Biochem. J. (2012) 441, 39–59 (Printed in Great Britain) 39 doi:10.1042/BJ20111226 REVIEW ARTICLE Insights into the PX (phox-homology) domain and SNX (sorting nexin) protein families: structures, functions and roles in disease Rohan D. TEASDALE and Brett M. COLLINS1 Institute for Molecular Bioscience, University of Queensland, St. Lucia, Queensland 4072, Australia The mammalian genome encodes 49 proteins that possess a PX (phox-homology) domain, responsible for membrane attachment to organelles of the secretory and endocytic system via binding of phosphoinositide lipids. The PX domain proteins, most of which are classified as SNXs (sorting nexins), constitute an extremely diverse family of molecules that play varied roles in membrane trafficking, cell signalling, membrane remodelling and organelle motility. In the present review, we present an overview of the family, incorporating recent functional and structural insights, and propose an updated classification of the proteins into distinct subfamilies on the basis of these insights. Almost all PX domain proteins bind PtdIns3P and are recruited to early endosomal membranes. Although other specificities and localizations have been reported for a select few family members, the molecular basis for binding to other lipids is still not clear. The PX domain is also emerging as an important protein–protein interaction domain, binding endocytic and exocytic machinery, transmembrane proteins and many other molecules. A comprehensive survey of the molecular interactions governed by PX proteins highlights the functional diversity of the family as trafficking cargo adaptors and membrane-associated scaffolds regulating cell signalling. Finally, we examine the mounting evidence linking PX proteins to different disorders, in particular focusing on their emerging importance in both pathogen invasion and amyloid production in Alzheimer’s disease. INTRODUCTION binds to PtdInsP lipids (phosphoinositides) on the cytoplasmic leaflets of different organelles, similar in function to other lipid-binding domains such as the PH (pleckstrin homology), FYVE (Fab1p/YOTB/Vac1p/EEA1) and ENTH (epsin N-terminal homology) domains. This association defines the cellular localization of the different PX proteins and hence is critical to their function. The present review summarizes recent progress in defining the structural and functional properties of the PX domain-containing proteins, and presents an update of some previously ambiguous structural classifications of the proteins. We provide a survey of the diverse biomolecular interactions that are regulated by these molecules, demonstrating their central functions as both cargo adaptors in cell trafficking and scaffolds regulating signal transduction, and discuss some of the recent work on previously poorly characterized PX domain-containing protein families. Finally, we highlight the emerging roles of PX proteins in disease. Eukaryotic cells contain numerous membrane-bound organelles involved in secretion and endocytosis. Transmembrane proteins, lipids and other critical cargo molecules must be selectively sorted and transported between these compartments via membrane trafficking processes, involving either vesicular or tubulovesicular carriers or compartmental maturation and fusion events. Furthermore, it is now apparent that endocytic compartments are also critical platforms for the regulation of cell signalling, well beyond the traditional function of controlling receptor degradation [1,2]. The mammalian Phox-homology (PX) domaincontaining molecules constitute a large family of at least 49 proteins. They are predominantly found in the endosomal system, but also localize to other cellular compartments and regulate diverse trafficking and signalling processes in the cell. The PX domain is a membrane recruitment module that Key words: Alzheimer’s disease, endosome, phosphoinositide, phox-homology domain (PX domain), retromer, sorting nexin (SNX). Abbreviations used: AD, Alzheimer’s disease; ADAM, a disintegrin and metalloproteinase; AP2, adaptor protein complex 2; ApoE, apolipoprotein E; APP, amyloid precursor protein; BACE, β-secretase; BAR, Bin/amphiphysin/Rvs; CASP, cytohesin-associated scaffolding protein; CGD, chronic granulomatous disease; CI-MPR, cation-independent mannose-6-phosphate receptor; DGK, diacylglycerolkinase; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; ERK, extracellular-signal-regulated kinase; FERM, band4.1/ezrin/radixin/moesin; FHA, forkhead association; FISH, five SH3 domains; FYVE, Fab1p/YOTB/Vac1p/EEA1; GAP, GTPase-activating protein; GC-GAP, GAB/cdc42 GAP; GPCR, G-protein-coupled receptor; Hrs, hepatocyte growth factor-regulated tyrosine kinase substrate; HS1BP3, HS1-binding protein 3; 5-HT4 R, 5-hydroxytryptamine type 4 receptor; IRAS, imidazoline receptor antisera selected; IRS, insulin receptor substrate; KIF, kinesin-family protein; Krit1, Krev-interaction trapped 1; LDLR, low-density lipoprotein receptor; LRP1, LDLR-related protein 1; LRR, leucine-rich repeat; MIT domain, microtubule-interacting and trafficking molecule domain; MMEP, microcephaly, micropthalmia, ectrodactyly and prognathism; MVB, multivesicular body; NCF, neutrophil cytosolic factor; NMDA, N -methyl-D-aspartate; NOXO1, NADPH oxidase organizer 1; NR, NMDA receptor; PA, phosphatidic acid; PDZ, postsynaptic density 95/discs large/zonula occludens; PDZbm, PDZ-binding motif; PH, pleckstrin homology; PI3K, phosphoinositide 3-kinase; PLCγ1, phospholipase Cγ1; PLD, phospholipase D; PS1, presenilin 1; PSGL, P-selectin glycoprotein ligand; PTB, phosphotyrosine binding; PX, phox-homology; PXA domain, PX-associated domain A; PXC, PX-associated domain C; RA, Ras-association; RGS, regulator of G-protein signalling; RPS6K, ribosomal protein S6 kinase; SCV, Salmonella -containing vacuole; SGK, serum- and glucocorticoid-induced protein kinase; SH, Src homology; SH3PXD2A, SH3 and PX domain protein 2A; siRNA, small interfering RNA; SLIC-1, selectin ligand interactor cytoplasmic-1; SNARE, soluble N -ethylmaleimide-sensitive factor-attachment protein receptor; snazarus, sorting nexin lazarus; SNX, sorting nexin; S/T kinase, serine/threonine kinase; STx, Shiga toxin; TC-GAP, TC10/cdc42 GAP; TGF-β, transforming growth factor β; TGN, trans -Golgi network; TPR, tetratricopeptide repeat; VPS, vacuolar protein sorting-associated protein; VSV, vesicular stomatitis virus; WASP, Wiskott–Aldrich syndrome protein; WASH, WASP and SCAR (suppressor of cAMP receptor) homologue; N-WASP, neuronal WASP. 1 To whom correspondence should be addressed (email [email protected]). c The Authors Journal compilation c 2012 Biochemical Society 40 R. D. Teasdale and B. M. Collins THE PX DOMAIN Structure of the PX domain and binding to the endosomal lipid marker PtdIns3P The PX domain is a globular fold approximately 110 residues in length, composed of three β-strands followed by three α-helices (Figure 1). Specific features of this protein fold are a long proline-rich loop between helices α1 and α2, and an electropositive basic pocket that is involved in direct binding of negatively charged phosphate groups of phosphoinositides [3]. In addition several studies have highlighted the role of adjacent hydrophobic residues in membrane penetration in at least some PX domains, and an additional positively charged surface patch, which is able to associate weakly with other anionic lipids, such as phosphatidylserine [4–8]. A survey of the known structures of mammalian and yeast PX proteins, their localization and lipid binding in vitro reveals a number of insights into the PX domain phosphoinositide specificity (Figure 1, and Supplementary Movie S1, Table S1 and Figure S1 at http://www.BiochemJ.org/bj/441/bj4410039add.htm). A global assessment of Saccharomyces cerevisiae PX domain molecules demonstrated an almost absolute PtdIns3P specificity [9], and a review of the literature presents an overwhelming consensus that mammalian PX domains also show a preference for PtdIns3P (see Supplementary Table S1). PtdIns3P is primarily found on early endosomal limiting membranes and therefore most commonly directs PX protein recruitment to PtdIns3P-enriched endosomal organelles. It should be noted, however, that distinct pools of PtdIns3P can be found in other compartments, such as the plasma membrane, being often generated during specific signalling processes [10]. Currently there are 27 structures of mammalian PX domains available in the PDB (from 14 different proteins), but only two have