1,167
Views
19
CrossRef citations to date
0
Altmetric
Thematic Issue: 52nd International Conference on the Bioscience of Lipids

Do annexins participate in lipid messenger mediated intracellular signaling? A question revisited

, &
Pages 229-242 | Received 22 Feb 2012, Accepted 25 Apr 2012, Published online: 14 Jun 2012

Abstract

Annexins are physiologically important proteins that play a role in calcium buffering but also influence membrane structure, participate in Ca2+-dependent membrane repair events and in remodelling of the cytoskeleton. Thirty years ago several peptides isolated from lung perfusates, peritoneal leukocytes, neutrophiles and renal cells were proven inhibitory to the activity of phospholipase A2. Those peptides were found to derive from structurally related proteins: annexins AnxA1 and AnxA2. These findings raised the question whether annexins may participate in regulation of the production of lipid second messengers and, therefore, modulate numerous lipid mediated signaling pathways in the cell. Recent advances in the field of annexins made also with the use of knock-out animal models revealed that these proteins are indeed important constituents of specific signaling pathways. In this review we provide evidence supporting the hypothesis that annexins, as membrane-binding proteins and organizers of the membrane lateral heterogeneity, may participate in lipid mediated signaling pathways by affecting the distribution and activity of lipid metabolizing enzymes (most of the reports point to phospholipase A2) and of protein kinases regulating activity of these enzymes. Moreover, some experimental data suggest that annexins may directly interact with lipid metabolizing enzymes and, in a calcium-dependent or independent manner, with some of their substrates and products. On the basis of these observations, many investigators suggest that annexins are capable of linking Ca2+, redox and lipid signaling to coordinate vital cellular responses to the environmental stimuli.

Introduction

The annexins belong to a multigenic family of calcium-dependent, pH-sensitive, membrane-binding proteins which are present in all eukaryotic organisms examined to date, including plants (Konopka-Postupolska et al. Citation2011, Laohavisit and Davies Citation2011), yeasts (Morgan et al. Citation2004), lower eukaryotes such as parasites (Hofmann et al. Citation2010) and bacteria (Morgan et al. Citation2006). In mammals there are currently 12 identified genes encoding structurally related annexins (Gerke and Moss Citation2002). Within the annexin molecule unique calcium-binding sites embedded in the highly conserved annexin repeat motifs, building their conserved C-terminal core, have been identified (Gerke and Moss Citation2002). In addition to the C-terminal core annexins contain a variable N-terminal domain containing binding sites for various protein ligands, as well as sites for functionally relevant posttranslational modifications, such as proteolysis and phosphorylation (Gerke and Moss Citation2002).

It is a strong belief of many annexin investigators that the reason for annexin multiplicity in various organisms may lie in the large number and diversity of their functions. First of all, as proteins that bind membranes in a calcium-dependent and -independent manner, they integrate calcium signaling with membrane-related cellular events including membrane repair mechanisms (Draeger et al. Citation2011). Furthermore, they participate in vesicular traffic (Futter and White Citation2007) and various signaling pathways that lead to cell differentiation, migration, and proliferation (Mussunoor and Murray Citation2008, Grewal and Enrich Citation2009, Monastyrskaya et al. Citation2009b, Grewal et al. Citation2010). Annexins also contribute to the maintenance of cellular calcium homeostasis (Babiychuk and Draeger Citation2000, Gerke et al. Citation2005, Monastyrskaya et al. Citation2009a).

Some members of the annexin family are ubiquitously expressed and function as intracellular Ca2+ concentration ([Ca2+]c) sensors endowed with individual Ca2+-sensitivity (Monastyrskaya et al. Citation2009a, Citation2009b). The Ca2+ sensitivity of annexins is modulated by their interaction with other proteins (Gerke et al. Citation2005), especially those of the S100 family of Ca2+-binding proteins (Rescher et al. Citation2004, Gokhale et al. Citation2005, Hayes et al. Citation2009, Illien et al. Citation2010, Cmoch et al. Citation2011, Rezvanpour et al. Citation2011), and by proteolytic cleavage (Chung et al. Citation2004, Khau et al. Citation2011, Blume et al. Citation2012). Perhaps due to their variable calcium sensitivity, most cells contain multiple annexins (Monastyrskaya et al. Citation2007, Potez et al. Citation2011).

A growing number of evidence suggests that annexins exert their biological functions through influencing membrane dynamics, promoting membrane segregation and membrane fusion (Babiychuk and Draeger Citation2000, Gerke et al. Citation2005). Furthermore, annexins were found to participate in plasma membrane repair mechanisms by reacting to Ca2+ influx and rise in intracellular calcium concentration upon mechanical stress, and to the presence of toxins, pathogens, etc (Gerke et al. Citation2005, Draeger et al. Citation2011). In addition, annexins perform unique functions such as remodeling of cytoskeleton (Hayes et al. Citation2004a, Citation2006), and regulation of membrane ion conductance (Riquelme et al. Citation2004, Muimo Citation2009, Monastyrskaya et al. Citation2009a). Annexins have also been implicated in a range of pathologies, such as the progression of cancer, diabetes, the autoimmune disorder, anti-phospholipid syndrome and others frequently related to deregulated vesicular traffic (Hayes et al. Citation2007, Fatimathas and Moss Citation2010).

Originally, some annexins, such as annexin A7 (AnxA7), were reported to support vesicle aggregation (Chander et al. Citation2006). Later, the plasma membrane attachment, vesicle association and involvement in vesicular traffic have been shown for several other annexins, including AnxA1, AnxA2, AnxA5 and AnxA6. These proteins were described as participating in intracellular vesicle movement, formation of multivesicular bodies and endosomal functioning, including endocytic membrane traffic and the proper positioning of recycling endosomes (Futter et al. Citation1993, Gerke and Moss Citation2002, Mayran et al. Citation2003, Zobiack et al. Citation2003, Hayes et al. Citation2004b, White et al. Citation2006, Futter and White Citation2007, Lim and Pervaiz Citation2007, Morel et al. Citation2009). AnxA6, like AnxA1, has been associated with the EGF receptor signaling (Grewal and Enrich Citation2009, Vilá de Muga et al. Citation2009, Grewal et al. Citation2010).

For many years after their discovery, annexins were thought to bind to membrane anionic phospholipids, especially to phosphatidylserine (PS) and phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2 or PIP2) in a calcium-dependent manner (Gerke and Moss Citation2002, Hayes et al. Citation2009). Changes in intracellular calcium ion concentration were found to regulate the interaction of annexins with membranes. On the basis of these observations it was suggested that multiple annexins of different Ca2+ sensitivity present in a single cell, may interact with distinct plasma and/or internal membrane regions promoting processes important for cell function such as membrane segregation, membrane fusion and membrane repair, in response to membrane injury (Draeger et al. Citation2011). It is also worth mentioning calcium-independent annexin-membrane interactions, modulated by cholesterol, as these could be of particular importance in cellular signaling involving receptors localized in cholesterol and sphingomyelin-enriched membrane microdomains (Cornely et al. Citation2011).

A well described feature of annexins, i.e., their ability to interact with anionic phospholipids is commonly employed in the laboratory practice to recognize apoptotic cells characterized by the exposure of PS molecules on the external leaflet of the plasma membrane (Schutters and Reutelingsperger Citation2010). Biological relevance of identification and clearance of apoptotic cells could be related to the fact that this prevents the release of harmful cell contents thereby suppressing inflammation and autoimmune reactions (Rosenbaum et al. Citation2011). It is postulated that annexins may modulate cell removal by phagocytosis by acting as bridging molecules to PS, a characteristic phagocytosis signal of dying cells. Indeed, it has been shown that AnxA3, AnxA4, AnxA5 and AnxA13 can bind to PS in apoptotic cells, while the evolutionary younger AnxA8 cannot (Rosenbaum et al. Citation2011).

It must be emphasized that the repertoire of lipid molecules interacting with annexins is not limited to PS. It includes a relatively large number of phospholipids, such as phosphatidic acid (PA), phosphatidylinositol (PI), PtdIns(4,5)P2 (PIP2), fatty acids (arachidonic acid), ceramides and lipid-derived metabolites (Russo-Marie Citation1999, Monastyrskaya et al. Citation2009b, Cornely et al. Citation2011, Domon et al. Citation2012). Almost all of these molecules as second messengers are important constituents of lipid mediated signal transduction pathways that regulate vital cellular processes (Schug et al. Citation2012).

Lipid mediated transduction pathways can be defined as signaling events involving a lipid messenger that binds a protein target, which in turn exerts a specific effect on a given signaling pathway. Lipid messengers are not stored but rather are formed ‘on demand' at their site of action. They cannot circulate freely in solution but, rather, exist bound to special carrier proteins or remain within the membrane. In this respect, annexins which possess the ability to interact with amphipathic ligands and with membranes, seem to be well suited to play an intermediary role.

Since the discovery of annexins as molecules interfering with phospholipase A2 activity, a source of important lipid messenger molecules, more than 30 years ago (Flower and Blackwell Citation1979), the current literature has provided little updated information on this topic. Therefore, in this review we present available evidence concerning the participation of annexins in regulation of lipid signaling via combination of various mechanisms including: (1) regulation of membrane lateral heterogeneity thus providing appropriate environment for enzymes and proteins involved in signal transduction; (2) interference with the activity of phospholipase A2 and perhaps other phospholipases or lipid kinases by limiting their access to membranous substrates or directly interacting with these proteins; and (3) interaction with lipid mediators of signal transduction pathways.

The role of membrane lateral heterogeneity in lipid mediated signaling

One of the most intriguing feature of annexins, recently gaining attention of many laboratories worldwide, is their role in assembly, organization and stabilization of cholesterol and sphingomyelin enriched microdomains, frequently called rafts, which provide platforms for specific signaling pathways, as well as a proper environment for membrane enzymes and transport proteins, including ion channels and ion pumps (Cornely et al. Citation2011, Reverter et al. Citation2011, Domon et al. Citation2012). In addition, annexins were described as proteins whose intracellular distribution is affected by the distribution of cholesterol (Grewal et al. 2000, de Diego et al. Citation2002, te Vruchte et al. Citation2004, Grewal et al. Citation2010, Enrich et al. Citation2011); these proteins were found to co-localize with cholesterol at the plasma membrane and to follow intracellular cholesterol trafficking. Moreover, some evidence has been presented suggesting participation of annexins as cholesterol interacting proteins in the mechanism of formation of cholesterol-rich domains not only in the plasma membrane but also in membranes of the intracellular organelles (Ayala-Sanmartin et al. Citation2001, Lambert et al. Citation2004, Cubells et al. Citation2007, Domon et al. Citation2010, Vats et al. Citation2010, Cornely et al. Citation2011, Domon et al. Citation2012).

