibrosis, which can be explained by marked reductions of detrimental lipid mediators such as LTs and PAF [13]. Pla2g4a −/− mice or wild-type mice treated with a cPLA2α inhibitor are less susceptible to experimental autoimmune encephalomyelitis or collagen-induced arthritis [4, 5], are protected from brain injuries caused by ischemia or Aβ amyloid [6, 7], and have reduced incidences of intestinal and lung cancer [8, 9], all of which can be attributed to reduced PGE2 signaling. Consistent with the protective role of PGE2 in the gastrointestinal mucosa, the intestinal epithelium of Pla2g4a −/− mice has numerous small ulcerative lesions [9]. The impairment of female fertility observed in Pla2g4a −/− mice suggests that cPLA2α has an important role in parturition and implantation by providing PGF and PGE2 [10, 7]. Because of reduced production of thromboxane A2 (TXA2) by platelets, Pla2g4a −/− mice are protected from thromboembolism and have prolonged bleeding times [11]. Furthermore, ablation or knockdown of cPLA2α ameliorates metabolic disorders including atherosclerosis, hepatic fibrosis, insulin resistance, and adipose tissue inflammation [1214]. In all cases, the levels of lipid mediators responsible for the corresponding pathophysiological events are markedly reduced in Pla2g4a −/− mice relative to wild-type mice. In humans, an inherited PLA2G4A mutation is linked to impaired eicosanoid biosynthesis, ulceration of the small intestine, and platelet dysfunction [15].

2.3 Other cPLA2s

cPLA2β, δ, ε, and ζ (group IVB, IVD, IVE, and IVF PLA2s) map to the same chromosomal locus and are therefore evolutionally more related [16]. cPLA2β is a dual PLA1/PLA2 enzyme, although cPLA2δ has a robust PLA1 activity in preference to PLA2 activity. cPLA2γ (group IVC PLA2) is unique in that it lacks the C2 domain and displays lysophospholipase and transacylase activities in addition to PLA2 activity [17]. The in vivo functions of these cPLA2 isoforms are entirely unknown because knockout studies have yet to be performed. A recent study has shown that cPLA2ε may drive recycling through clathrin-independent endocytosis [18].

3 The iPLA2/PNPLA Family

3.1 General Aspects of iPLA2s

The human genome encodes nine iPLA2/PNPLA enzymes, which share a protein motif known as the “patatin domain” with an unusual folding topology that differs from classical lipases (Fig. 2.2). The cPLA2 and iPLA2 families seem to have evolved from a common ancestral gene, as their catalytic domains are commonly characterized by a three-layer α/β/α architecture employing a conserved Ser/Asp catalytic dyad instead of the classical catalytic triad [19]. iPLA2/PNPLA enzymes are found in virtually all eukaryotes including yeast, plants, invertebrates, and vertebrates, suggesting that they possess fundamental roles in cellular lipid metabolism conserved in the eukaryote kingdom. The designation “PNPLA” appears to be more appropriate than “iPLA2,” as some of the isoforms have enzymatic activities apparently distinct from bona fide PLA2 activity. For instance, iPLA2ζ/PNPLA2 functions as a major TG lipase in adipose and many other tissues, whereas iPLA2ε/PNPLA3 may act mainly as an acyltransferase or transacylase for accumulation of TG, particularly in the liver [20]. Here, we focus on two particular iPLA2s, iPLA2β/PNPLA9 and iPLA2γ/PNPLA8, which have robust PLA2 activity.