been solved in complex with PtdIns3P: p40phox (PDB code 1H6H) and SNX9 (sorting nexin 9; PDB code 2RAK). There are five known structures of yeast PX domains, with one structure of Grd19p/Snx3p in complex with PtdIns3P (PDB code 1OCU). In addition, the binding of PtdIns3P to the SNX17 and SNX22 PX domains has been analysed using NMR and restrained computational docking approaches [11,12]. These studies reveal the key determinants for PtdIns3P binding (Figure 1 and Supplementary Table S1) [3]. Each moiety of the PtdIns3P headgroup interacts with a specific and strictly conserved side chain of the PX domain. The defining 3-phosphate forms electrostatic bonds with a conserved arginine side chain. The inositol group forms a key stacking interaction with a conserved tyrosine residue (or in some cases phenylalanine) immediately one residue downstream of the conserved arginine. The 4- and 5-hydroxy groups of the inositol form hydrogen bonds with a conserved arginine side chain, which helps orient the PtdIns3P in the binding pocket and also sterically precludes binding of 4- or 5-phosphorylated lipids. In most cases, the polyproline loop also creates a steric boundary against incoming 5-phosphate groups, although this loop is inherently flexible and may be able to alter its conformation. Finally, the 1-phosphate is co-ordinated by a conserved lysine side chain, which is immediately downstream of the polyproline loop. The necessary consensus sequence in PX domains for PtdIns3P binding is thus R[Y/F]x23–30 Kx13–23 R. Apart from lipid binding by the PX domain, a very important additional factor is the contribution of associated domains and other molecular interactions that drive and enhance membrane attachment through ‘coincidence detection’ [13]. Thus membrane attachment specificity may be controlled not only by the PX domain, but also by combination with domains that associate independently with membrane lipids [e.g. BAR (Bin/amphiphysin/Rvs), FERM (band4.1/ezrin/radixin/moesin) c The Authors Journal compilation c 2012 Biochemical Society Figure 1 Structure of the PX domain (A) Ribbon and surface structures of a representative PX domain from p40phox in complex with PtdIns3P (PDB code 1H6H). Key determinants of the interaction are the arginine side chain electrostatic association with the 3-phosphate (ArgP3 ), stacking of the inositol ring with the tyrosine (or phenylalanine) side chain immediately downstream from the conserved arginine residue (Tyrinositol ), contact of a lysine side chain with the 1-phosphate (LysP1 ), and hydrogen bonds of the 4- and 5-hydroxy groups to a second arginine side chain (Arg4,5-hydroxyl ). The polyproline loop also forms a steric block of potential 5-phosphate groups in the binding pocket. The PtdIns3P headgroup is located within a deep positively charged pocket. The consensus PX domain-binding sequence is shown below. See also Supplementary Movie S1 at http://www.BiochemJ.org/bj/441/bj4410039add.htm. (B) Structure of the human SNX5 PX domain [22] overlaid with the human PX domain of SNX9 within the SNX9 PX-BAR assembly [18]. The SNX9 protein is coloured in grey, whereas SNX5 is coloured dark blue to light blue (Nto C-terminal). Bound PtdIns3P molecules are shown as spheres, with carbon atoms coloured green. A schematic membrane is indicated with a curvature that follows the curvature of the SNX9 PX-BAR assembly. The SNX5 helical insertion, indicated by the broken red circle, will clearly form steric interactions with the membrane environment in this orientation. SNX5 is only shown aligned with one subunit of the PX-BAR dimer for clarity. and PH domains] or bind to other membrane-associated proteins such as Rabs or cargo molecules. Do PX domains bind other phosphoinositides? PtdIns3P is the most common phosphoinositide bound by PX domains; however, many other phosphoinositide interactions have The PX protein family been reported, suggesting a diverse role in membrane trafficking or signalling at different cellular compartments (Supplementary Table S1). Typical reported affinities (K d values) in vitro are of the order of 0.1–10 μM, but vary wildly from nanomolar to millimolar values depending on the PX domain studied and sometimes even for the same PX domain. Assessing the phosphoinositide binding of PX domain proteins is complicated by the range of different methods used and the relative reliability of these methods [14,15]. Despite the wide variety of other phosphoinositide specificities reported, there is still no conclusive structural indication of how these interactions occur. Indeed, a naı̈ve analysis of the available structural data predicts that other phosphoinositides apart from PtdIns3P will not be able to associate with PX domains, unless their binding mode is significantly different from that demonstrated for PtdIns3P (Supplementary Table S1 and Figure S1). In most cases there are steric considerations that preclude the association of 4- and 5-phosphate groups, and where this is not the case there are no obvious determinants for specific coordination of these phosphates as might be expected. This lack of a clear mechanistic basis for binding of alternative phosphoinositides to PtdIns3P suggests that cases where other specificities are reported could merit further scrutiny, and we have selected two cases to illustrate this point. SNX9 has been reported to have a functional preference for PtdIns(4,5)P2 in liposomebinding assays and in vitro assays for stimulation of dynamin and N-WASP [neuronal WASP (Wiskott–Aldrich syndrome protein)] activities, but it can associate with all other phosphoinositide lipids, including PtdIns3P [16–21]. This liposome association, however, requires co-operation of the SNX9 C-terminal BAR domain, thus clouding the PX domain specificity [18,20]. The structure of SNX9 was determined by crystallography in the presence of PtdIns3P (PDB code 2RAK), but notably, despite soaking with other phosphoinositide lipids, none were observed to bind the PX domain [18]. Furthermore, additional biochemical data shows that mutation of the PX domain abrogates binding of the PX-BAR unit of SNX9 to PtdIns3P- but not PtdIns(4,5)P2 -containing liposomes [18]. This suggests that the SNX9 PX domain is specific for PtdIns3P, whereas the binding of PtdIns(4,5)P2 is mediated primarily by the SNX9 BAR domain. SNX5, and its related homologues SNX6 and SNX32 have an unusual PX domain with a large conserved insertion following the polyproline loop [3]. The structure of the SNX5 PX domain shows that this insertion forms an extended helix–turn–helix structure [22] (PDB code 3HPC). The authors used NMR to map the binding of different phosphoinositides and suggested that PtdIns(4,5)P2 binds to a site overlapping the approximate location where PtdIns3P binds other PX domains. However, their data shows that the affinity is extremely weak (K d > 400 μM). It also contradicts previous studies from their own group [23] and perhaps, more importantly, does not explain cell localization data showing that SNX5 is only membrane-associated in the presence of SNX1 [24]. As the structure of SNX5 reveals that it lacks both conserved residues required for phosphoinositide binding and an identifiable binding pocket for PtdIns(4,5)P2 , the specificity of SNX5 certainly merits closer scrutiny. In conclusion, although mammalian PX domains have been reported to have specificities for phosphoinositides other than PtdIns3P, a detailed structural explanation is required to understand precisely how they are associated. The PX domain as a protein-interaction module Studies of the PX domain have almost exclusively focused on its ability to bind phosphoinositides, but there is accumulating 41 evidence that the PX domain can also act as a protein-scaffolding device. We have surveyed all reported protein–protein interactions of PX domain-containing molecules, and a number of these occur directly via the PX domain itself (Supplementary Table S2 at http://www.BiochemJ.org/bj/441/bj4410039add.htm). Compared with the detailed molecular understanding we now have of PtdIns3P binding, how PX domains control protein–protein interactions remains very poorly understood. One common feature of PX domains is the presence of a polyproline loop between helices α1 and α2, which may potentially be involved in interactions with SH3 (Src homology 3) domains. Indeed both intramolecular and intermolecular SH3 domain binding has been observed. An example of the former is the binding of C-terminal SH3 domains of p47phox to the