The opinion existing in the literature in the late '90s of the twentieth century about membrane binding properties of annexins and the consequences of this binding on membrane structure and properties, was based mostly on the results of in vitro experiments showing calcium-dependent binding of annexins to membranes and their ability to self-assemble on the membrane surface (Skrahina et al. Citation2008). These results led to the creation of the model depicted in . The model considers a calcium signal, i.e., the rise in [Ca2+]c concentration upon cell stimulation as a trigger of annexin interaction with membranes. At low resting [Ca2+]c annexins exist in the soluble state. Upon cell stimulation annexins bind to the membrane surface and self-associate. The authors of the model also considered the lateral reorganization of lipids in the cytosolic leaflet of the membrane into lipid-protein microdomains as an additional trigger for annexin-membrane interactions (Seaton and Dedman Citation1998). A consequence of this model is that the presence of organized arrays of surface-bound annexins alters membrane properties and the properties of other membrane-bound proteins and enzymes (Kaetzel and Dedman Citation1995, Seaton and Dedman Citation1998, Russo-Marie Citation1999, Kaetzel and Dedman Citation2004). Additionally, one may consider the role of such surface-bound annexin aggregates in the lateral organization of the membrane, right beneath the arrays of the protein.

Figure 1. Schematic visualization of annexins as regulators of formation of lipid second messengers interfering with the activity of cytosolic phospholipase A2 (PLA2) and other phospholipases, as well as mediators of the activity of various protein and lipid kinases (including proteins kinase C isoforms) playing regulatory roles in different lipid mediated signaling pathways. The scheme illustrates the idea formulated by Dedman and his co-workers (Kaetzel and Dedman Citation1995, Seaton and Dedman Citation1998) suggesting that upon a rise in intracellular [Ca2+]c annexin monomers (depicted in green), bind initially to certain lipid constituents of the inner leaflet of the plasma membrane, preferably to lipid microdomains enriched either in anionic phospholipids (such as phosphatidylserine, phosphatidylinositol or phosphatidylinositol 4,5-bisphosphate) or to cholesterol and sphingomyelin or ceramides (depicted in the figure in various colors). Then, annexins bound to the membranes interact with each other to form larger arrays on the membrane surface and, therefore, prevent the interaction of phospholipases, other lipid hydrolyzing enzymes, as well as protein and lipid kinases, with their substrates at the membrane. Other explanations are in the text. Redrawn from Kaetzel and Dedman (Citation1995), modified. This Figure is reproduced in color in the online version of Molecular Membrane Biology.

Figure 1. Schematic visualization of annexins as regulators of formation of lipid second messengers interfering with the activity of cytosolic phospholipase A2 (PLA2) and other phospholipases, as well as mediators of the activity of various protein and lipid kinases (including proteins kinase C isoforms) playing regulatory roles in different lipid mediated signaling pathways. The scheme illustrates the idea formulated by Dedman and his co-workers (Kaetzel and Dedman Citation1995, Seaton and Dedman Citation1998) suggesting that upon a rise in intracellular [Ca2+]c annexin monomers (depicted in green), bind initially to certain lipid constituents of the inner leaflet of the plasma membrane, preferably to lipid microdomains enriched either in anionic phospholipids (such as phosphatidylserine, phosphatidylinositol or phosphatidylinositol 4,5-bisphosphate) or to cholesterol and sphingomyelin or ceramides (depicted in the figure in various colors). Then, annexins bound to the membranes interact with each other to form larger arrays on the membrane surface and, therefore, prevent the interaction of phospholipases, other lipid hydrolyzing enzymes, as well as protein and lipid kinases, with their substrates at the membrane. Other explanations are in the text. Redrawn from Kaetzel and Dedman (Citation1995), modified. This Figure is reproduced in color in the online version of Molecular Membrane Biology.

In fact, membrane lipid and protein lateral distribution and their assembly into membrane microdomains of variable life-time, are tightly regulated processes (Lemmon Citation2008, Levental et al. Citation2010, Surma et al. Citation2011). A growing number of evidence suggests that the optimal function of many membrane proteins depends on their residence at the lipid rafts. Examples of these proteins include ion pumps (Baron et al. Citation2010) and ion channels such as human ether-à-go-go-related gene (HERG) (Ganapathi et al. Citation2010), and calcium-activated chloride channels (Sones et al. Citation2010), protein complexes involved in calcium entry (Orai1, TRPCs and STIM1) (Galan et al. Citation2010), membrane transporters (Mrp1 or ABCC1) (Hummel et al. Citation2011), membrane receptors (Lasley Citation2011), enzymes (NADPH oxidase subunits) (Zhang and Li Citation2010), as well as caveolin 1 (Lin et al. Citation2010), cadherins (Gentil-dit-Maurin et al. Citation2010), supervillin (Fang et al. Citation2010) and other proteins associated with various signaling pathways. As a consequence of such localization many vital signaling pathways are regulated at membrane microdomains where the signal complexes are assembled, organized and fine-tuned (Staubach and Hanisch Citation2011). Examples of such microdomain-dependent processes include signaling triggered by a member of the tumor necrosis factor (TNF) receptor family, CD40, expressed on a variety of immune and non-immune cells, and its ligand, CD154, playing a role in humoral and cell-mediated immunity (Nadiri et al. Citation2011). Activation of this signaling pathway causes changes in the compartmentalization of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway normally residing within plasma membrane microdomains. This, in turn, may lead to development of insulin resistance (Gao et al. Citation2011) and involvement of PI3K-associated cholesterol-enriched microdomains in CD204-mediated low density lipoprotein (LDL) uptake in human macrophages, that may be critical in inflammatory processes (Kiyanagi et al. Citation2011).

Despite the results of in vitro experiments (using liposomes, monolayer technique, solid supported membranes) showing interaction of some annexins (AnxA2, AnxA5, AnxA6 and AnxA13) with the lipid constituents of membrane microdomains, controversy still exists whether this process occurs in vivo and what its functional outcome might be (te Vruchte et al Citation2004, Gokhale et al. Citation2005, Grewal and Enrich Citation2009). The nature of the signal triggering annexin re-localization from the cytoplasm to the membranes, in particular microdomain-enriched regions, is not well understood either. Differential membrane binding ability of annexins is suggested to be the result of their different calcium and pH sensitivity, lipid and partner protein specificity, and posttranslational modifications (for review, see Domon et al. Citation2012).

Many members of the annexin family of proteins have been localized at membrane microdomains resembling rafts (Domon et al. Citation2012). This was observed for AnxA2 and its heterotetramer with the S100A10 protein (AnxA22S100A102); the latter is most probably implicated in microdomain-supported exocytosis of neurotransmitters and promotes the lateral association of glycosphingolipid- and cholesterol-enriched lipid microdomains into larger assemblies (Umbrecht-Jenck et al. Citation2010). The association of AnxA2 with lipid rafts was found to be influenced not only by intracellular [Ca2+] but also by N-terminal phosphorylation at Tyr23 residue of annexin. In addition it has been observed that the binding of AnxA2 to the lipid rafts is followed by the transport of protein along the endocytic pathway (Valapala and Vishwanatha Citation2011).

In neuroendocrine cells, AnxA2 and its heterotetramer were found to be implicated in the assembly of monosialotetrahexosylganglioside (GM1)-containing lipid microdomains as well as PIP2-enriched microdomains that are required for calcium-regulated exocytosis (Chasserot-Golaz et al. Citation2005). Furthermore, upon stimulation of adrenergic chromaffin cells the translocation of cytosolic AnxA2 to the plasma membrane, where it co-localizes with a member of the SNARE family of proteins, SNAP-25, and S100A10 (a partner protein of VAMP2), was observed, suggesting close relation between AnxA2-mediated lipid microdomains and SNAREs during exocytosis (Umbrecht-Jenck et al. Citation2010). In addition, AnxA2 and HIV-1 Gag interact at the PIP2-enriched membrane microdomains, sites of Gag-mediated viral assembly; this suggests that AnxA2 supports viral assembly, and functions in the late stages of HIV-1 replication (Harrist et al. Citation2009). AnxA13b, much like AnxA2, has been assigned to raft localization and functions in raft-dependent apical traffic in MDCK cells, as first described by Simons and co-workers (Lafont et al. Citation1998, Lecat et al. Citation2000) and further confirmed by other investigators (Astanina et al. Citation2010). AnxA8, another member of the family, was shown to bind, in a Ca2+-dependent manner, to F-actin-associated PIP2-enriched membrane microdomains in HeLa cells. The appearance of PIP2-enriched membrane microdomains seems to be stimulated upon infection with Escherichia coli (Goebeler et al. Citation2006). Similar observation was earlier made for AnxA2 (Zobiack et al. Citation2002).

The existing data on intracellular distribution and function of AnxA6 suggest that this protein, as a protein involved in the lateral organization of the plasma membrane, interacting with cholesterol and modulating the remodeling of actin cytoskeleton, is also involved in the vesicular transport along endocytotic and exocytotic pathways by interacting with the target membrane SNAP receptors (t-SNAREs) (Umbrecht-Jenck et al. Citation2010, Reverter et al. Citation2011) and working as a scaffold protein for several signaling proteins (Cornely et al. Citation2011). The best characterized to date is the Ca2+-dependent membrane targeting of p120GAP to down-regulate Ras activity (Enrich et al. Citation2011) and the stimulatory effect of AnxA6 on protein kinase C (PKC) important in the regulation of HRas and epidermal growth factor receptor (EGFR) signal transduction pathways (Grewal et al. Citation2010). One of the most important and intriguing observations is that AnxA6, by sequestering cholesterol in the late endosomes, as for example in skin fibroblasts of patients suffering from the Niemann-Pick type C disease (Grewal et al. Citation2000, Citation2010, de Diego et al. Citation2002, te Vruchte et al. Citation2004, Sztolsztener et al. Citation2010, Enrich et al. Citation2011), may contribute to lowering the levels of cholesterol in the Golgi and the plasma membrane. This leads to reduced cytoplasmic phospholipase A2 (cPLA2) activity, retention of caveolin in the Golgi apparatus and a reduced number of caveolae at the cell surface (Cubells et al. Citation2008).