Fig. 2.2
figure 2

Evolutional relationship between the cPLA2 and iPLA2 families. The iPLA2 family is present in all eukaryotes, whereas the cPLA2 family emerged from the iPLA2 family at the stage of divergence of vertebrates

3.2 iPLA2β

iPLA2β (PNPLA9 or group VIA PLA2), the best characterized iPLA2, has long been thought to be involved in homeostatic phospholipid remodeling through deacylation of phospholipids in the Lands’ cycle. Indeed, the composition of phospholipids, particularly those containing docosahexaenoic acid (DHA), is noticeably altered in the brain (but not other tissues) of mice lacking iPLA2β (Pla2g6 −/−) [21]. Notably, human PLA2G6 mutations are associated with neurodegenerative diseases such as infantile neuroaxonal dystrophy (INAD), neurodegeneration with brain iron accumulation (NBIA), and Schindler’s disease, which share the distinctive pathological feature of axonal degeneration with spheroid bodies in the nervous system [22]. Similar neurodegenerative phenotypes are also evident in Pla2g6 −/− mice or Pla2g6 mutant mice (Pla2g6-inad, in which the Pla2g6 gene harbors a point mutation), which show motor dysfunction caused by widespread degeneration of axons and synapses, accompanied by the appearance of spheroids and vacuoles [23, 24]. iPLA2β has also been proposed to have more diverse signaling roles. These Pla2g6 −/− phenotypes include male infertility [25], defective opening of store-operated Ca2+ entry, probably caused by reduced production of lysophosphatidylcholine (LPC) [26], impaired insulin secretion by pancreatic β-cells [27], reduced apoptosis [28], decreased eicosanoid generation in vascular cells [29], and protection from ovarian cancer, possibly through reduction of lysophosphatidic acid (LPA) [30]. In most cases, however, the iPLA2β-driven lipid metabolic processes underlying these events are poorly characterized.

3.3 iPLA2γ

iPLA2γ, also known as PNPLA8 or group VIB PLA2, is localized to mitochondria or peroxisomes and displays PLA2 or PLA1 activity depending on the substrates involved [31]. Mice null for iPLA2γ (Pnpla8 −/−) exhibit bioenergetic dysfunctional phenotypes, including growth retardation, cold intolerance, reduced exercise endurance, increased mortality from cardiac stress, and abnormal mitochondrial function with an altered cardiolipin composition [32]. Furthermore, Pnpla8 −/− mice are resistant to diet-induced obesity, fatty liver, and hyperlipidemia [33, 34]. These mice also display lipodystrophy, impaired glucose-stimulated insulin secretion, and decreased mitochondrial β-oxidation. Myocardium-specific Pnpla8-transgenic mice show a dramatic reduction of myocardial phospholipid mass, marked accumulation of TG, impaired mitochondrial function, and hemodynamic dysfunction [35]. Thus, iPLA2γ appears to be crucial for maintaining efficient bioenergetic mitochondrial function by tailoring mitochondrial lipid metabolism. However, considering that defective β-oxidation usually leads to increased fat accumulation in peripheral tissues, the protective effect of iPLA2γ ablation against diet-induced metabolic disorders might involve an as yet unknown mechanism. Pnpla8 −/− mice also display a profound alteration in hippocampal mitochondrial homeostasis, leading to cognitive dysfunction [36]. The Pnpla8 −/− hippocampus has an increased level of cardiolipin and a decrease of plasmalogen, implying a function of iPLA2γ in remodeling of these phospholipids. Overall, the neurological abnormalities in Pnpla8 −/− mice are reminiscent of features in patients with Barth syndrome, a disease caused by disturbed cardiolipin metabolism [37].

4 The sPLA2 Family

4.1 General Aspects of sPLA2s

More than one third of the PLA2 enzymes belong to the sPLA2 family, which contains ten catalytically active isoforms (IB, IIA, IIC, IID, IIE, IIF, III, V, X, XIIA). Individual sPLA2s exhibit unique tissue and cellular localizations and enzymatic properties, suggesting their distinct pathophysiological roles. Classical group I/II/V/X sPLA2s are closely related, 14- to 19-kDa secreted enzymes with a highly conserved Ca2+-binding loop and a His/Asp catalytic dyad as well as conserved disulfide bonds, whereas group III and XII sPLA2s are atypical and classified into distinct classes. As sPLA2s are secreted, their target membranes should reside in the extracellular spaces. Individual sPLA2s contribute to various biological events through production of lipid mediators, promotion of membrane remodeling, modification of extracellular noncellular lipid components such as surfactant, microparticles, and lipoproteins, or degradation of foreign phospholipids such as those originating from microbes and dietary components. Here we overview the pathophysiological functions of several classical sPLA2s (IB, IIA, IID, IIE, V, X) and an atypical sPLA2 (group III), as revealed by their transgenic overexpression or gene targeting in mice. Several examples of these sPLA2-mediated lipid networks are illustrated in Fig. 2.3.