p47phox PX domain [25], an interaction that regulates its activation [26]. An example of an intermolecular association is the binding of the PLCγ 1 (phospholipase Cγ 1) SH3 domain to the polyproline loop of the PLD1 (phospholipase D1) and PLD2 PX domains, important in EGF (epidermal growth factor) signalling [27]. Given the importance of the dynamic remodelling of the polyproline loop on PtdIns3P binding, this also suggests a possible role for protein– protein interactions in regulating membrane association [28]. A number of other direct protein–protein interactions involving PX domains have also been reported. The PX domains of PLD1 and PLD2 have been found to bind to the GTPase domain of dynamin and are able to act as GAPs (GTPase-activating proteins) affecting dynamin-mediated endocytosis [29]. PLD1 and PLD2 PX domains are also able to associate with the exocytic SM (Sec/Munc) protein Munc18-1, and this association potently inhibits the phospholipase activity of the proteins [30]. The PX domains of a number of PX proteins, including SNX1, SNX2, SNX4 and SNX6, can associate with the cytoplasmic tails of members of the TGF-β (transforming growth factor β) receptor family [31]. The functional significance of these interactions have not been well characterized, but overexpression of SNX6 was found to downregulate TGF-β signalling. The SNX2 PX domain was also found to bind to the DEAD-box helicase Abstrakt [32], although again the functional importance of this interaction is not clear. The vesicle coat protein clathrin can bind to several PX domains through a novel inverted clathrin box sequence, including SNX1, SNX2, SNX3 and SNX4 [33]. The SNX6 PX domain can drive association with the oncogene Pim-1, resulting in SNX6 phosphorylation and nuclear translocation [34], although once more the role of this nuclear localization remains a mystery. Intriguingly, the SNX9 PX domain is able to bind to and stimulate phosphoinositide kinases, implying that a positive feedback mechanism between phosphoinositide generation and effector binding may be involved in SNX9-mediated endocytosis and signalling [16,19]. A final example of PX domain protein interactions involves the binding of SNX20 to the cytoplasmic tail of the transmembrane cell-surface protein PSGL-1 (Pselectin glycoprotein ligand 1), discussed below in more detail [35]. Taken together, the evidence is substantial that PX domains act not only as lipid recognition modules, but play a key role in protein–protein interactions, and it will be of major interest to determine the molecular details of how these interactions occur. There is an obvious parallel here between the PX domain and the PTB (phosphotyrosine binding)/PH domains [36]. Like the PX domain, these are small modules of approximately 120 amino acids that bind to different phosphoinositides and are found in a number of different endocytic adaptors and signalling molecules. In addition, they are known to associate directly with other proteins, typically via short peptide motifs. A similar understanding of the protein-binding partners of the PX c The Authors Journal compilation c 2012 Biochemical Society 42 Table 1 R. D. Teasdale and B. M. Collins Subfamilies of mammalian PX proteins on the basis of domain architecture Subfamily Member(s) PX-BAR SNX1, SNX2, SNX4, SNX5, SNX6, SNX7, SNX8, SNX30, SNX32 SNX9 (SH3PX1), SNX18, SNX33 SH3-PX-BAR PX-SH3 PX-SH3-GAP PX-FERM PXA-RGS-PX-PXC PX-serine/threonine kinase PI3K-PX PX-PH-PLD PX-PXB PX-only Kinesin-PX PX-MIT PX-LRR-IRAS PX-SNX16 SNX29-PX PX-SNX34 Notes Each protein has a C-terminal BAR domain involved in membrane tubulation. Several also have N-terminal extended sequences, thought to be involved in protein–protein interactions (e.g. SNX1 and SNX2) These molecules are very similar in architecture to the PX-BAR proteins, except that they have an N-terminal SH3 domain that binds to polyproline sequences and an intervening low-complexity region containing AP2and clathrin-binding motifs SH3PXD2A (SH3MD1, FISH), SH3PXD2B, SNX28 SNX28, p47phox and p40phox are components of the NADPH oxidase complex. The PX-SH3 proteins have various numbers of SH3 domains C-terminal to the PX domain (from 1 to 5) (NOXO1), p47phox (NCF1), p40phox (NCF4) SNX26 (TC-GAP), PX-RICS (GC-GAP, p250GAP) The PX-SH3-GAP proteins have very long extended C-terminal domains with no identified structural motifs SNX17, SNX27, SNX31 The FERM domain has all three modules of a typical FERM domain (F1, F2 and F3), except that the F2 module is significantly smaller. SNX27 contains an additional N-terminal PDZ domain SNX13, SNX14, SNX19, SNX25 Each PXA-RGS-PX-PXC protein possesses two N-terminal hydrophobic stretches predicted to form transmembrane helices. Each PXA-RGS-PX-PXC protein possesses an RGS domain, except for SNX19 PXK, RPK118 (RPS6KC1), SGK3 (CISK) PXK and RPK118 are predicted to have non-catalytic serine/threonine-kinase domains. RPK118 may also possess an MIT domain PI3K-C2α, PI3K-C2β, PI3K-C2γ These are type II PI3K enzymes with multiple domains, including Ras-binding domains, C2 domains, a PI3K domain and a PI3KC2 homology domain of unknown structure and function PLD1, PLD2 These proteins have two separate membrane-recruitment domains (PX and PH) coupled to a PLD domain SNX20 (SLIC-1), SNX21 SNX20 and SNX21 are highly homologous and share a conserved helical domain (PXB) of ∼ 140 residues downstream of the PX domain. The PXB domain may be composed of up to three TPR helical repeats SNX3, SNX10, SNX11, SNX12, SNX22, SNX24, These proteins have been annotated as having no identified domains outside of the PX domain [3,37]. In all SNX29, HS1BP3 cases, the proteins possess short extra sequences without predicted secondary structure. The C-terminal tail of HS1BP3 is a proline-rich region SNX23 (KIF16B) SNX23 is a member of the kinesin family of microtubule motors. It includes an N-terminal kinesin motor domain, a putative forkhead association (FHA) domain and a central coiled-coil dimerization domain SNX15 SNX15 includes a C-terminal MIT domain IRAS (nischarin) IRAS is a large protein ∼ 1504 residues in length. Apart from the N-terminal PX domain, IRAS has a predicted leucine-rich repeat region from approximately residues 220 to 397, and secondary structure predictions indicate that the C-terminal region from approximately residue 570 onwards is composed of one or more structural domains with mixed α/β topologies. SNX16 SNX16 possesses significant predicted helical structure from between approximately residues 220 and 310 downstream of the PX domain SNX29 SNX29 possesses significant predicted helical structure between approximately residues 80 and 250 upstream of the PX domain SNX34 (C6ORF145) SNX34 has not been annotated previously and was identified in the present study via bioinformatics searches. It potentially contains a small C-terminal β-sheet domain between approximately residues 160 and 230 domain will allow us to discern common features governing their association, will lead to the identification of other interacting molecules, and will drive the design of specific mutants aimed at dissecting the functional importance of PX domain protein recruitment. THE MAMMALIAN PX PROTEIN FAMILY – AN UPDATE There are currently 49 known proteins that contain a PX domain encoded in the human genome. Most of these are modular proteins containing one or more additional domains with diverse functions, including membrane remodelling, phosphoinositide kinase activity, phospholipase activity, protein–protein interaction and numerous other functions both known and unknown. We have revisited recent catalogues of the PX domain proteins [3,37–40], and in Table 1 and Figure 2 we propose a modified classification based not only on sequence homology or known domains, but also on the presence of conserved structural elements defined by careful analysis of secondary-structure predictions. This reveals that in some cases proteins have been misclassified and in other cases there is strong evidence for the presence of conserved domains not previously characterized. The classification of PX proteins into related structural subfamilies provides an important basis for considering both the common and distinct functions of these diverse proteins. Some of these subfamilies contain many members, such as the PX-BAR and PX-only families, whereas c The Authors Journal compilation c 2012 Biochemical Society others contain only a single member. The majority of PX domain proteins have been named SNXs [3,37]. This terminology