The latter observation raises the question whether lipid microdomains play a role in regulation of signaling pathways in which phospholipases are engaged. It has been shown for example that phosphatidylcholine (PC)-specific phospholipase C (PC-PLC) selectively accumulates in the plasma membrane of HER2-overexpressing cells, where it co-localizes and associates with the EGFR family member, HER2, in raft domains. This may be of particular importance for targeting the molecular mechanisms controlling HER2 overexpression in the membrane of breast cancer cells by altering the rates of its endocytosis and lysosomal degradation (Paris et al. Citation2010). Furthermore, PI-specific PLC (PI-PLC) isoform δ1, which is localized in caveolae/rafts where it hydrolyzes PIP2 and initiates calcium mobilization and PKC activation, was found to be involved in noradrenaline-induced PIP2 hydrolysis and sustained contraction of vascular tissue (Clarke et al. Citation2008).

Taking together observations described in this paragraph it is worth to note the participation of annexins in the lateral organization of membranes, especially their ability to participate in the assembly and organization of lipid microdomains that constitute specific signaling platforms, may affect the function of lipid microdomain-associated proteins, including phospholipases. Bearing in mind that various phospholipases exert their activities also through lipid microdomains, in the next paragraph we address the question how annexins, either in a membrane-bound raft-associated or a soluble cytosolic form, may affect the functioning of phospholipases in certain signaling pathways in the cell.

Annexins as regulators of lipid-hydrolyzing enzyme activities

Cytosolic phospholipases A2

Phospholipases A2 release fatty acids (frequently arachidonic acid) from the second carbon of glycerol because they specifically recognize the sn-2 acyl bond of phospholipids. The superfamily includes several unrelated protein subfamilies with common enzymatic activity. The two most notable subfamilies are the secreted PLA2 (they require Ca2+ for their activity) and the cytosolic PLA2 (Dennis et al. Citation2011). The latter family is also Ca2+-dependent, but its members have a different structure (a C2 domain and a large catalytic domain) and are significantly larger than the secreted PLA2. Cytosolic PLA2 are involved in cell signaling processes, such as inflammatory responses. Arachidonic acid is both a signaling molecule and the precursor for other signalling molecules: eicosanoids (leukotrienes and prostaglandins). Other subfamilies include Ca2+-independent PLA2 (i PLA2) and lipoprotein-associated PLA2 (lp-PLA2), also known as platelet activating factor acetylhydrolases (PAF-AH) (Freedman et al. Citation2004, Saavedra et al. Citation2006, Leslie et al. Citation2010, Dennis et al. Citation2011).

Among many factors regulating their activity, the most important are phosphorylation (Hirabayashi et al. Citation2004), changes in [Ca2+], and interactions with various proteins, including vimentin (Hirabayashi et al. Citation2004), NADPH oxidase (Hirabayashi et al. Citation2004), PLA2-activating protein (PLAA) (Zhang et al. Citation2008) and annexins (Buckland and Wilton Citation2003). As for annexins, ever since the publication of Flower and Blackwell more than 30 years ago, describing for the first time a factor present in lung perfusate that was induced as a result of treatment with anti-inflammatory steroids and was an inhibitor of PLA2 activity (further identified as AnxA1) (Flower and Blackwell Citation1979, Blackwell et al. Citation1980, de Caterina et al. Citation1993, Hayashi et al. Citation1993, Croxtall et al. Citation1996a, Citation1996b, Citation1998), the dilemma over the mechanism by which annexins exert their inhibitory effect on PLA2 activity still persists. Various hypotheses were formulated so far including reduced availability of membrane substrates for PLA2 due to the presence of annexins (substrate depletion concept), an indirect mechanism in which signaling pathways leading to modulation of phospholipase activity are affected and, last but not least, a direct interaction of annexins with lipid-hydrolyzing enzymes (Buckland and Wilton Citation2000a, Citation2000b, Citation2003).

The first hypothesis considering substrate depletion or interfacial competition as a mechanism of PLA2 activity inhibition by annexins was formulated with respect to the ability of annexins to bind in a calcium-dependent manner to membranous anionic phospholipids, thus preventing PLA2 binding to the membrane (Kaetzel and Dedman Citation1995, Citation2004, Seaton and Dedman Citation1998). Furthermore, it has been postulated that membrane-bound annexins self-associate to form trimers that further assemble into sheets that cover the membrane surface and alter crucial properties of the membrane such as fluidity and permeability to metabolites and solutes. According to this concept, this submembranous annexin shield alters the functions of membrane integral proteins such as ion pumps and channels, and protects the membrane surface from phospholipid binding proteins such as cytosolic (or secreted) phospholipases and PKC isoforms (Kaetzel and Dedman Citation2004). This mechanism was analyzed in detail by Buckland and Wilton, and a positive conclusion has been reached concerning inhibition of the secreted PLA2 activity by annexins that could be best explained by competition of the two proteins for the same membrane surface (Buckland and Wilton Citation1998a, Citation2003). The same investigators asked this question in relation to cytosolic PLA2 activity (Buckland and Wilton Citation1998b, Citation2003). They stated that the regulation of cytosolic PLA2 was more complex and could involve both direct protein-protein interactions and indirect effects involving various signaling pathways.

The concept of interfacial competition was further extended into a hypothesis that annexins could interfere with the calcium and phospholipid signaling pathways by affecting the activity of various calcium-dependent and independent, anionic phospholipid-stimulated isoforms of PKC, and cytosolic PLA2 (Russo-Marie Citation1999).

Buckland and Wilton in their excellent review (Buckland and Wilton Citation2003) analyzed the glucocorticoid effects in relation to annexins and cPLA2 interactions. These authors discussed the potential roles of these interactions taking into account that glucocorticoids control up to 1% of the genome and thus exert profound physiological effects. These effects include regulation of the production of mediators of inflammation and related metabolic disturbances. The authors also provided numerous evidence showing that annexins could control PLA2 activity via protein-protein or protein-lipid interactions or through signalling cascades (Buckland and Wilton Citation2003). In this context, it is worth to note that annexins, especially AnxA1, may target the cytosolic PLA2 both by direct enzyme inhibition and by suppression of cytokine-induced activation of phospholipase. AnxA1 inhibits the expression and activity of other inflammatory enzymes such as the inducible nitric oxide synthase (iNOS) in macrophages and inducible cyclooxygenase (COX-2) in activated microglia (Parente and Solito Citation2004). This, together with the role of AnxA1 in cell migration (Meliton et al. Citation2008) and its neuroprotective effects (Liu et al. Citation2005), point to the complexity of cellular events affected by the interplay between annexins and cPLA2 (Chen et al. Citation2010).

Below we would like to focus on the results of experiments showing a direct interaction of annexins with PLA2 and its possible physiological consequences.

It has been proposed that cPLA2 activity is inhibited by AnxA1 by a direct interaction rather than by substrate depletion (Kim et al. Citation1994). By systematic approach it has been shown by the same investigators that inhibition of cPLA2 by a specific interaction is not a general function of all annexins. It was characteristic for AnxA1, N-terminally truncated AnxA1, heterotetramer AnxA22S100A102 and to a lesser extent for AnxA5, but not AnxA2 and AnxA3 (Kim et al. Citation2001a). The interaction site within the AnxA1 molecule was then identified using AnxA1 deletion mutants. It was observed that the deletion mutant AnxA1-(1–274), exhibited no cPLA2 inhibitory activity, whereas the deletion mutant AnxA1-(275–346), containing the C terminus, retained full activity. In addition, the AnxA1-(1–274) mutant did not interact with cPLA2 (Kim et al. Citation2001b).

The mechanism of direct interaction of the AnxA22S100A102 heterotetramer with PLA2 in terms of regulatory capacity is described for group IVA cPLA2α. Group IVA cPLA2α translocates to intracellular membranes, including the Golgi apparatus, in response to a rise in [Ca2+]c (Leslie et al. Citation2010). Group IVA cPLA2α has been reported to be phosphorylated at multiple serine residues, of which phosphorylation of the Ser727 residue was found to modulate protein-protein interactions. It has been shown that the AnxA22S100A102 heterotetramer but not S100A10 or AnxA2 alone, directly binds cPLA2α via Ser727. This interaction prevents binding of the phospholipase to the membrane and inhibits its activity. On the other hand phosphorylation of the Ser727 residue of PLA2 disrupts the interaction and activates cPLA2α () (Tian et al. Citation2008).

Figure 2. The activity of ubiquitous mammalian group IVA cPLA2 (cPLA2α) is tightly regulated by various factors, such as phosphorylation at multiple serine residues, [Ca2+], lipid mediators and various proteins, including annexin A2/S100A10 heterotetramer (AnxA22S100A102). A cPLA2α calcium-dependent binding to the plasma membrane is assured by the presence of an NH2-terminal C2 domain (C2) within the enzyme molecule. According to the mechanism proposed by Cho and his co-workers (Tian et al. Citation2008) upon rise in intracellular [Ca2+] AnxA22S100A102 heterotetramer interacts with cPLA2α (the binding occurs via the hydroxyl group of Ser727 (S727) of cPLA2α that forms hydrogen bonds with S100A10 in the heterotetramer) and thus prevents its binding to the membrane and inhibits the enzyme. Phosphorylation of the Ser727 residue affects the cPLA2α-AnxA22S100A102 interaction, thereby allowing phospholipase activation and subsequent lipid hydrolysis through binding to the membrane. Phosphorylation of another residue of cPLA2α, Ser505 at C2 domain, does not appear to influence the cPLA2α-AnxA22S100A102 interaction. On the Figure the lipid microdomain (pink) enriched in phosphatidylinositol 4,5-bisphosphate, a well known partner molecule for AnxA2, is depicted as a potential membrane target for the heterotetramer. Such domain could be stabilized by cholesterol molecules (Gokhale et al. Citation2005). Other explanations are in the text. Adapted from Tian et al. (Citation2008), with some modifications. This Figure is reproduced in color in the online version of Molecular Membrane Biology.