Fig. 2.3
figure 3

Several examples of sPLA2-driven lipid networks. (a) sPLA2-V in M2 macrophages facilitates the Th2 response and that in airway epithelial cells degrades lung surfactant. sPLA2-X from the airway epithelium acts on eosinophils to augment LTC4 generation. Accordingly, the two sPLA2s independently promote asthma. (b) sPLA2-III released from immature MCs acts on fibroblasts to promote L-PGDS-dependent generation of PGD2, which in turn acts on the PGD2 receptor DP1 on MCs to promote MC maturation. Accordingly, PLA2G3 facilitates MC-dependent anaphylaxis. Activation of cPLA2α in mature MCs is highlighted in Fig. 2.1. (c) In lymph nodes, sPLA2-IID in DCs hydrolyzes PE to release DHA, which is then converted to the pro-resolving lipid mediator resolving D1 (RvD1) to sequester Th1 immunity. Accordingly, sPLA2-IID ameliorates Th1-dependent contact dermatitis

4.2 sPLA2-IB

sPLA2-IB, often called “pancreatic sPLA2,” is abundantly expressed in the pancreas. After secretion into the duodenal lumen, an N-terminal heptapeptide of the inactive zymogen is cleaved by trypsin to yield an active enzyme. The main role of this enzyme is digestion of dietary and biliary phospholipids. Gene disruption of sPLA2-IB (Pla2g1b −/−) results in decreased phospholipid digestion and absorption in the gut [38]. The reduced intestinal production of LPC, which is a causal factor for hepatic insulin resistance, results in protection from diet-induced obesity, glucose intolerance, hyperlipidemia, and atherosclerosis in Pla2g1b −/− mice [3941].

4.3 sPLA2-IIA

sPLA2-IIA is the only isozyme detectable in the circulation, particularly under pathological conditions. It is often referred to as “inflammatory sPLA2” as its levels in sera or inflammatory exudates correlate with the severity of inflammatory diseases, and it is robustly induced by pro-inflammatory stimuli in various cells [42]. However, the precise role of sPLA2-IIA in inflammation remains debatable, as a natural mutation in its gene (Pla2g2a) in C57BL/6 and 129Sv mice [43] prevents adequate evaluation of its functions by gene targeting. So far, therefore, most of the in vivo functions of sPLA2-IIA have currently been addressed mainly using Pla2g2a-transgenic mice.

Pla2g2a-transgenic mice have skin abnormalities manifested by hair loss and epidermal hyperplasia [44] and by increased carcinogen-induced skin cancer [45]. In line with clinical evidence that the serum level of sPLA2-IIA correlates with cardiovascular diseases [46], Pla2g2a-transgenic mice develop advanced atherosclerotic lesions [47]. Given that atherosclerosis represents chronic inflammation in the aorta, sPLA2-IIA can be regarded as a pro-inflammatory enzyme in atherosclerosis. The most probable physiological role of sPLA2-IIA is degradation of bacterial membranes, thereby providing a first line of antimicrobial defense [48]. sPLA2-IIA is capable of hydrolyzing phosphatidylethanolamine (PE) and phosphatidylglycerol in marked preference to PC, which can account for the preferential action of this enzyme on bacteria rather than on mammalian cells. Accordingly, Pla2g2a-transgenic mice or wild-type mice treated with sPLA2-IIA are resistant to pneumonia and sepsis following bacterial infection [49]. For this reason, sPLA2-IIA is often referred to as a “bactericidal sPLA2.”