was originally used by Kurten et al. [41], after identification of SNX1, and later this nomenclature was refined to refer to proteins that have greater than 50 % sequence similarity to SNX1 in the PX domain [42,43]. A difficulty with this nomenclature arises from the fact that the classification of PX proteins as SNXs remains somewhat arbitrary. As just one example, it is not obvious why TC-GAP (TC10/cdc42 GAP) should also have the name SNX26, whereas GC-GAP (GAB/cdc42 GAP) does not have an SNX moniker. Therefore we propose that current common names for these proteins remain in place, but that a new SNX name is assigned for any future molecules identified for the PX family (e.g. C6ORF145, which we have named SNX34; see below). We have also produced a curated list of proteins that are found to associate with PX domain proteins and, where possible, have defined interacting domains, further highlighting the functional relationships between members of the same subfamilies (Supplementary Table S2). In constructing this Table, we restricted interactions to those that appeared to be direct as shown by immunoprecipitations or other assays, including yeast two-hybrid, pull-downs with purified proteins or cocrystal structures. Importantly, these analyses also reveal that ligands for PX proteins can be assigned to several major functional groups: transmembrane cargo molecules, regulators of intracellular membrane trafficking, cellular signalling and cytoskeletal reorganization/attachment. The PX protein family Figure 2 43 Classification and domain organization of human PX proteins Structural classification of PX proteins was based on known domains, and novel conserved domains were identified by secondary-structure prediction and sequence comparison. Note that the diagrams are not to scale. Domains with broken outlines indicate a domain that is only found in some members of the subfamily (PDZ domain in SNX27; MIT domain in RPK118; RGS domain missing in SNX19; TM domains missing in SNX25), or found in variable numbers (SH3 domains of PX-SH3 proteins). PX-BAR and SH3-PX-BAR subfamilies The subfamily of PX proteins possessing a C-terminal BAR domain are perhaps the best characterized of all PX subfamilies. An excellent review by van Weering et al. [39] highlights their importance in endocytic and endosomal membrane sorting processes. Sorting of proteins by PX-BAR molecules is dependent on the membrane remodelling and sensing properties of the BAR domain, which drives the formation of membrane tubules and preferentially associates with curved membrane domains [44,45]. Examples include the formation of endosomal transport tubules by SNX1 [46] and membrane deformation during clathrin-coated vesicle formation by SNX9 [18,47]. Therefore the overarching question about the PX-BAR proteins is this: why are there so many? Does each protein control the sorting of cargo molecules within specific membrane domains, are they regulated by different signals or do they function in different intracellular compartments? Attempts to answer these questions have borne significant fruit in the last few years; however, dissecting the individual roles of PX-BAR proteins is severely complicated by the requirement that they form homo- or hetero-dimers (a feature of the BAR domain) and higher-order oligomeric complexes during membrane tubulation. It is plain that in some cases different PX-BAR proteins can have overlapping functions. For example, one of the bestcharacterized roles of the PX-BAR proteins is to co-operate with the retromer protein complex in endosome-to-TGN (trans-Golgi network) recycling of a variety of transmembrane cargo proteins. Retromer is a heterotrimeric assembly composed of the proteins VPS35 (vacuolar protein sorting-associated protein 35), VPS29 and VPS26 thought to participate in cargo loading into membrane tubules coated by PX-BAR proteins, including SNX1, SNX2 c The Authors Journal compilation c 2012 Biochemical Society 44 R. D. Teasdale and B. M. Collins SNX5 and SNX6 [40,48,49]. The highly homologous SNX1 and SNX2 have been shown to participate somewhat interchangeably in retrograde endosomal protein trafficking of both the bacterial toxin StxB (Shiga-like toxin β subunit), where siRNA (small interfering RNA) knockdown of both proteins results in a greater defect in protein trafficking than that of either one alone [50], and the CI-MPR (cation-independent mannose-6-phosphate receptor) whereby knockdown of both proteins together is required to observe a defect in retromer membrane tethering and CI-MPR endosome-to-Golgi retrieval [51]. Each protein has been shown to both self-associate and form heterodimers with each other, suggesting that they have very similar biochemical properties [51–56]. These data are somewhat clouded, however, by separate reports that SNX2 does not participate in either trafficking process [46,57,58], whereas other evidence suggests that SNX1 and SNX2 can also have independent functions, as shown by their differential roles in down-regulation of activated EGFR (EGF receptor) or PAR1 (protease-activated receptor 1) [52,59]. Perhaps the most compelling evidence for functional overlap is that deletion of both SNX1 and SNX2 in mice results in a lethal phenotype, strongly implying that the two proteins can functionally compensate for each other to a certain extent [60]. Other examples of PX-BAR proteins with overlapping functions include the highly similar SNX5 and SNX6 proteins, as well as the endocytic SH3 domain-containing proteins SNX9, SNX18 and SNX33. In the former case, both SNX5 and SNX6 were found to play similar roles in the endosome-to-Golgi retrieval of the CI-MPR, and both form heteromeric complexes with the retromer-associated SNX protein SNX1 [24,56,61]. Interestingly, SNX5 has been shown to have a significantly different membrane recruitment response to EGFR-mediated signalling, with SNX5 showing both endosomal and plasma membrane localization in stimulated cells, perhaps regulated by an affinity for PtdIns(4,5)P2 , whereas SNX1 remains endosomally bound [22,62]. This is strong evidence that PX-BAR proteins may have differing functions depending on whether they are partnered with one particular PX-BAR protein or another. SNX9 plays a well-established role in clathrin-mediated endocytosis, in part controlling the membrane remodelling of the narrow neck region of the forming vesicle, and acting as a molecular scaffold for assembly and recruitment of essential vesicle components, such as dynamin, AP2 (adaptor protein complex 2), clathrin and components of the actin cytoskeleton (reviewed in [47]). Studies of SNX18 and SNX33 indicate that they have overlapping, but also distinct, intracellular localizations with SNX9 [63–65]. However, all bind to dynamin and the effector WASP, and SNX9 and SNX33 have a similar ability to affect endocytosis of APP (amyloid precursor protein) [66]. The data are conflicting as to whether these proteins can form heteromeric assemblies with each other [63–65], but these initial studies suggest that the three proteins probably have both overlapping and distinct roles in dynamin-dependent endocytosis and protein trafficking. The functional interplay between the different members of the PX-BAR subfamilies is therefore highly complex, and the task now will be to dissect both the common and unique properties of each PX-BAR protein and protein assembly. Ongoing goals will be to determine the repertoires of functional PX-BAR dimeric complexes, examine which of these possess inherent membrane associating and tubulating properties, and how these complexes correlate to the in vivo assemblies based on criteria such as overlapping tissue distribution and cellular colocalization. Studies of the SNX9 PX and BAR domains provide important details on how dimeric SH3-PX-BAR complexes are assembled and how their structures contribute to membrane attachment and c The Authors Journal compilation c 2012 Biochemical Society remodelling (Figure 1B) [18,67]. An interesting finding was the presence of a so-called ‘yoke’ domain, which is critical for holding the PX domain in a fixed orientation with respect to the BAR domain. Although it is assumed that related PX-BAR proteins will adopt overall similar tertiary assemblies, the recent structure of the SNX5 PX domain may have important implications for the assembly of SNX5 into functional complexes [22]. SNX5 possesses a novel helix–turn–helix insertion following the polyproline loop that forms an extended structure protruding