Figure 2. The activity of ubiquitous mammalian group IVA cPLA2 (cPLA2α) is tightly regulated by various factors, such as phosphorylation at multiple serine residues, [Ca2+], lipid mediators and various proteins, including annexin A2/S100A10 heterotetramer (AnxA22S100A102). A cPLA2α calcium-dependent binding to the plasma membrane is assured by the presence of an NH2-terminal C2 domain (C2) within the enzyme molecule. According to the mechanism proposed by Cho and his co-workers (Tian et al. Citation2008) upon rise in intracellular [Ca2+] AnxA22S100A102 heterotetramer interacts with cPLA2α (the binding occurs via the hydroxyl group of Ser727 (S727) of cPLA2α that forms hydrogen bonds with S100A10 in the heterotetramer) and thus prevents its binding to the membrane and inhibits the enzyme. Phosphorylation of the Ser727 residue affects the cPLA2α-AnxA22S100A102 interaction, thereby allowing phospholipase activation and subsequent lipid hydrolysis through binding to the membrane. Phosphorylation of another residue of cPLA2α, Ser505 at C2 domain, does not appear to influence the cPLA2α-AnxA22S100A102 interaction. On the Figure the lipid microdomain (pink) enriched in phosphatidylinositol 4,5-bisphosphate, a well known partner molecule for AnxA2, is depicted as a potential membrane target for the heterotetramer. Such domain could be stabilized by cholesterol molecules (Gokhale et al. Citation2005). Other explanations are in the text. Adapted from Tian et al. (Citation2008), with some modifications. This Figure is reproduced in color in the online version of Molecular Membrane Biology.

The results of subsequent in vitro experiments using Calu-3 cells and the plasmon surface resonance technique revealed interaction of S100A10 with CFTR (NBD1 domain), AnxA1 and cPLA2α. This was accompanied in Calu-3 cells by a partial redistribution of all four proteins into detergent resistant membranes induced by tumor necrosis factor-α (TNF-α), increased IL-8 synthesis, activation of cPLA2α and overproduction of eicosanoids. This suggests that the putative cPLA2α/AnxA1/S100A10/CFTR complex may participate in the regulation of the levels of inflammation mediators (Borot et al. Citation2009).

Using Madin-Darby canine kidney (MDCK) type II cells it has been observed that the stoichiometry of S100A10-cPLA2 interaction may be controlled by cell confluence, therefore the phospholipase activation can be controlled by the establishment of cell-cell contacts (Bailleux et al. Citation2004). In addition, it has been found that the confluence-dependent interaction of cPLA2α and AnxA1 at the Golgi membranes in endothelial cells may act as a molecular switch controlling cPLA2α activity and endothelial cell prostaglandin generation (Herbert et al. Citation2007).

In Chinese hamster ovary (CHO) cells overexpressing AnxA6, sequestration of cholesterol in late endosomes led to reduced amounts of cholesterol and cPLA2 in the Golgi membranes, and to inhibition of cPLA2 activity. This correlated with an impairment of caveolin export suggesting that AnxA6 interfered with caveolin transport through inhibition of cPLA2 activity; (Cubells et al. Citation2008). It is suggested that the AnxA6/cPLA2 interaction may regulate vital physiological processes including proliferation, differentiation, inflammation and cell migration although a direct interaction between AnxA6 and cPLA2 in the cell has not been demonstrated (Grewal et al. Citation2010).

The importance of potential cPLA2/annexin interactions in pathological processes is supported by observations suggesting that in prostate cancer cPLA2 function is affected by aberrant upregulation of the secreted enzymes and concomitant downregulation of their endogenous inhibitors: AnxA1 and AnxA2 (Dong et al. Citation2006).

Moreover, it was reported that in normal human keratinocytes (NHK), Ca2+-dependent binding of S100A11 to its partner annexin, AnxA1, facilitated the binding of the latter to cPLA2, resulting in inhibition of cPLA2 activity, which is essential for the growth of NHK cells. Upon NHK exposure to the epidermal growth factor, AnxA1 was cleaved at the Trp12 residue by cathepsin D, and this was accompanied by growth inhibition (Sakaguchi et al. Citation2007).

Other phospholipases

In contrast to the cytoplasmic and secretory PLA2 isoforms, the number of reports related to the possible effects of annexins on phospholipase C (PLC), phospholipase D (PLD) and sphingomyelinase (SMase) activities is very limited. The reports mostly refer to the molecular events that are related to the potential role of annexins in the formation of membrane microdomains that provide a proper environment for the lipid hydrolysing enzymes. No data supporting a direct interaction of annexins with these enzymes is available so far. Therefore, further studies are required to answer the question whether annexins may modulate the activity of other lipid-hydrolysing enzymes. A brief analysis of the properties of these enzymes, i.e., their intracellular localization, substrates and interacting partners, makes the presumption about annexins as important factors for lipid-hydrolyzing enzymes very likely.

Phospholipases C belong to a class of enzymes that cleave phospholipids into diacylglycerol (DAG) and phosphoethanolamine or phosphocholine. When PIP2 is subjected to digestion, DAG and inositol 1,4,5-trisphosphate (IP3) are produced. In this respect, PLC is a key enzyme regulating intracellular calcium homeostasis and modulating phosphoinositide balance. Thirteen isoforms of mammalian PLC are classified into six isotypes (β, γ, δ, ε, ζ, η) (Fukami et al. Citation2010). In the next paragraph the ability of annexins to interact with phosphatidylinositol and phosphoinositides will be analyzed in conjunction with the PLC activity and cellular functions. Here, we refer to a spectacular example of biological activity of AnxA7 in the alveolar type II cells responsible for secretion of lung surfactants. It has been shown that the membrane fusion activity of AnxA7 involved in the fusion of lamellar body membranes with the plasma membrane during the secretory process is greatly modulated when DAG and PIP2, the product and the substrate of PLC, respectively, are present (Chander et al. Citation2007).

Phospholipases D (PLD) are enzymes which are located in the plasma membrane and catalyze the hydrolysis of glycerophospholipids and PA (McDermott et al. Citation2004). Phosphatidylcholine-specific PLD isoforms cleave PC to form PA and a soluble choline head group which is released into the cytoplasm. Two mammalian isoforms of phospholipase D: PLD1 and PLD2, were identified. Mammalian PLD directly interacts with several kinases including PKC isoforms and control signaling pathways mediated by these kinases. PLD isoforms are activated by ADP-ribosylation factor (Arf) (Selvy et al. Citation2011). In the course of studies using neutrophiles several PA binding proteins were identified including vesicle amine transport protein-1 (VAT-1), AnxA3, Rac2, Cdc42 and RhoG, suggesting the role of PLD-derived PA in cell physiology, also in processes that are annexin-dependent (Faugaret et al. Citation2011).

SMase, especially the acid SMase (also known as sphingomyelin phosphodiesterase-1), plays an important role in normal membrane turnover through the hydrolysis of sphingomyelin (SM), and is one of the key enzymes responsible for the production of ceramide (Schiffman Citation2010, Schuchman Citation2010). Some results of biochemical studies and studies using SMase knockout mice suggest that acid SMase may be involved in the pathogenesis of several common diseases through its ability to reorganize membrane microdomains (Claus et al. Citation2009, Jin and Zhou Citation2009, Schuchman Citation2010). It has been reported that the Niemann-Pick disease type A or B, a lysosomal storage disorder characterized at the cellular level by accumulation of SM within the late endosome/lysosome compartment, may arise because of the dysfunction and deficit of acid SMase as a result of a mutation in the enzyme encoding gene, SMPD-1 (Jenkins et al. Citation2009, Tamasawa et al. Citation2012). Although highly speculative at this stage it is possible that the effect of SMase deficit could be related to the proposed role of annexins in membrane microdomain organization and their involvement in several pathologies related to deregulated intracellular vesicular transport and storage of metabolites (Monastyrskaya et al. Citation2009b, Grewal et al. Citation2010, Enrich et al. Citation2011, Domon et al. Citation2012).

Lipid-derived second messengers as ligands for annexins

Phosphatidylinositol-derived second messengers

In the course of in vitro studies numerous evidence have been presented that annexins bind to anionic phospholipids (for review see Babiychuk and Draeger Citation2000, Gerke and Moss Citation2002, Gerke et al. Citation2005, Draeger et al. Citation2011), including phosphatidylinositol and an important second messenger, phosphatidylinositol 3,4-bisphosphate (PI(4,5)P2) (Rescher et al. Citation2004, Hayes et al. Citation2004a, Citation2004b, Citation2009). Data using mutated proteins revealed that individual annexin repeat domains may have characteristic affinities for different lipids, including PI(4,5)P2 (Sohma et al. Citation2001). In the cell, the PI(4,5)P2 messenger system consists of the G-protein coupled receptors and two effectors, PI-PLC and phosphoinositide 3-kinase (PI3K). PI-PLC produces two different second messengers, inositol trisphosphate (IP3) and diacylglycerol (DAG). IP3 diffuses freely into the cytoplasm and is recognized by the inositol trisphosphate receptor (IP3R) family in the endoplasmic reticulum membrane. Thus IP3 participates in the overall intracellular calcium homoeostasis. DAG remains bound to the membrane where it recruits and activates both conventional and non conventional members of the PKC family of protein kinases (Nishizuka Citation1995). PI3K, as an effector enzyme in the system, phosphorylates PIP2 to phosphatidylinositol (3,4,5)-trisphosphate (PI(3,4,5)P3) (Kanaho and Suzuki Citation2002). PI3K belongs to a family of phosphoinositide kinases that catalyze the phosphorylation of PI and its phosphorylated forms to produce phosphoinositides. These phosphoinositides have specific physiological functions, such as reorganization of actin cytoskeleton, vesicular transport, cell proliferation and survival (Kanaho and Suzuki Citation2002).