Mouse strains with natural disruption of the Pla2g2a gene (see foregoing) are more sensitive to intestinal tumorigenesis [50]. Transgenic transfer of the Pla2g2a gene into these strains reduces the incidence of intestinal polyposis [51]. Thus, sPLA2-IIA appears to have an antitumorigenic role in the gastrointestinal tract. Presumably, bactericidal sPLA2-IIA may affect the gastrointestinal microflora, thereby influencing tumor development. On the other hand, sPLA2-IIA expression is positively correlated with the malignancy of prostate cancer [52], revealing distinct impacts of sPLA2-IIA on different types of cancer. Recently, the mutated Pla2g2a allele in the C57BL/6 mouse strain was delivered into the BALB/c mouse strain to produce Pla2g2a −/− BALB/c mice. Autoantibody-induced arthritis is attenuated in these Pla2g2a −/− mice relative to Pla2g2a-sufficent mice, whereas it is conversely aggravated in Pla2g2a-transgenic mice [53]. This study has provided the first compelling evidence for the pro-inflammatory role of sPLA2-IIA. Recently, it has been shown that sPLA2-IIA targets phospholipids in extracellular mitochondria, and thereby amplifies inflammation by producing eicosanoids as well as mitochondrial DNA, a kind of danger-associated molecular pattern (DAMP) [54].

4.4 sPLA2-IID

sPLA2-IID is structurally most related to sPLA2-IIA and is expressed preferentially in dendritic cells (DCs) in secondary lymphoid organs [55], suggesting its immunoregulatory role. In a model of Th1-dependent contact dermatitis, resolution of inflammation is compromised in skin and lymph nodes of mice lacking sPLA2-IID (Pla2g2d −/−) [55]. sPLA2-IID in regional lymph nodes mobilizes a pool of polyunsaturated fatty acids that can be metabolized to pro-resolving lipid mediators such as DHA-derived resolvin D1, which reduces Th1 cytokine production and DC activation. sPLA2-IID preferentially hydrolyzes DHA-containing PE in lymph node membranes. In accordance with its antiinflammatory role, sPLA2-IID expression in DCs is downregulated after cell activation. Furthermore, administration of sPLA2-IID-Fc protein attenuates autoimmune diseases in mice [56]. Together, the existing data suggest that sPLA2-IID is a “resolving sPLA2” that ameliorates inflammation by mobilizing DHA-derived pro-resolving lipid mediators.

4.5 sPLA2-IIE

Similar to sPLA2-IID, sPLA2-IIE is structurally most homologous to sPLA2-IIA. Expression of sPLA2-IIE is markedly induced in adipocytes during adipogenesis in vitro and after high-fat feeding in vivo. Mice deficient in sPLA2-IIE (Pla2g2e −/−) are modestly protected from diet-induced obesity, fatty liver, and hyperlipidemia [57]. sPLA2-IIE preferentially hydrolyzes minor lipoprotein phospholipids, phosphatidylserine (PS), and PE, with no apparent fatty acid selectivity. As such, sPLA2-IIE alters lipid composition in lipoproteins, thereby affecting fat deposition in adipose tissue and liver. Thus, sPLA2-IIE is a “metabolic sPLA2” that controls systemic metabolic states by modulating lipoprotein phospholipids. These findings shed light on the importance of the minor lipoprotein phospholipids (PS and PE) in metabolic regulation.