from the PX domain. When the SNX5 PX domain is aligned with SNX9, the helical insertion protrudes in such a way that it will penetrate significantly into the predicted plane of the membrane (Figure 1B). This suggests that either the PX domain is oriented differently in SNX5 compared with SNX9, or that SNX5 may have different membrane-modulating properties compared with related SH3-PX-BAR proteins. High-resolution studies of homoand hetero-dimeric PX-BAR complexes will be required for a firm understanding of their assembly and membrane interaction. To generate or sense tubular membrane structures, BAR domain proteins must organize into polymeric arrays, involving lateral interactions between membrane-associated dimers. Multiscale structural characterization, such as performed in landmark studies of F-BAR proteins (where F is the FCH domain) [68], will hopefully provide a more detailed understanding of the membrane association and higher-order oligomerization properties of PXBAR proteins. An important question, as outlined previously [37,39], is how different SNX proteins may control formation or sorting into alternative membrane transport tubules in vivo. For example, how exclusive are PX-BAR membrane tubules? Can mixtures of different PX-BAR proteins exist on the same tubules, and how do cargo receptors and accessory proteins (for example the retromer complex) integrate into these membrane domains? All of this information will ultimately help us to understand how different homo- and hetero-dimeric combinations of PX-BAR proteins control protein sorting within the endocytic system. Other questions of immediate interest include determining the function of the uncharacterized PX-BAR proteins. For example, does SNX32 participate in endosomal retrograde transport similarly to the homologous proteins SNX5 and SNX6? Given the highly dynamic nature of PX-BAR- and SH3-PX-BAR-derived membrane structures, another question of prime interest will be to determine how these proteins associate with other regulatory molecules and with cargo receptors themselves (Supplementary Table S2). For example SNX1 was originally identified by its ability to associate with EGFR and other signalling receptors [41,54], and with the protein Hrs (hepatocyte growth factorregulated tyrosine kinase substrate) [69]. Subsequent studies have also suggested direct interactions between SNX1 and GPCRs (G-protein-coupled receptors) [59,70]. Perhaps the bestcharacterized interaction of SNX1 is with the retromer complex [51–53,71], but even in this case the mechanism of association is very poorly understood. One of the most exciting discoveries has been the identification of interactions between endosomal PXBAR proteins and components of the cytoskeleton [39,56,72]. These findings demonstrate the importance of dynein-mediated microtubule translocation for PX-BAR trafficking processes and have opened an important area of investigation for these molecules. PX-FERM subfamily The PX-FERM molecules serve as a prime example of the important endosomal scaffolding and trafficking functions of many PX proteins. Recent work from our laboratory has shown The PX protein family Figure 3 A network of molecular interactions regulated by the PX-FERM proteins in endosomal membrane transport and signalling The PX domain binds to PtdIns3P with high specificity, and the similarity of the F3 module of the FERM domain suggests that it will be responsible for binding cargo receptors via NPxY sequences. The proteins can bind Ras-GTP, and this is proposed to be via the F1 module similar to structurally related Ras-association domains. SNX17 has been reported to use its polyproline sequence within the PX domain to interact with a variety of SH3 domain proteins, including Src-related kinases and PLCγ 1. SNX27 possesses an additional N-terminal PDZ domain that binds cytosolic and transmembrane proteins via C-terminal type I PDZbms, ExE[S/T]x[F/V]. β2-AR, β 2 adrenergic receptor; CNKRS2, connector enhancer of kinase suppressor of Ras2; FEEL1, fasciclin, EGF-like, laminin-type EGF-like and link domain-containing scavenger receptor 1 (also called stabilin-1); Grb2, growth-factor-receptor-bound protein 2; MAP1A, microtubule-associated protein 1A; Nck1, non-catalytic region of tyrosine kinase adaptor protein 1; PTCH1, protein patched homologue 1; PTPN2, protein tyrosine phosphatase non-receptor type 2; RhoGDI, Rho GDP-dissociation inhibitor; VLDLR, very-low-density lipoprotein receptor; vl-GPCR, very large GPCR. that three proteins, SNX17, SNX31 and SNX27, constitute a conserved subfamily containing a C-terminal FERM domain [11]. In addition SNX27 possesses an N-terminal PDZ (postsynaptic density 95/discs large/zonula occludens) domain. FERM domains are approximately 300 amino acids in length and are found in numerous signalling and scaffolding proteins, where they serve a role in membrane tethering and/or binding to the cytosolic domains of transmembrane proteins [73]. The FERM domain is composed of three subdomains or modules named F1, F2 and F3. The F1 subdomain possesses a ubiquitin-like fold and the C-terminal F3 subdomain possesses a similar structure to the PH and PTB domains. The F1 and F3 modules are oriented with relation to each other by the central F2 subdomain composed of four α-helices. The sequence homology of the PX-associated FERM domains with the canonical FERM domain is low, and the central F2 subdomain is significantly smaller than normal [11]. The PX domains are essential for endosomal localization via PtdIns3P binding [11,74–80], and the FERM domains of these proteins regulate endosomal cargo interactions and, possibly, scaffolding of signalling complexes [11] (Figure 3). 45 SNX17 and SNX31 are the most closely related within this subfamily. There are currently no published data on the function of SNX31, but several groups have determined that SNX17 is endosome-associated and controls intracellular trafficking of a number of transmembrane cargo proteins. Proteins whose cellular localization is regulated by SNX17 include LRP1 [LDLR (lowdensity lipoprotein receptor)-related protein 1] [76,81–83], LDLR [83,84], P-selectin [75,85,86] and APP [87]. Overexpression of SNX17 enhances LDLR endocytosis [83], whereas expression of a truncated SNX17 molecule causes LDLR to be mislocalized [84]. It was subsequently found that SNX17 regulates the recycling of LDLR from endosomes to the cell surface in nonpolarized cells [76] and from basolateral sorting endosomes to the basolateral plasma membrane in polarized MDCK (Madin– Darby canine kidney) cells [82]. The role of SNX17 in trafficking of LDLR and related lipoprotein receptors suggests a potentially central function in lipid metabolism, and it will be important to determine whether this is indeed the case. SNX17 was also found to promote cellular uptake of the endothelial receptor P-selectin and, additionally, it colocalizes with P-selectin on endosomal organelles where it is able to inhibit its transport to late endosomes/lysosomes for degradation [75]. Finally, SNX17 is expressed in neurons, associates with APP on endosomes and inhibits its proteolytic processing to the toxic amyloid Aβ peptide [87]. The role of PX proteins in inflammation and AD (Alzheimer’s disease) is discussed further below. In addition to transmembrane cargo proteins, SNX17 has been found to associate with the cytosolic protein Krit1 (Krevinteraction trapped 1), which is able to enhance recruitment of SNX17 to endosomes [74]. SNX17 binds receptors and effectors via short peptide motifs with the consensus Asn-Pro-Xaa-Tyr (NPxY) via its C-terminal FERM domain. In addition to these NPxY-motif interactions, a proteomics study has also identified SNX17 as a ligand for various SH3 domain-containing proteins, including a number of Src-related tyrosine kinases and PLCγ 1 [88]. In these cases, the interaction involves the SH3 domain binding directly to the SNX17 PX domain polyproline loop. The determination that SNX27 shares structural similarity with SNX17 and SNX31 led to the demonstration that it also possesses NPxY-motif binding affinity, although it has yet to be confirmed that SNX27 is able to direct endosomal sorting of NPxY cargo proteins [11]. SNX27 was annotated as possessing a C-terminal RA (Ras-association) or B41 domain [3,37,43]. However, it is now clear that it possesses a similar FERM C-terminal domain to SNX17 and SNX31. This confusion is due to the fact that both RA and B41 domains share a ubiquitin-like fold, as does the F1 