Among different annexins, AnxA2 was identified as a phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2)-interacting protein, targeted together with its ligand, the S100A10 protein, to specific PI(4,5)P2-enriched membrane microdomains where also F-actin was found to accumulate. Membrane-associated AnxA2 may play a role in membrane-associated actin assembly and bacterial adhesion during bacteria invasion. In vitro the AnxA22S100A102 heterotetramer binds to PI(4,5)P2 with an affinity comparable to that of the PH domain of PLCδ1 (Rescher et al. Citation2004). Moreover, AnxA2 was stimulating macropinocytic rocketing in a calcium-dependent manner in fibroblasts from patients with Lowe Syndrome. This suggests that AnxA2 plays a role of an actin nucleator on PI(4,5)P2-enriched membranes (Hayes et al. Citation2009). Recombinant AnxA2 binds to PI(4,5)P2, but not to other poly- and monophosphoinositides, with affinity comparable to that of other PI(4,5)P2-binding proteins (Hayes et al. Citation2004a, Citation2004b). It has been observed that in pro-B cell acute lymphoblastic leukemia cells AnxA2 expression was regulated by the Ras/PI3K pathway (Matsunaga et al. Citation2004).

In epithelium PI(4,5)P2-AnxA2 interactions were found to be important in the generation of an apical region of the plasma membrane, and thereby in integrating cellular polarity with epithelial architecture (Martin-Belmonte and Mostov Citation2007). Furthermore, AnxA2 is recruiting other proteins such as GTPase Cdc42 and aPKC into this region of the plasma membrane where PTEN (phosphatase and tensin homolog deleted on chromosome ten), a phosphatase of a tumor suppressor function which plays a role in dephosphorylation of PI(3,4,5)P3 to PI(4,5)P2, is also residing (Choi et al. Citation2002, Mahimainathan and Choudhury Citation2004). PTEN was found to be present in the apical region of the plasma membrane. Its malfunction prevented development of the apical domain of the plasma membrane (Martin-Belmonte et al. Citation2007).

Whether annexins may participate in regulation of kinases which phosphorylate various forms of PI remains to be elucidated. However, the data obtained so far on the skeletal muscle triad system responsible for propagation of the calcium signal point to the fact that AnxA6, despite its capacity to bind to negatively charged phospholipids including PI and PI(4,5)P2, does not affect the kinase activities responsible for their generation (Barrientos and Hidalgo Citation2002). On the other hand, 1α,25-dihydroxyvitamin D3, via binding to its membrane receptor, AnxA2, induces activation of the PI3K/Ras/MEK/extracellular signal regulated kinase 1/2 and c-Jun N-terminal kinase 1 signal transduction pathway resulting in increased expression of c-Fos, Fra1, and c-Jun, and subsequently, increased activator protein 1 DNA binding activity and gene transcription (Johansen et al. Citation2003).

Other lipid-derived second messengers

Information about interaction of annexins with other lipid-derived second messengers such as lysophosphatidic acid (LPA), platelet activating factor (PAF), prostaglandins, leukotrienes, sphingosine-1-phosphate (S1P), ceramide and ceramide-1-phosphate, glucosylceramide, as well as retinol derivatives, endocannabinoids, and steroid hormones is very limited. Interaction of annexins with membrane regions enriched in precursors of the above mentioned compounds, i.e., specific lipids including sphingomyelin, phosphatidic acid, phospholipids acylated in the sn-2 position with polyunsaturated fatty acids, other anionic phospholipids, etc. (Zschörnig et al. Citation2007, Monastyrskaya et al. Citation2009b, Draeger et al. Citation2011, Domon et al. Citation2011, Citation2012), makes this presumption very likely. Moreover, steroid hormones are synthesized from isoprenoids and structurally resemble cholesterol, a molecule identified as a ligand for annexins (Domon et al. Citation2010, Citation2012).

Annexins may participate in the metabolism of arachidonic acid derivatives, such as leukotrienes, which are produced by 5-lipoxygenase. For example, it has been shown that AnxA7 possesses a tumor suppressor function demonstrated by cancer susceptibility of ANX7 +/- mice and by the loss of ANXA7 expression in human cancers (Torosyan et al. Citation2009). It was observed that AnxA7, by regulating 5-lipoxygenase transcription that is potentially relevant to the arachidonic acid-mediated cell growth control in tumor suppression, is associated with the arachidonic acid cascade (Torosyan et al. Citation2006).

In addition, it has been shown that vitamin A (all-trans retinol) and all-trans retinoid acid interact in vitro with recombinant human AnxA6, and stimulated the calcium-dependent binding of AnxA6 to liposomes, accompanied by oligomerization of AnxA6. This may support the hypothesis of a direct implication of AnxA6 in vitamin A-dependent tissue mineralization (Balcerzak et al. Citation2006).

Conclusions and future perspectives

In this review the possible participation of annexins in lipid metabolism, with a special focus on the metabolism of secondary lipid messengers, has been revisited. Our renewed interest in this subject is due to the growing number of observations suggesting that annexins, as membrane-binding proteins, participate in the regulation of membrane lateral organization, assembly of enzyme and signaling complexes, vesicular traffic, membrane repair mechanisms, and, therefore, may play a role in lipid-mediated signal transduction. As a consequence of these functions, deregulation of annexin expression or mistargeting of these proteins in the cell accompany or may directly lead to development of serious pathologies, including certain types of cancer or lipid storage diseases, such as the Niemann-Pick type C disease. Identification of other diseases in which malfunction of annexins is the pathological factor is only a question of time.

The focus of the review is on lipid-hydrolyzing enzymes, especially PLA2 isoforms, that regulate the amount of important lipid messengers, or of their precursors, such as fatty acids, especially arachidonic acid. Of equal importance are also the secondary products of the reaction catalyzed by PLA2, i.e., lysophospholipds that are gaining attention of many investigators as potent biologically active lipid mediators that exert a wide range of cellular effects through specific G protein-coupled receptors. These receptors are expressed in a large number of tissues and cell types, allowing for a wide variety of cellular responses including cell adhesion, cell motility, reorganization of cytoskeleton, proliferation, angiogenesis and cell survival (Rivera and Chun Citation2008). Their activation initiates different intracellular pathways that induce profound changes in [Ca2+], intracellular pH and production of lipid second messengers (Monastyrskaya et al. Citation2009b). In this respect, annexins, as Ca2+-sensitive proteins binding to a large variety of phospholipids, cholesterol and lipid-derived molecules, and playing important roles in organization and function of lipid microdomains, are key elements of the cellular machinery responsible for signal transduction during stress response. In summary, on the basis of available data, a still complex picture is emerging as to the relationship between annexins and PLA2. It is strongly believed that detailed studies of cell lines (Solito et al. Citation2001, Croxtall et al. Citation2003) and knockout animals (especially ANXA1 -/- and ANXA2 -/-) (Bensalem et al. Citation2005, Damazo et al. Citation2007, Ayoub et al. Citation2008, Flood and Hajjar, Citation2011, He et al. Citation2011) should clarify the role of the annexin/phospholipase interaction in regulation of the amount as well as spatial and temporal distribution of lipid-derived second messengers in the cell.

Acknowledgements

The annexin project in the authors' laboratories is supported in part by a grant NN401642740 from the National Science Center to JBP, and by the Nencki Institute of Experimental Biology statutory grant to SP. The authors would like to apologize for not being able to cite all original articles related to the topic, due to space limitations.

Declaration of interest: The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