4.6 sPLA2-V

Because sPLA2-V is able to hydrolyze PC more efficiently than sPLA2-IIA, most investigators in this research field have been interested in the potential roles of this enzyme in inflammation in the context of AA metabolism. Indeed, zymosan-induced peritonitis or lipopolysaccharide (LPS)-induced air pouch inflammation is partially attenuated in mice lacking sPLA2-V (Pla2g5 −/−) [58, 59]. sPLA2-V is highly expressed in the myocardium, and Pla2g5 −/− mice exhibit a markedly decreased infarct size in a myocardial ischemia and reperfusion model [60]. sPLA2-V is expressed in bronchial epithelial cells and alveolar macrophages, and Pla2g5 −/− mice are protected from airway disorders such as antigen-induced asthma and LPS-induced respiratory distress syndrome [61, 62]. Moreover, in keeping with the view that hydrolysis of phospholipids in low density lipoprotein (LDL) by sPLA2-V can promote foam cell formation by macrophages in vitro [63], Ldlr −/− mice transplanted with Pla2g5 −/− bone marrow cells are partially protected from atherosclerosis [64]. Although most of these studies support the offensive roles of sPLA2-V, the underlying mechanisms by which sPLA2-V regulates each of these pathologies have remained controversial. Of note, sPLA2-V prefers phospholipids bearing fatty acids with a lower degree of unsaturation (e.g., oleate and linoleate) to those containing highly polyunsaturated fatty acids (e.g., AA and DHA), making it unclear whether sPLA2-V indeed mobilizes AA-derived eicosanoids in vivo. Because increased inflammation is generally accompanied by cPLA2α activation, the observed changes in eicosanoid levels in Pla2g5 −/− mice might simply reflect disease-associated changes in cPLA2α activation, rather than hydrolytic liberation of AA by sPLA2-V. Indeed, transgenic overexpression of sPLA2-V leads to respiratory distress and neonatal death without alterations in pulmonary eicosanoid levels [65]. This phenotype has been ascribed to aberrant hydrolysis of surfactant phospholipids (dipalmitoyl-PC) and is apparently eicosanoid independent.

The roles of sPLA2-V in inflammation have been proven to be more complex. Although sPLA2-V was thought to be induced by pro-inflammatory stimuli (as in the case of sPLA2-IIA), it has recently become obvious that its expression is induced by the Th2 cytokines IL-4 and IL-13, rather than proinflammatory stimuli including LPS, zymosan, and Th1 cytokines, which decrease sPLA2-V expression [57, 66]. sPLA2-V is expressed in IL-4-driven M2 macrophages and Th2 cells, which facilitate Th2-type inflammation while attenuating Th1 or Th17 immunity. Importantly, Th2 responses, as monitored by IL-4 expression and IgE production, are greatly reduced in Pla2g5 −/− mice, thus underscoring the reduced asthmatic phenotype from the lack of sPLA2-V. Thus, sPLA2-V appears to function in at least two regulatory steps in asthma: (1) in antigen-presenting cells to regulate antigen processing and thereby the Th2 response, and (2) in airway-resident cells to promote airway inflammation that may involve surfactant degradation. Pla2g5 −/− mice are more susceptible to Candida infection (Th1 immunity) and arthritis (Th17 immunity) [53, 67], which could also be partly explained by the ability of sPLA2-V to promote Th2 immunity (and therefore to suppress Th1/Th17 immunity).

The function of sPLA2-V as a “Th2-prone sPLA2” also influences obesity, as Th2 or M2 response dampens adipose tissue inflammation. In obesity, sPLA2-V is induced in hypertrophic adipocytes [57]. When fed a high-fat diet, Pla2g5 −/− mice display hyperlipidemia with higher plasma levels of lipid-rich LDL and increased obesity, fatty liver, and insulin resistance. sPLA2-V has a protective function against metabolic disorders by hydrolyzing and thereby normalizing PC in LDL and by tipping the immune balance toward an Th2/M2 state that counteracts adipose tissue inflammation. Mechanistically, sPLA2-V-driven oleate and linoleate from PC in LDL dampen M1 macrophage polarization by saturated fatty acids (e.g., palmitate), probably through attenuation of endoplasmic reticulum stress. Clinically also, sPLA2-V expression in human visceral adipose tissue inversely correlates with plasma LDL levels. These studies underscore the physiological relevance of lipoprotein hydrolysis by sPLA2s, highlight two adipocyte-driven “metabolic sPLA2s” (sPLA2-IIE and sPLA2-V) as integrated regulators of immune and metabolic responses, and bring about a paradigm shift toward a better understanding of the roles of the sPLA2 family as a metabolic coordinator.