module of FERM proteins. Nonetheless, as mentioned above, the F1 module of FERM domains shares structural similarity with the RA domain, and this prompted an investigation of the Ras-binding properties of the PX-FERM proteins [11]. These preliminary experiments show that PX-FERM proteins are able to bind the activated H-Ras GTPase, and suggest a potential link between endosomal Ras-mediated signalling and membrane trafficking that requires further investigation [89]. A hint that the PX-FERM proteins can indeed affect Ras-mediated signalling comes from recent studies showing that SNX27 knockdown by siRNA results in enhanced ERK (extracellular-signal-regulated kinase) phosphorylation [79]. However, the actual Ras isoform(s) targeted by the PX-FERM proteins in vivo remains unknown. SNX27 is unique amongst the PX-FERM proteins in that it also contains a PDZ domain upstream of the PX domain. Like SNX17, SNX27 is highly expressed in brain tissue, and was in fact first identified as a molecule up-regulated upon stimulation of dopamine receptors with methamphetamines [90–92]. The PDZ domain of SNX27 has been implicated in binding to a number c The Authors Journal compilation c 2012 Biochemical Society 46 R. D. Teasdale and B. M. Collins of proteins via a C-terminal type I PDZbm (PDZ-binding motif) (E[S/T]x[V/F]), and structural studies of the SNX27 PDZ domain show that the preferred motif is enhanced by the presence of an upstream glutamic acid side chain (ExE[S/T]xV/F]) [90]. The first identified binding partner for SNX27 was 5-HT4 R (5-hydroxytryptamine type 4 receptor), a GPCR involved in feeding and respiratory control [77]. SNX27 was found to be essential for transport of 5-HT4 R to EEA1-positive early endosomes. DGKζ (diacylglycerolkinase ζ ) was identified as a SNX27 binding partner by proteomics and can regulate SNX27 association with early and recycling endosomes [80]. Other proteins found to associate with the SNX27 PDZ domain are CASP (cytohesinassociated scaffolding protein), a cellular adaptor protein found in cells of haemopoietic origin [93,94], Kir3, G-protein-gated inwardly rectifying potassium channels that control a slow inhibitory postsynaptic response in neurons [78,90,95], and most recently the NR2C [NMDA (N-methyl-D-aspartate) receptor 2C] ligand-gated ion channel [96]. SNX27 and Kir3 channels show colocalization in endosomes, and overexpression of SNX27 results in reduced Kir3.3 expression at the cell surface, suggesting that it either promotes Kir3.3 endocytosis or inhibits its recycling. Finally, a recent paper by Lauffer et al. [97] demonstrated that SNX27 can bind to β2AR (β 2 -adrenergic receptor) via its C-terminal PDZbm and regulate its endosomal recycling to the cell surface, perhaps analogously to the recycling role demonstrated for SNX17. The exact function of the PX-FERM molecules in protein trafficking remains unclear and their mechanisms of action are unknown. Do they form a protein coat regulating membrane transport, do they act as adaptors for other membrane scaffolding molecules or do they play a facilitating role by concentrating transport cargoes into membrane domains destined for recycling to the cell surface? The first clue to the function of these proteins emerged with the identification of SNX27 as a binding partner for the WASH [WASP and SCAR (suppressor of cAMP receptor) homologue] complex [98]. The WASH complex is an endosomeassociated assembly that induces actin polymerization in an Arp2/3 complex (actin-related protein 2/3 complex)-dependent manner, and has an important function in endosomal trafficking and membrane dynamics (reviewed in [99]). Interestingly, WASH is believed to co-ordinate endosomal sorting in association with the retromer complex [100,101]. Knockdown of retromer perturbs cell-surface recycling of the β 1 - and β 2 -adrenergic receptors similarly to SNX27, and SNX27 was found to be associated with retromer after cross-linking [98]. The authors propose that SNX27 can act as a cargo-specific adaptor for endosomal sorting of PDZbm-containing receptors within retromer-coated membrane tubules, and also propose a role for retromer and SNX27 in rapid cell-surface recycling via a Rab4-dependent pathway, which contrasts significantly with the canonical role of retromer in endosome–TGN retrograde transport. This specific process is unique to SNX27 as it only operates on PDZbm-containing proteins, so it remains to be seen whether PX-FERM molecules also co-ordinate endosomal trafficking of NPxY cargo, such as APP and LDLR family members, with WASH and retromer. SNX14 and SNX25. Two other domains (each approximately 150 amino acids) have previously been annotated as the PXA domain (PX-associated domain A) lying N-terminal to the RGS and canonical PX domains, and the C-terminal PX-associated domain lying downstream of the PX domain [3,37]. For clarity, we refer to the N- and C-terminal domains as the PXA and PXC (PX-associated domain C) domains respectively, to denote that they are as yet uncharacterized PX-associated domains. Finally, each member of the family possesses two putative N-terminal transmembrane helices resulting in a predicted topology of a short cytoplasmic leader, two closely spaced transmembrane domains, and a long C-terminal structure containing the three (SNX19; PXA-PX-PXC) or four (SNX13, SNX14 and SNX25; PXA-RGS-PX-PXC) modular domains (Figure 4B). Currently, SNX25 is predominantly annotated as lacking transmembrane sequences, but our analyses of the rat, mouse and human genomes identified a conserved N-terminal coding sequence that is clearly transcribed and encodes two potential transmembrane helices. There are only a small number of reports that address the function of the PXA-RGS-PX-PXC proteins. These describe the role of SNX13 (also called RGS-PX1) as a GAP and regulator of the trimeric G-protein subunit Gα s via its RGS domain [102–104], and of SNX25 as a modulator of TGF-β signalling [105]. The former studies confirmed SNX13 as an important member of the family of RGS proteins, a family with diverse roles in modulating GPCR-mediated signalling [106,107]. Initial work identified SNX13 after database searches for the putative GAP for Gα s , which at the time had not been identified. SNX13 was subsequently cloned and shown to both bind to and promote GTPase activity of Gα s with high specificity and attenuate Gα s -mediated signalling [103]. Endosomes are increasingly being recognized as critical sites of signal regulation [1,2,108], and the affinity of SNX13 for PtdIns3P and localization to endosomes suggests a potential role in GPCR signal attenuation in this compartment. SNX13 was subsequently shown to form a heteromeric complex with both Gα s and Hrs on endosomes and co-operate in the lysosomal targeting of the EGFR, suggesting a role both in signalling and trafficking [102]. A model for SNX13 function in endosomal signal attenuation is presented in Figure 4(B). SNX13-null mice show embryonic lethality, and yolk-sac endoderm cells of these embryos display highly disrupted endosome morphology, demonstrating that it plays a role in endosomal function that is critical for normal development and that other PXA-RGS-PX-PXC proteins are unable to compensate for its loss [104]. SNX25 was identified through bioinformatics searches for novel PX domain proteins and is proposed to modulate TGF-β signalling via endosomal sorting of TGF-β receptors for lysosomal degradation [105]. Another study has implicated SNX19 in chondrogenic regeneration in cartilage, although the mechanism for this function is not proposed [109]. A final interesting finding is that the Drosophila protein snazarus (sorting nexin lazarus), for which SNX25 is the closest homologue, plays a role in regulating longevity, as snazarus mutants display significantly increased lifespans [110]. PX-only and PX proteins with putative novel domains PXA-RGS-PX-PXC subfamily Four proteins, SNX13, SNX14, SNX19 and SNX25, display a unique architecture consisting of a central PX domain flanked by several conserved domains (Figure 4A). The first domain identified was the RGS (regulator of G-protein signalling) domain, found in a number of molecules that attenuate GPCR and related G-protein signalling. This domain is seen only in SNX13, c The Authors Journal compilation c 2012 Biochemical Society The subfamily of proteins that have been annotated as PX-only proteins is a relatively poorly characterized group of molecules [3,37]. As the name suggests, bioinformatics analyses do not