References

  • Astanina K, Delebinski CI, Delacour D, Jacob R. 2010. Annexin XIIIb guides raft-dependent and -independent apical traffic in MDCK cells. Eur J Cell Biol 89:799–806.
  • Ayala-Sanmartin J, Henry JP, Pradel LA. 2001. Cholesterol regulates membrane binding and aggregation by annexin 2 at submicromolar Ca2+ concentration. Biochim Biophys Acta 1510:18–28.
  • Ayoub SS, Yazid S, Flower RJ. 2008. Increased susceptibility of annexin-A1 null mice to nociceptive pain is indicative of a spinal antinociceptive action of annexin-A1. Br J Pharmacol 154:1135–1142.
  • Babiychuk EB, Draeger A. 2000. Annexins in cell membrane dynamics. Ca2+–regulated association of lipid microdomains. J Cell Biol 150:1113–1124.
  • Bailleux A, Wendum D, Audubert F, Jouniaux AM, Koumanov K, Trugnan G, 2004. Cytosolic phospholipase A2-p11 interaction controls arachidonic acid release as a function of epithelial cell confluence. Biochem J 378:307–315.
  • Balcerzak M, Bandorowicz-Pikula J, Buchet R, Pikula S. 2006. A novel retinoid binding property of human annexin A6. FEBS Lett 580:3065–3069.
  • Baron S, Vangheluwe P, Sepúlveda MR, Wuytack F, Raeymaekers L, Vanoevelen J. 2010. The secretory pathway Ca2+-ATPase 1 is associated with cholesterol-rich microdomains of human colon adenocarcinoma cells. Biochim Biophys Acta 1798:1512–1521.
  • Barrientos G, Hidalgo C. 2002. Annexin VI is attached to transverse-tubule membranes in isolated skeletal muscle triads. J Membr Biol 188:163–173.
  • Bensalem N, Ventura AP, Vallée B, Lipecka J, Tondelier D, Davezac N, 2005. Down-regulation of the anti-inflammatory protein annexin A1 in cystic fibrosis knock-out mice and patients. Mol Cell Proteomics 4:1591–1601.
  • Blackwell GJ, Carnuccio R, Di Rosa M, Flower RJ, Parente L, Persico P. 1980. Macrocortin: A polypeptide causing the anti-phospholipase effect of glucocorticoids. Nature 287:147–150.
  • Blume KE, Soeroes S, Keppeler H, Stevanovic S, Kretschmer D, Rautenberg M, 2012. Cleavage of annexin A1 by ADAM10 during secondary necrosis generates a monocytic ‘find-me' signal. J Immunol 188:135–145.
  • Borot F, Vieu DL, Faure G, Fritsch J, Colas J, Moriceau S, 2009. Eicosanoid release is increased by membrane destabilization and CFTR inhibition in Calu-3 cells. PLoS One 4:e7116.
  • Buckland AG, Wilton DC. 1998a. Inhibition of secreted phospholipases A2 by annexin V. Competition for anionic phospholipid interfaces allows an assessment of the relative interfacial affinities of secreted phospholipases A2. Biochim Biophys Acta 1391:367–376.
  • Buckland AG, Wilton DC. 1998b. Inhibition of human cytosolic phospholipase A2 by human annexin V. Biochem J 329:369–372.
  • Buckland AG, Wilton DC. 2000a. Anionic phospholipids, interfacial binding and the regulation of cell functions. Biochim Biophys Acta 1483:199–216.
  • Buckland AG, Wilton DC. 2000b. The antibacterial properties of secreted phospholipases A2. Biochim Biophys Acta 1488:71–82.
  • Buckland AG, Wilton DC. 2003. Annexins and phospholipases. In: Bandorowicz-Pikula J, editor. Annexins: Biological importance and annexin-related pathologies. New York: Kluwer Academic/Plenum Publishers, Landes Bioscience. pp 207–217.
  • Chander A, Naidu DG, Chen XL. 2006. A ten-residue domain (Y11-A20) in the NH2-terminus modulates membrane association of annexin A7. Biochim Biophys Acta 1761:775–784.
  • Chander A, Chen XL, Naidu DG. 2007. A role for diacylglycerol in annexin A7-mediated fusion of lung lamellar bodies. Biochim Biophys Acta 1771:1308–1318.
  • Chasserot-Golaz S, Vitale N, Umbrecht-Jenck E, Knight D, Gerke V, Bader MF. 2005. Annexin 2 promotes the formation of lipid microdomains required for calcium-regulated exocytosis of dense-core vesicles. Mol Biol Cell 16:1108–1119.
  • Chen Z, Yoshihara E, Son A, Matsuo Y, Masutani H, Sugie K, 2010. Differential roles of Annexin A1 (ANXA1/lipocortin-1/lipomodulin) and thioredoxin binding protein-2 (TBP-2/VDUP1/TXNIP) in glucocorticoid signaling of HTLV-I-transformed T cells. Immunol Lett 131:11–18.
  • Choi Y, Zhang J, Murga C, Yu H, Koller E, Monia BP, 2002. PTEN, but not SHIP and SHIP2, suppresses the PI3K/Akt pathway and induces growth inhibition and apoptosis of myeloma cells. Oncogene 21:5289–5300.
  • Chung YW, Oh HY, Kim JY, Kim JH, Kim IY. 2004. Allergen-induced proteolytic cleavage of annexin-1 and activation of cytosolic phospholipase A2 in the lungs of a mouse model of asthma. Proteomics 4:3328–3334.
  • Clarke CJ, Forman S, Pritchett J, Ohanian V, Ohanian J. 2008. Phospholipase C-δ1 modulates sustained contraction of rat mesenteric small arteries in response to noradrenaline, but not endothelin-1. Am J Physiol Heart Circ Physiol 295:H826–H834.
  • Claus RA, Dorer MJ, Bunck AC, Deigner HP. 2009. Inhibition of sphingomyelin hydrolysis: Targeting the lipid mediator ceramide as a key regulator of cellular fate. Curr Med Chem 16:1978–2000.
  • Cmoch A, Strzelecka-Kiliszek A, Palczewska M, Groves P, Pikula S. 2011. Matrix vesicles isolated from mineralization-competent Saos-2 cells are selectively enriched with annexins and S100 proteins. Biochem Biophys Res Commun 412:683–687.
  • Cornely R, Rentero C, Enrich C, Grewal T, Gaus K. 2011. Annexin A6 is an organizer of membrane microdomains to regulate receptor localization and signalling. IUBMB Life 63:1009–1017.
  • Croxtall JD, Newman SP, Choudhury Q, Flower RJ. 1996a. The concerted regulation of cPLA2, COX2, and lipocortin 1 expression by IL-1 beta in A549 cells. Biochem Biophys Res Commun 220:491–495.
  • Croxtall JD, Choudhury Q, Newman S, Flower RJ. 1996b. Lipocortin 1 and the control of cPLA2 activity in A549 cells. Glucocorticoids block EGF stimulation of cPLA2 phosphorylation. Biochem Pharmacol 52:351–356.
  • Croxtall JD, Choudhury Q, Flower RJ. 1998. Inhibitory effect of peptides derived from the N-terminus of lipocortin 1 on arachidonic acid release and proliferation in the A549 cell line: Identification of E-Q-E-Y-V as a crucial component. Br J Pharmacol 123:975–983.
  • Croxtall JD, Gilroy DW, Solito E, Choudhury Q, Ward BJ, Buckingham JC, 2003. Attenuation of glucocorticoid functions in an Anx-A1-/- cell line. Biochem J 371:927–935.
  • Cubells L, Vilà de Muga S, Tebar F, Wood P, Evans R, Ingelmo-Torres M, 2007. Annexin A6-induced alterations in cholesterol transport and caveolin export from the Golgi complex. Traffic 8:1568–1589.
  • Cubells L, Vilà de Muga S, Tebar F, Bonventre JV, Balsinde J, Pol A, 2008. Annexin A6-induced inhibition of cytoplasmic phospholipase A2 is linked to caveolin-1 export from the Golgi. J Biol Chem 283:10174–10183.
  • Damazo AS, Moradi-Bidhendi N, Oliani SM, Flower RJ. 2007. Role of annexin 1 gene expression in mouse craniofacial bone development. Birth Defects Res A Clin Mol Teratol 79:524–532.
  • De Caterina R, Sicari R, Giannessi D, Paggiaro PL, Paoletti P, Lazzerini G, 1993. Macrophage-specific eicosanoid synthesis inhibition and lipocortin-1 induction by glucocorticoids. J Appl Physiol 75:2368–2375.
  • De Diego I, Schwartz F, Siegfried H, Dauterstedt P, Heeren J, Beisiegel U, 2002. Cholesterol modulates the membrane binding and intracellular distribution of annexin 6. J Biol Chem 277:32187–32194.
  • Dennis EA, Cao J, Hsu YH, Magrioti V, Kokotos G. 2011. Phospholipase A2 enzymes: Physical structure, biological function, disease implication, chemical inhibition, and therapeutic intervention. Chem Rev 111:6130–6185.
  • Domon MM, Matar G, Strzelecka-Kiliszek A, Bandorowicz-Pikula J, Pikula S, Besson F. 2010. Interaction of annexin A6 with cholesterol rich membranes is pH-dependent and mediated by the sterol OH. J Colloid Interface Sci 346:436–441.
  • Domon MM, Besson F, Bandorowicz-Pikula J, Pikula S. 2011. Annexin A6 is recruited into lipid rafts of Niemann-Pick type C disease fibroblasts in a Ca2+-dependent manner. Biochem Biophys Res Commun 405:192–196.
  • Domon MM, Nasir MN, Matar G, Pikula S, Besson F, Bandorowicz-Pikula J. 2012. Annexins as organizers of cholesterol- and spingomyelin-enriched membrane microdomains in Niemann-Pick type C disease. Cell Mol Life Sci 69:1773–1785.
  • Dong Q, Patel M, Scott KF, Graham GG, Russell PJ, Sved P. 2006. Oncogenic action of phospholipase A2 in prostate cancer. Cancer Lett 240:9–16.
  • Draeger A, Monastyrskaya K, Babiychuk EB. 2011. Plasma membrane repair and cellular damage control: The annexin survival kit. Biochem Pharmcol 81:703–712.
  • Enrich C, Rentero C, de Muga SV, Reverter M, Mulay V, Wood P, 2011. Annexin A6-linking Ca2+ signaling with cholesterol transport. Biochim Biophys Acta 1813:935–947.
  • Fang Z, Takizawa N, Wilson KA, Smith TC, Delprato A, Davidson MW, 2010. The membrane-associated protein, supervillin, accelerates F-actin-dependent rapid integrin recycling and cell motility. Traffic 11:782–799.
  • Fatimathas L, Moss SE. 2010. Annexins as disease modifiers. Histol Histopathol 25:527–532.
  • Faugaret D, Chouinard FC, Harbour D, El azreq MA, Bourgoin SG. 2011. An essential role for phospholipase D in the recruitment of vesicle amine transport protein-1 to membranes in human neutrophils. Biochem Pharmacol 81:144–156.
  • Flood EC, Hajjar KA. 2011. The annexin A2 system and vascular homeostasis. Vascul Pharmacol 54:59–67.
  • Flower RJ, Blackwell GJ. 1979. Anti-inflammatory steroids induce biosynthesis of a phospholipase A2 inhibitor which prevents prostaglandin generation. Nature 278:456–459.