4.7 sPLA2-X

As in the case of sPLA2-IB, sPLA2-X is synthesized as a zymogen, and removal of an N-terminal pro-peptide produces an active mature enzyme [68]. Among the sPLA2s, sPLA2-X has the highest binding affinity for PC and thus exhibits the most potent ability to hydrolyze plasma membrane phospholipids in intact cells [69]. Because of this property, many investigators have speculated that sPLA2-X has a major role in inflammation. In line with this scenario, mice lacking sPLA2-X (Pla2g10 −/−) are refractory to antigen-induced asthma, with markedly reduced infiltration of eosinophils and lymphocytes, attenuated goblet cell hyperplasia and smooth muscle layer thickening, and decreased levels of Th2 cytokines and pro-asthmatic eicosanoids [70]. The attenuated asthmatic responses in Pla2g10 −/− mice are fully restored by knock-in of human sPLA2-X, and treatment of the knock-in mice with an inhibitor specific for human sPLA2-X suppresses airway inflammation [71]. Mechanistically, sPLA2-X secreted from the airway epithelium may act on infiltrating eosinophils to augment LT production in a process involving LPC-dependent activation of cPLA2α [72]. Pla2g10 −/− mice are also protected from the early phase of influenza infection [73], further highlighting the role of this enzyme in the airway. Moreover, sPLA2-X is one of the major sPLA2 isoforms detected in the airway of patients with asthma [74], thus directing attention to sPLA2-X as a novel therapeutic target for asthma. In contrast to sPLA2-V, however, sPLA2-X does not influence the Th2 response itself, as antigen-sensitized Pla2g10 −/− mice have normal IgE and IL-4 levels.

Several phenotypes have been reported for Pla2g10 −/− mice, but the data are controversial. These phenotypes include protection from myocardial infarction or aneurysm [75, 76], exacerbation or attenuation of atherosclerosis [77, 78], increased or decreased adiposity [79, 80], altered macrophage responses [81], and lower response to peripheral pain [79]. In some of these studies, experiments were performed under the assumption that sPLA2-X is expressed in immune cells such as neutrophils and macrophages. However, the expression of sPLA2-X in such immune cells is very low or almost undetectable [75, 79], raising a question as to the physiological relevance of studies involving adoptive transfer of Pla2g10 −/− bone marrow-derived cells. Rather, the possibility that paracrine sPLA2-X may alter the properties of inflammatory cells should be taken into account. Because sPLA2-X is abundantly expressed in the gut epithelium, it is likely that the decreased digestion and absorption of dietary and biliary phospholipids are eventually linked to reduced fat accumulation in adipose tissue of Pla2g10 −/− mice [79], a situation similar to Pla2g1b −/− mice (see foregoing).

sPLA2-X is most abundantly expressed in the testis, where it is stored in acrosomes (secretory granules) in the head of sperm cells [82]. Pla2g10 −/− spermatozoa display an impaired acrosome reaction and low fertility despite showing a normal number and motility [83, 82]. Thus, sPLA2-X plays a specific role in sperm activation, boosting the acrosome reaction by producing LPC from sperm membranes in a paracrine or autocrine manner. Last, a striking skin phenotype characterized by alopecia in Pla2g10-transgenic mice points to a unique role of sPLA2-X in hair homeostasis [84]. Although grossly the coat hairs of Pla2g10 −/− mice appear normal, they have ultrastructural abnormalities including a hypoplasic outer root sheath and reduced melanin granules in their hair follicles.