detect any conserved domains outside the defining PX domain. Structurally, however, these proteins are of various lengths and typically contain long extended sequences with no predicted secondary structure. In several cases, such sequences have The PX protein family Figure 4 47 Architecture and interactions of PXA-RGS-PX-PXC proteins (A) Schematic diagrams of the human PXA-RGS-PX-PXC proteins SNX13, SNX14, SNX19 and SNX25. Transmembrane (TM), N-terminal PXA, RGS, PX and C-terminal PXC domains are indicated with shaded boxes. Proteins and domains are drawn to scale. (B) Model for SNX13 function in GPCR signalling [103]. SNX13 is predicted to possess two transmembrane domains. After activation of a GPCR by ligand binding, GDP is displaced and exchanged for GTP in the Gα s subunit of the trimeric G-protein, releasing the Gβ and Gγ subunits and resulting in signal transduction. The RGS domain of SNX13 binds to Gα s and SNX13 acts as a GAP to stimulate GTPase activity. The G-protein complex can then reform and signal transduction is ablated. Because SNX13 has a PtdIns3P -binding PX domain and is localized to endosomes, it probably functions in signal attenuation in this intracellular compartment and also interacts with Hrs, a component of the ubiquitin-mediated protein sorting and degradation pathway in late endosomes. been found to be critical for function [111,112]. Of the PXonly proteins, SNX3 is perhaps the best characterized. Like the majority of PX proteins, SNX3 binds to PtdIns3P via its PX domain and this interaction drives its association with the limiting membrane of early endosomes [113] and MVBs (multivesicular bodies) [114]. Overexpression of SNX3 was found to alter endosome morphology [113], and the protein is essential for the formation of the intralumenal vesicles of MVBs [114]. Ubiquitylation is a critical feature of protein sorting into MVBs, and SNX3 itself has been found to be ubiquitylated, with its stability being regulated by the de-ubiquitylating enzyme Usp10 [115]. SNX3 has also been linked to neurite outgrowth, and its expression is induced by lithium treatment [116]. The yeast homologue Grd19p/Snx3p appears to co-operate with the retromer coat complex in endosome-to-Golgi retrieval [117–119], and elegant studies have recently shown that it also co-operates with retromer in endosome-to-Golgi recycling of the Wnt receptor Wntless in Caenorhabditis elegans, Drosophila melanogaster and mammalian cells, indicating an evolutionarily conserved function in retromer-mediated membrane trafficking [120]. HS1BP3 (HS1-binding protein 3) is a unique PX domain protein identified by proteomic screening as a binder of the HS1 protein, which in turn is a substrate of the Lck tyrosine kinase involved in T-cell differentiation [121]. HS1BP3 has an extended C-terminal region rich in proline residues that is bound by the HS1 SH3 domain. Expression of a truncated HS1BP3 construct resulted in reduced IL-2 (interleukin-2) production, suggesting a role in T-cell-receptor-mediated signalling, and genetic studies also indicate that a variant (A265G) is associated with familial essential tremor [122–124]. The only other PX-only protein that has been characterized to any extent is SNX10, which induces the formation of giant vacuoles when overexpressed [111]. This vacuolization is sensitive to disruption of the Golgi, suggesting that SNX10 may in some way enhance the fusion of Golgi-derived vesicles with endosomes. In addition to the PX-only proteins, several PX molecules possess regions of predicted secondary structure that almost certainly form folded domains of unknown function. SNX20 and SNX21 are a prime example. These proteins are highly homologous and share a conserved domain of ∼ 140 residues containing six α-helices downstream of the PX domain on the basis of secondary-structure predictions (Supplementary Figure S2 at http://www.BiochemJ.org/bj/441/bj4410039add.htm). We propose that this be named the PXB domain (PX-associated domain B), continuing the nomenclature adopted for the PXA-RGSPX-PXC proteins. Results from the TPRpred server suggest that this region may contain up to three TPRs (tetratricopeptide repeats), α–α-hairpin structures that form helical solenoids [125]. SNX16 possesses a predicted helical structure between residues ∼ 220 and 310 C-terminal to the PX domain. SNX29 has significant predicted helical structure between residues ∼ 80 and 250 upstream of the PX domain. Therefore technically, SNX16, SNX29, SNX20 and SNX21 proteins should not be classified as ‘PX-only’ molecules. During bioinformatic searches we also identified the novel open reading frame C6ORF145, which we have designated SNX34. SNX34 is 231 residues in length and secondary-structure predictions indicate the presence of a small domain composed of β-strands at the C-terminus (residues 160– 225). SNX16 associates with endosomes via PtdIns3P binding to its PX domain, and the helical C-terminal domain appears to be important for self-association, possibly by forming a coiled-coil [126,127]. This domain regulates SNX16 distribution between early and late endosomes and is important for processing of c The Authors Journal compilation c 2012 Biochemical Society 48 R. D. Teasdale and B. M. Collins cargo molecules, such as EGFR, into the late compartments. Interestingly, SNX16 was recently found to be a PtdIns3P effector that is able to inhibit the export of viral RNA into the cytoplasm from late endosomes, although the mechanism of function is unknown [128]. A recent study of D. melanogaster SNX16 identified an interaction with the WASP-activating F-BAR molecule Nwk on recycling endosomes, and highlighted a role for this interaction in down-regulating BMP (bone morphogenetic protein) and Wg (Wingless) synaptic growth signalling pathways in nerve terminals [129]. One final PX protein of interest with novel structural domains is IRAS (imidazoline receptor antisera-selected), also called nischarin in mice. IRAS/nischarin is a large protein of 1504 amino acids. Apart from the N-terminal PX domain, IRAS/nischarin also has a predicted leucine-rich repeat region (residues ∼ 220–397). Leucine-rich repeats display a characteristic three-dimensional structure composed of repeating β-strands and α-helices, and often participate in protein–protein interactions [130]. In addition, secondary-structure predictions indicate that the C-terminal region of IRAS/nischarin (from residue ∼ 570 onwards) is composed of one or more structural domains with mixed α/β topologies. IRAS/nischarin was originally cloned as a putative imidazoline receptor, although it is probably not a bona fide receptor [131], and is localized to endosomes in HEK (human embryonic kidney)-293 cells via its PX domain [132]. It was subsequently found to bind to IRSs (insulin receptor substrates) and enhanced IRS-dependent insulin stimulation of ERK [133]. A number of other papers have centred on the ability of IRAS/nischarin to bind to α5 integrins [132,134–138]. IRAS/nischarin binds to a short peptide in the cytoplasmic tail of α5 integrin via a C-terminal region (residues ∼ 710–810) [132]. It also regulates Rac-induced cell migration and Rac/PAK (p21activated kinase) signalling [134,135,137,138], although these data must be viewed with some degree of caution as all of these studies employed a mouse version of IRAS/nischarin that lacks the N-terminal PX domain [132,135], apparently due to a truncation in the original IRAS/nischarin clone. The current mouse IRAS/nischarin entry in the NCBI database (accession number NM_022656) has the PX domain and is homologous with the full-length human gene. It is possible, therefore, that the previous studies of IRAS/nischarin function in Rac signalling employed a dominant-negative version of the protein. Other unique PX proteins SNX15, which includes a C-terminal MIT domain (microtubuleinteracting and trafficking molecule domain), was first identified via database searches for SNX1 homologues, and despite the lack of a C-terminal BAR domain for dimerization was found to associate with SNX1, SNX2 and SNX4 [139]. It was also found to associate with the receptor for PDGF (plateletderived growth factor), and interestingly all of these identified interactions were dependent on the PX domain itself. SNX15 is localized to endosomes and overexpression of the protein has numerous phenotypic consequences, including disrupted endosomal morphology, reduced processing of several growth factor receptors and inhibited trafficking of several proteins such