  • Freedman SD, Blanco PG, Zaman MM, Shea JC, Ollero M, Hopper IK, 2004. Association of cystic fibrosis with abnormalities in fatty acid metabolism. N Engl J Med 350:560–569.
  • Fukami K, Inanobe S, Kanemaru K, Nakamura Y. 2010. Phospholipase C is a key enzyme regulating intracellular calcium and modulating the phosphoinositide balance. Prog Lipid Res 49:429–437.
  • Futter CE, White IJ. 2007. Annexins and endocytosis. Traffic 8:951–958.
  • Futter CE, Felder S, Schlessinger J, Ullrich A, Hopkins CR. 1993. Annexin I is phosphorylated in the multivesicular body during the processing of the epidermal growth factor receptor. J Cell Biol 120:77–83.
  • Galan C, Woodard GE, Dionisio N, Salido GM, Rosado JA. 2010. Lipid rafts modulate the activation but not the maintenance of store-operated Ca2+ entry. Biochim Biophys Acta 1803:1083–1093.
  • Ganapathi SB, Fox TE, Kester M, Elmslie KS. 2010. Ceramide modulates HERG potassium channel gating by translocation into lipid rafts. Am J Physiol Cell Physiol 299:C74–C86.
  • Gao X, Lowry PR, Zhou X, Depry C, Wei Z, Wong GW, 2011. PI3K/Akt signaling requires spatial compartmentalization in plasma membrane microdomains. Proc Natl Acad Sci USA 108:14509–14514.
  • Gentil-dit-Maurin A, Oun S, Almagro S, Bouillot S, Courçon M, Linnepe R, 2010. Unraveling the distinct distributions of VE- and N-cadherins in endothelial cells: A key role for p120-catenin. Exp Cell Res 316:2587–2599.
  • Gerke V, Moss SE. 2002. Annexins: From structure to function. Physiol Rev 82:331–371.
  • Gerke V, Creutz CE, Moss SE. 2005. Annexins: Linking Ca2+ signalling to membrane dynamics. Nat Rev Mol Cell Biol 6:449–461.
  • Goebeler V, Ruhe D, Gerke V, Rescher U. 2006. Annexin A8 displays unique phospholipid and F-actin binding properties. FEBS Lett 580:2430–2434.
  • Gokhale NA, Abraham A, Digman MA, Gratton E, Cho W. 2005. Phosphoinositide specificity of and mechanism of lipid domain formation by annexin A2-p11 heterotetramer. J Biol Chem 280:42831–42840.
  • Grewal T, Enrich C. 2009. Annexins – modulators of EGF receptor signalling and trafficking. Cell Signal 21:847–858.
  • Grewal T, Heeren J, Mewawala D, Schnitgerhans T, Wendt D, Salomon G, 2000. Annexin VI stimulates endocytosis and is involved in the trafficking of low density lipoprotein to the prelysosomal compartment. J Biol Chem 275:33806–33813.
  • Grewal T, Koese M, Rentero C, Enrich C. 2010. Annexin A6-regulator of the EGFR/Ras signalling pathway and cholesterol homeostasis. Int J Biochem Cell Biol 42:580–584.
  • Harrist AV, Ryzhova EV, Harvey T, González-Scarano F. 2009. Anx2 interacts with HIV-1 Gag at phosphatidylinositol (4,5) bisphosphate-containing lipid rafts and increases viral production in 293T cells. PLoS One 4:e5020.
  • Hayashi J, Liu P, Ferguson SE, Wen M, Sakata T, Teraoka H, 1993. Arachidonic acid metabolism in cells transfected with sense and antisense cDNA to annexin-1. Biochem Mol Biol Int 31:143–151.
  • Hayes MJ, Rescher U, Gerke V, Moss SE. 2004a. Annexin-actin interactions. Traffic 5:571–576.
  • Hayes MJ, Merrifield CJ, Shao D, Ayala-Sanmartin J, Schorey CD, Levine TP, 2004b. Annexin 2 binding to phosphatidylinositol 4,5-bisphosphate on endocytic vesicles is regulated by the stress response pathway. J Biol Chem 279:14157–14164.
  • Hayes MJ, Shao D, Bailly M, Moss SE. 2006. Regulation of actin dynamics by annexin 2. EMBO J 25:1816–1826.
  • Hayes MJ, Longbottom RE, Evans MA, Moss SE. 2007. Annexinopathies. Subcell Biochem 45:1–28.
  • Hayes MJ, Shao DM, Grieve A, Levine T, Bailly M, Moss SE. 2009. Annexin A2 at the interface between F-actin and membranes enriched in phosphatidylinositol 4,5,-bisphosphate. Biochim Biophys Acta 1793:1086–1095.
  • He KL, Sui G, Xiong H, Broekman MJ, Huang B, Marcus AJ, 2011. Feedback regulation of endothelial cell surface plasmin generation by PKC-dependent phosphorylation of annexin A2. J Biol Chem 286:15428–15439.
  • Herbert SP, Odell AF, Ponnambalam S, Walker JH. 2007. The confluence-dependent interaction of cytosolic phospholipase A2-alpha with annexin A1 regulates endothelial cell prostaglandin E2 generation. J Biol Chem 282:34468–34478.
  • Hirabayashi T, Murayama T, Shimizu T. 2004. Regulatory mechanism and physiological role of cytosolic phospholipase A2. Biol Pharm Bull 27:1168–1173.
  • Hofmann A, Osman A, Leow CY, Driguez P, McManus DP, Jones MK. 2010. Parasite annexins--new molecules with potential for drug and vaccine development. Bioessays 32:967–976.
  • Hummel I Klappe K, Ercan C, Kok JW. 2011. Multidrug resistance-related protein 1 (MRP1) function and localization depend on cortical actin. Mol Pharmacol 79:229–240.
  • Illien F, Finet S, Lambert O, Ayala-Sanmartin J. 2010. Different molecular arrangements of the tetrameric annexin 2 modulate the size and dynamics of membrane aggregation. Biochim Biophys Acta 1798:1790–1796.
  • Jenkins RW, Canals D, Hannun YA. 2009. Roles and regulation of secretory and lysosomal acid sphingomyelinase. Cell Signal 21:836–846.
  • Jin S, Zhou F. 2009. Lipid raft redox signaling platforms in vascular dysfunction: Features and mechanisms. Curr Atheroscler Rep 11:220–226.
  • Johansen C, Kragballe K, Henningsen J, Westergaard M, Kristiansen K, Iversen L. 2003. 1α,25-dihydroxyvitamin D3 stimulates activator protein 1 DNA-binding activity by a phosphatidylinositol 3-kinase/Ras/MEK/extracellular signal regulated kinase 1/2 and c-Jun N-terminal kinase 1-dependent increase in c-Fos, Fra1, and c-Jun expression in human keratinocytes. J Invest Dermatol 120:561–570.
  • Kaetzel MA, Dedman JR. 1995. Annexins: Novel Ca2+-dependent regulators of membrane function. News Physiolog Sci 10:171–176.
  • Kaetzel MA, Dedman JR. 2004. Annexin VI regulation of cardiac function. Biochem Biophys Res Commun 322:1171–1177.
  • Kanaho Y, Suzuki T. 2002. Phosphoinositide kinases as enzymes that produce versatile signaling lipids, phosphoinositides. J Biochem 131:503–509.
  • Khau T, Langenbach SY, Schuliga M, Harris T, Johnstone CN, Anderson RL, 2011. Annexin-1 signals mitogen-stimulated breast tumor cell proliferation by activation of the formyl peptide receptors (FPRs) 1 and 2. FASEB J 25:483–496.
  • Kim KM, Kim DK, Park YM, Kim CK, Na DS. 1994. Annexin-I inhibits phospholipase A2 by specific interaction, not by substrate depletion. FEBS Lett 343:251–255.
  • Kim S, Ko J, Kim JH, Choi EC, Na DS. 2001a. Differential effects of annexins I, II, III, and V on cytosolic phospholipase A2 activity: Specific interaction model. FEBS Lett 489:243–248.
  • Kim SW, Rhee HJ, Ko J, Kim YJ, Kim HG, Yang JM, 2001b. Inhibition of cytosolic phospholipase A2 by annexin I. Specific interaction model and mapping of the interaction site. J Biol Chem 276:15712–15719.
  • Kiyanagi T, Iwabuchi K, Shimada K, Hirose K, Miyazaki T, Sumiyoshi K, 2011. Involvement of cholesterol-enriched microdomains in class A scavenger receptor-mediated responses in human macrophages. Atherosclerosis 215:60–69.
  • Konopka-Postupolska D, Clark G, Hofmann A. 2011. Structure, function and membrane interactions of plant annexins: An update. Plant Sci 181:230–241.
  • Lafont F, Lecat S, Verkade P, Simons K. 1998. Annexin XIIIb associates with lipid microdomains to function in apical delivery. J Cell Biol 142:1413–1427.
  • Lambert O, Cavusoglu N, Gallay J, Vincent M, Rigaud JL, Henry JP, 2004. Novel organization and properties of annexin 2-membrane complexes. J Biol Chem 279:10872–10882.
  • Laohavisit A, Davies JM. 2011. Annexins. New Phytol 189:40–53.
  • Lasley RD. 2011. Adenosine receptors and membrane microdomains. Biochim Biophys Acta 1808:1284–1289.
  • Lecat S, Verkade P, Thiele C, Fiedler K, Simons K, Lafont F. 2000. Different properties of two isoforms of annexin XIII in MDCK cells. J Cell Sci 113:2607–2618.
  • Lemmon MA. 2008. Membrane recognition by phospholipid-binding domains. Nat Rev Mol Cell Biol 9:99–111.
  • Leslie CC, Gangelhoff TA, Gelb MH. 2010. Localization and function of cytosolic phospholipase A2alpha at the Golgi. Biochimie 92:620–626.
  • Levental I, Grzybek M, Simons K. 2010. Greasing their way: Lipid modifications determine protein association with membrane rafts. Biochemistry 49:6305–6316.
  • Lim LH, Pervaiz S. 2007. Annexin 1: The new face of an old molecule. FASEB J 21:968–975.
  • Lin S, Wang XM, Nadeau PE, Mergia A. 2010. HIV infection upregulates caveolin 1 expression to restrict virus production. J Virol 84:9487–9496.
  • Liu J, Zhu X, Myo S, Lambertino AT, Xu C, Boetticher E, 2005. Glucocorticoid-induced surface expression of annexin 1 blocks beta2-integrin adhesion of human eosinophils to intercellular adhesion molecule 1 surrogate protein. J Allergy Clin Immunol 115:493–500.
  • Mahimainathan L, Choudhury GG. 2004. Inactivation of platelet-derived growth factor receptor by the tumor suppressor PTEN provides a novel mechanism of action of the phosphatase. J Biol Chem 279:15258–15268.
  • Martin-Belmonte F, Mostov K. 2007. Phosphoinositides control epithelial development. Cell Cycle 6:1957–1961.
  • Martin-Belmonte F, Gassama A, Datta A, Yu W, Rescher U, Gerke V, 2007. PTEN-mediated apical segregation of phosphoinositides controls epithelial morphogenesis through Cdc42. Cell 128:383–397.
  • Matsunaga T, Inaba T, Matsui H, Okuya M, Miyajima A, Inukai T, 2004. Regulation of annexin II by cytokine-initiated signaling pathways and E2A-HLF oncoprotein. Blood 103:3185–3191.