4.8 sPLA2-III

sPLA2-III, an atypical sPLA2, more closely resembles bee venom sPLA2 rather than other mammalian sPLA2s [85]. Transgenic overexpression of sPLA2-III in mice with an ApoE −/− background results in increased atherosclerosis from accelerated LDL hydrolysis and increased TXA2 synthesis [86]. These mice also develop systemic inflammation as they age because of elevated eicosanoid formation [87]. Thus, beyond the overexpression strategy, sPLA2-III has a pro-inflammatory potential.

sPLA2-III is highly expressed in the epididymal epithelium. Studies using mice lacking sPLA2-III (Pla2g3 −/−) have revealed that epididymal sPLA2-III acts on immature sperm cells passing through the duct in a paracrine manner to regulate phospholipid remodeling. During epididymal transit of spermatozoa, PC in the sperm membrane undergoes a dramatic shift in its acyl groups from oleate, linoleate, and AA to docosapentaenoic acid (DPA) and DHA, and the increased proportion of DPA/DHA consequently contributes to increased sperm membrane fluidity and thereby sperm motility. In Pla2g3 −/− mice, this sperm membrane remodeling is severely compromised. Accordingly, spermatozoa from Pla2g3 −/− mice have a low DPA/DHA content, aberrant acrosomes and flagella with an abnormal axoneme configuration, and display hypomotility and reduced fertility [88]. Thus, the two “reproductive sPLA2s” (sPLA2-III and sPLA2-X), which are expressed in different locations within the male genital organs, exert nonredundant but interrelated functions in two major steps of male fertility, the former during sperm maturation in the epididymis and the latter during capacitation and the acrosome reaction, likely after ejaculation in the uterus and oviduct.

Microenvironmental alterations in MC phenotypes affect susceptibility to allergy, yet the mechanisms underlying the proper maturation of MCs toward an allergy-sensitive phenotype were poorly understood. sPLA2-III is stored in and released from MC granules, and MC-associated passive and active anaphylactic responses are markedly attenuated in Pla2g3 −/− mice, whereas they are augmented in Pla2g3-transgenic mice [89]. Tissue MCs in Pla2g3 −/− mice are immature and are therefore resistant to IgE-dependent and -independent activation. Similar MC abnormalities are also seen in mice lacking lipocalin-type prostaglandin D2 (PGD2) synthase (L-PGDS) or those lacking the PGD2 receptor DP1, suggesting their functional relationship. Indeed, genetic or pharmacological ablation of DP1 in MCs or L-PGDS in fibroblasts phenocopies that of sPLA2-III in MCs in terms of defective MC maturation and anaphylaxis. Taken together, the data suggest that sPLA2-III secreted from immature MCs is coupled with fibroblastic L-PGDS to provide microenvironmental PGD2, which in turn promotes MC maturation via DP1. The sPLA2-III/L-PGDS/DP1 paracrine loop is a novel lipid-orchestrated mechanism, providing a missing microenvironmental cue that underlies the proper maturation of MCs.

5 Concluding Remarks

With the growing list of knockout and transgenic mouse strains for PLA2s, much progress has been made in delineating the physiological functions of each PLA2. It is now becoming obvious that cPLA2α is a central regulator of AA metabolism, supported by the view that the molecular evolution of cPLA2α coincided with that of eicosanoid receptors when vertebrates evolved, that the iPLA2 family is a fundamental regulator of membrane homeostasis and energy metabolism, and that individual sPLA2s exert unique and tissue-specific biological functions by acting on extracellular phospholipids, which include adjacent cell membranes, noncellular lipid components, and foreign phospholipids such as those in microbes and the diet. The diversity of target phospholipids and products may explain why each PLA2 family contains many isoforms. Further advances in this research field and their integration for therapeutic applications are likely to benefit from improved, time- and space-resolved lipidomics technology that will allow monitoring of individual PLA2s and their associated forms of lipid metabolism within specific tissue niches. Hopefully, the next decade will yield a comprehensive map of the PLA2-driven lipid networks, which will allow the therapeutic application of inhibitors for some PLA2s central to human diseases.