  • Mayran N, Parton RG, Gruenberg J. 2003. Annexin II regulates multivesicular endosome biogenesis in the degradation pathway of animal cells. EMBO J 22:3242–3253.
  • McDermott M, Wakelam MJ, Morris AJ. 2004. Phospholipase D. Biochem Cell Biol 82:225–253.
  • Meliton AY, Munoz NM, Zhu X, Leff AR. 2008. Attenuated translocation of group IVa phospholipase A2 and up-regulated annexin-1 synthesis by glucocorticoid blocks beta 2-integrin adhesion in neutrophils. J Leukoc Biol 83:344–351.
  • Monastyrskaya K, Babiychuk EB, Hostettler A, Rescher U, Draeger A. 2007. Annexins as intracellular calcium sensors. Cell Calcium 41:207–219.
  • Monastyrskaya K, Babiychuk EB, Hostettler A, Wood P, Grewal T, Draeger A. 2009a. Plasma membrane-associated annexin A6 reduces Ca2+ entry by stabilizing the cortical actin cytoskeleton. J Biol Chem 284:17227–17242.
  • Monastyrskaya K, Babiychuk EB, Draeger A. 2009b. The annexins: Spatial and temporal coordination of signaling events during cellular stress. Cell Mol Life Sci 66:2623–2642.
  • Morel E, Parton R, Gruenberg J. 2009. Annexin A2-dependent polymerization of actin mediates endosome biogenesis. Dev Cell 16:445–457.
  • Morgan RO, Martin-Almedina S, Iglesias JM, Gonzalez-Florez MI, Fernandez MP. 2004. Evolutionary perspective on annexin calcium-binding domains. Biochim Biophys Acta 1742:133–140.
  • Morgan RO, Martin-Almedina S, Garcia M, Jhoncon-Kooyip J, Fernandez MP. 2006. Deciphering function and mechanism of calcium-binding proteins from their evolutionary imprints. Biochim Biophys Acta 1763:1238–1249.
  • Muimo R. 2009. Regulation of CFTR function by annexin A2-S100A10 complex in health and disease. Gen Physiol Biophys 28:F14–F19.
  • Mussunoor S, Murray GI. 2008. The role of annexins in tumour development and progression. J Pathol 216:131–140.
  • Nadiri A, Polyak MJ, Jundi M, Alturaihi H, Reyes-Moreno C, Hassan GS, 2011. CD40 translocation to lipid rafts: Signaling requirements and downstream biological events. Eur J Immunol 41:2358–2367.
  • Nishizuka Y. 1995. Protein kinase C and lipid signaling for sustained cellular responses. FASEB J 9:484–496.
  • Parente L, Solito E. 2004. Annexin 1: More than an anti-phospholipase protein. Inflamm Res 53:125–132.
  • Paris L, Cecchetti S, Spadaro F, Abalsamo L, Lugini L, Pisanu ME, 2010. Inhibition of phosphatidylcholine-specific phospholipase C downregulates HER2 overexpression on plasma membrane of breast cancer cells. Breast Cancer Res 12:R27.
  • Potez S, Luginbühl M, Monastyrskaya K, Hostettler A, Draeger A, Babiychuk EB. 2011. Tailored protection against plasmalemmal injury by annexins with different Ca2+ sensitivities. J Biol Chem 286:17982–17991.
  • Rescher U, Ruhe D, Ludwig C, Zobiack N, Gerke V. 2004. Annexin 2 is a phosphatidylinositol (4,5)-bisphosphate binding protein recruited to actin assembly sites at cellular membranes. J Cell Sci 117:3473–3480.
  • Reverter M, Rentero C, de Muga SV, Alvarez-Guaita A, Mulay V, Cairns R, 2011. Cholesterol transport from late endosomes to the Golgi regulates t-SNARE trafficking, assembly, and function. Mol Biol Cell 22:4108–4123.
  • Rezvanpour A, Santamaria-Kisiel L, Shaw GS. 2011. The S100A10-annexin A2 complex provides a novel asymmetric platform for membrane repair. J Biol Chem 286:40174–40183.
  • Riquelme G, Llanos P, Tischner E, Neil J, Campos B. 2004. Annexin 6 modulates the maxi-chloride channel of the apical membrane of syncytiotrophoblast isolated from human placenta. J Biol Chem 279:50601–50608.
  • Rivera R, Chun J. 2008. Biological effects of lysophospholipids. Rev Physiol Biochem Pharmacol 160:25–46.
  • Rosenbaum S, Kreft S, Etich J, Frie C, Stermann J, Grskovic I, 2011. Identification of novel binding partners (annexins) for the cell death signal phosphatidylserine and definition of their recognition motif. J Biol Chem 286:5708–5716.
  • Russo-Marie F. 1999. Annexin V and phospholipid metabolism. Clin Chem Lab Med 37:287–291.
  • Saavedra G, Zhang W, Peterson B, Cummings BS. 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.
  • Sakaguchi M, Murata H, Sonegawa H, Sakaguchi Y, Futami J, Kitazoe M, 2007. Truncation of annexin A1 is a regulatory lever for linking epidermal growth factor signaling with cytosolic phospholipase A2 in normal and malignant squamous epithelial cells. J Biol Chem 282:35679–35686.
  • Schiffmann R. 2010. Therapeutic approaches for neuronopathic lysosomal storage disorders. J Inherit Metab Dis 33:373–379.
  • Schuchman EH. 2010. Acid sphingomyelinase, cell membranes and human disease: Lessons from Niemann-Pick disease. FEBS Lett 584:1895–1900.
  • Schug ZT, Frezza C, Galbraith LC, Gottlieb E. 2012. The music of lipids: How lipid composition orchestrates cellular behaviour. Acta Oncol 51:301–310.
  • Schutters K, Reutelingsperger C. 2010. Phosphatidylserine targeting for diagnosis and treatment of human diseases. Apoptosis 15:1072–1082.
  • Seaton BA, Dedman JR. 1998. Annexins. Biometals 11:399–404.
  • Selvy PE, Lavieri RR, Lindsley CW, Brown HA. 2011. Phospholipase D: Enzymology, functionality, and chemical modulation. Chem Rev 111:6064–6119.
  • Skrahina T, Piljić A, Schultz C. 2008. Heterogeneity and timing of translocation and membrane-mediated assembly of different annexins. Exp Cell Res 314:1039–1047.
  • Sohma H, Creutz CE, Gasa S, Ohkawa H, Akino T, Kuroki Y. 2001. Differential lipid specificities of the repeated domains of annexin IV. Biochim Biophys Acta 1546:205–215.
  • Solito E, de Coupade C, Canaider S, Goulding NJ, Perretti M. 2001. Transfection of annexin 1 in monocytic cells produces a high degree of spontaneous and stimulated apoptosis associated with caspase-3 activation. Br J Pharmacol 133:217–228.
  • Sones WR, Davis AJ, Leblanc N, Greenwood IA. 2010. Cholesterol depletion alters amplitude and pharmacology of vascular calcium-activated chloride channels. Cardiovasc Res 87:476–484.
  • Staubach S, Hanisch FG. 2011. Lipid rafts: Signaling and sorting platforms of cells and their roles in cancer. Expert Rev Proteomics 8:263–277.
  • Surma MA, Klose C, Simons K. 2011. Lipid-dependent protein sorting at the trans-Golgi network. Biochim Biophys Acta; DOI: 10.1016/j.bbalip.2011.12.008.
  • Sztolsztener ME, Strzelecka-Kiliszek A, Pikula S, Tylki-Szymanska A, Bandorowicz-Pikula J. 2010. Cholesterol as a factor regulating intracellular localization of annexin A6 in Niemann-Pick type C human skin fibroblasts. Arch Biochem Biophys 493:221–233.
  • te Vruchte D, Lloyd-Evans E, Veldman RJ, Neville DC, Dwek RA, Platt FM, 2004. Accumulation of glycosphingolipids in Niemann-Pick C disease disrupts endosomal transport. J Biol Chem 279:26167–26175.
  • Tamasawa N, Takayasu S, Murakami H, Yamashita M, Matsuki K, Tanabe J, 2012. Reduced cellular cholesterol efflux and low plasma high-density lipoprotein cholesterol in a patient with type B Niemann-Pick disease because of a novel SMPD-1 mutation. J Clin Lipidol 6:74–80.
  • Tian W, Wijewickrama GT, Kim JH, Das S, Tun MP, Gokhale N, 2008. Mechanism of regulation of group IVA phospholipase A2 activity by Ser727 phosphorylation. J Biol Chem 283:3960–3971.
  • Torosyan Y, Dobi A, Naga S, Mezhevaya K, Glasman M, Norris C, 2006. Distinct effects of annexin A7 and p53 on arachidonate lipoxygenation in prostate cancer cells involve 5-lipoxygenase transcription. Cancer Res 66:9609–9616.
  • Torosyan Y, Simakova O, Naga S, Mezhevaya K, Leighton X, Diaz J, 2009. Annexin-A7 protects normal prostate cells and induces distinct patterns of RB-associated cytotoxicity in androgen-sensitive and -resistant prostate cancer cells. Int J Cancer 125:2528–2539.
  • Umbrecht-Jenck E, Demais V, Calco V, Bailly Y, Bader MF, Chasserot-Golaz S. 2010. S100A10-mediated translocation of annexin-A2 to SNARE proteins in adrenergic chromaffin cells undergoing exocytosis. Traffic 11:958–971.
  • Valapala M, Vishwanatha JK. 2011. Lipid raft endocytosis and exosomal transport facilitate extracellular trafficking of annexin A2. J Biol Chem 286:30911–30925.
  • Vats K, Knutson K, Hinderliter A, Sheets ED. 2010. Peripheral protein organization and its influence on lipid diffusion in biomimetic membranes. ACS Chem Biol 5:393–403.
  • Vilá de Muga S, Timpson P, Cubells L, Evans R, Hayes TE, Rentero C, 2009. Annexin A6 inhibits Ras signalling in breast cancer cells. Oncogene 28:363–377.
  • White IJ, Bailey LM, Aghakhani MR, Moss SE, Futter CE. 2006. EGF stimulates annexin 1-dependent inward vesiculation in a multivesicular endosome subpopulation. EMBO J 25:1–12.
  • Zhang C, Li PL. 2010. Membrane raft redox signalosomes in endothelial cells. Free Radic Res 44:831–842.
  • Zhang F, Sha J, Wood TG, Galindo CL, Garner HR, Burkart MF, 2008. Alteration in the activation state of new inflammation-associated targets by phospholipase A2-activating protein (PLAA. Cell Signal 20:844–861.
  • Zobiack N, Rescher U, Laarmann S, Michgehl S, Schmidt MA, Gerke V. 2002. Cell-surface attachment of pedestal-forming enteropathogenic E. coli induces a clustering of raft components and a recruitment of annexin 2. J Cell Sci 115:91–98.
  • Zobiack N, Rescher U, Ludwig C, Zeuschner D, Gerke V. 2003. The annexin 2/S100A10 complex controls the distribution of transferrin receptor-containing recycling endosomes. Mol Biol Cell 14:4896–4908.
  • Zschörnig O, Opitz F, Müller M. 2007. Annexin A4 binding to anionic phospholipid vesicles modulated by pH and calcium. Eur Biophys J 36:415–424.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.