as furin, TGN38 and transferrin [139,140]. Although its exact function is not clear, it appears that SNX15 plays an important role in endosomal transport processes. MIT domains are found in a number of endosomal proteins, including Vps4 {an ATPase that controls the disassembly of the ESCRT (endosomal sorting complex required for transport) complex [141]}, spastin and another PX protein, the serine/threonine-pseudokinase RPK118 c The Authors Journal compilation c 2012 Biochemical Society [142]. The structure of the MIT domain reveals a small three-helix bundle, and its function appears to be to act as a protein–protein interaction module, specifically binding small MIT-interaction motifs [143,144]. The exact role of the SNX15 MIT domain, however, is unknown. KIF16B (kinesin-family protein 16B, also known as SNX23) is a member of the kinesin superfamily of proteins [145]. It contains kinesin motor, FHA (forkhead association) and coiledcoil dimerization domains. KIFs are microtubule-binding motors with numerous roles in the transport of organelles, protein complexes and RNA [146,147]. KIF16B is a plus-end-directed microtubule motor that associates with PtdIns3P-containing endosomes via its C-terminal PX domain and regulates the localization of endosomal organelles in the cell as well as receptor recycling within the endosomal system [145,148]. EVOLUTIONARY INSIGHTS FROM THE YEAST PX PROTEOME The conservation of PX proteins in yeast and other simple eukaryotes demonstrates a deep evolutionary heritage in eukaryotes, in line with their critical function in membrane trafficking, organization and cell signalling. Several interesting observations are made when the human PX proteome is compared with that of yeast. First, it is clear that higher eukaryotes have evolved an arsenal of PX domain proteins more diverse than those of their single-celled cousins. Secondly, it is seen that not all subfamilies of PX proteins are found in all eukaryotes. There are 15 known PX domain proteins in yeast, divided into several subfamilies that generally reflect those found in humans (Supplementary Table S3 at http://www.BiochemJ.org/bj/441/bj4410039add.htm). The most populous subfamily consists of the PX-BAR molecules (seven in yeast compared with nine in humans). Homologues are also seen for the PX-PH-PLD (Pld1p), PXA-RGS-PXPXC (Mdm1p) and PX-only subfamilies (Snx3p and Ypt35p). However, in yeast each of these families has only one or two members. Yeast does have a PX domain-containing GAP, Bem3p; however, this molecule has a different architecture to the PX-SH3GAP proteins found in humans, replacing the central SH3 domain with a membrane-binding PH domain. Many PX subfamilies found in humans are not identified in yeast, including the PX-FERM, PX-S/T-kinase (serine/threonine kinase) and SH3-PX-BAR proteins; conversely, yeast has three PX proteins not identified in humans. The SNARE (soluble N-ethylmaleimide-sensitive factor-attachment protein receptor) protein Vam7p has, in addition to the canonical PX domain, C-terminal t-SNARE (target SNARE) motifs for forming SNARE complexes with Vam3p and Vti1p during vacuolar membrane fusion. The SH3-PX-CAD (caspase-activated DNase) protein Bem1p has two SH3 domains N-terminal to the PX domain and a C-terminal CAD/PB1 domain, which belongs to the ubiquitinfold superfamily. The poorly characterized Ypr097Wp is a large protein and appears to contain a highly divergent PX domain on the basis of sequence and secondary-structure predictions (see [9]). Outside the PX domain, it is predicted to be primarily α-helical in structure, although no other known domains can be identified. Overall, it appears that eukaryotic organisms have employed both overlapping and distinct PX protein families throughout evolution. PX PROTEINS AND DISEASE There are now many reported links between PX proteins and various disease processes (Table 2). Currently, there is only one known case where a specific PX protein mutation is known to be The PX protein family Table 2 49 PX proteins implicated in disease Family PX protein Disease References PX-BAR SNX1 STx uptake Salmonella uptake Down-regulated in ovarian cancer Gefitinib-sensitive non-small lung cancer Down-regulated in colon cancer Aβ production (AD) STx uptake Salmonella uptake Epilepsy – Uptake of Ebola virus via macropinocytosis Aβ production (AD) – STx uptake – – EPEC (enteropathic Escherichia coli ) infection Aβ production (AD) – Prion disease mediated by PrPc (normal cellular prion protein) Aβ production (AD) Aβ production (AD) Tumour metastasis – – – Chronic granulomatous disease – – Aβ production (AD) – – – – Candidate gene involved in Jacobsen syndrome May be a chondrogenic factor in osteoarthritis Genetic risk factor in myocardial infarction Transcriptionally up-regulated in thyroid oncocytic tumours Potential oncogene in myeloid leukaemia Homozygous deletion of 3 exons found in B-cell non-Hodgkin’s lymphoma cell lines Systemic lupus erythematosus Akt-independent cancer cell survival Up-regulated and promotes cell survival in oestrogen-receptor-positive breast cancer – Prostate cancer risk Up-regulated in some glioblastoma brain tumours – Aβ production (AD) Up-regulated in endometrial cancer cells Invasion by metastatic breast cancer cells Colon cancer Aβ production (AD) Invasion by metastatic breast cancer cells Metastatic mammary adenocarcinoma Lymphoma cell proliferation Colon cancer – – Salmonella uptake An effector of lithium treatment for bipolar disorder, and a regulator of neurite outgrowth Gene disrupted in MMEP – – – – Oestrogen-regulated expression in breast cancer cell lines Familial essential tremor – [50,57,58,221] [212] [224] [225] [166,167] [186] [50] [213] [156] SNX2 SH3-PX-BAR SNX4 SNX5 SNX6 SNX7 SNX8 SNX30 SNX32 SNX9 SNX18 SNX33 PX-SH3 PX-SH3-GAP PX-FERM PXA-RGS-PX-PXC PX-S/T kinase PI3K-PX PX-PH-PLD SH3PXD2A (SH3MD1, FISH, Tks5) SH3PXD2B (Tks4) SNX28 (NOXO1, p41NOX, SH3PXD5) p40phox (NCF4, SH3PXD4) p47phox (NCF1, SH3PXD1A) SNX26 (TC-GAP) PX-RICS (GC-GAP, p250GAP, p200GAP, Grit) SNX17 SNX27 SNX31 SNX13 (RGSPX1) SNX14 SNX19 SNX25 PXK (MONaKA) RPS6KC1 (RPK118, humS6PKh1) SGK3 (CISK, SGKL) PIK3C2A (PI3K-C2α) PIK3C2B (PI3K-C2β) PIK3C2G (PI3K-C2γ ) PLD1 PLD2 PX-PXB PX-only SNX20 SNX21 SNX3 Kinesin-PX SNX10 SNX11 SNX12 SNX22 SNX24 HS1BP3 KIF16B (SNX23) [223] [190] [222] [226,227] [66] [66,228] [66] [199,200] [163,164] [149–151] [87] [229] [109] [230] [231] [232] [233] [155] [168] [169] [165] [234,235] [202–206] [236] [237–240] [241] [207] [240,242] [162] [243] [244,245] [213] [112,116] [153,154] [246] [122–124] c The Authors Journal compilation c 2012 Biochemical Society 50 Table 2 R. D. Teasdale and B. M. Collins Continued Family PX protein Disease References PX-MIT SNX15 [162] [213] PX-LRR-IRAS PX-SNX16 IRAS (nischarin) SNX16 SNX29-PX PX-SNX34 SNX29 SNX34 (C6ORF145) Metastatic mammary adenocarcinoma Salmonella uptake – VSV infection Differential expression used as a biomarker in bladder cancer Undergoes alternative splicing in certain melanoma cell lines – – the causative agent of a disorder: mutation of the p47phox subunit of the NADPH oxidase in CGD (chronic granulomatous disease) [149–151]. Patients with CGD have a primary failure in neutrophil function and do not mount the normal NADPH oxidase-mediated respiratory burst required for phagocytosis. In particular, it is known that the R42Q mutation is implicated in CGD and that this mutation disrupts the phosphoinositide-binding site and membrane coupling of the protein [152]. Another PX protein, SNX3, was found to be disrupted in a patient with MMEP (microcephaly, microphthalmia, ectrodactyly and prognathism) [153]; however, subsequent studies were unable to detect coding mutations in SNX3 in MMEP patients [154], so it remains to be determined whether SNX3 is directly involved in this family of diseases. Other PX molecules have been implicated in disease on the basis of differential transcription or protein expression. PXK (PX domain-containing serine/threonine-kinase) has been linked to SLE (systemic lupus erythematosus) by genome-wide association studies [155]. SNX19 was found to be up-regulated in cartilage cells during progression of osteoarthritis, and studies in a model chondrogenic cell line implicated it as a protective factor against cartilage degradation [109]. SNX2 was also isolated in a screen for genes with potential involvement in epilepsy, but it remains to be confirmed that SNX2 is important for the disease [156]. Several reviews highlight the critical role of dysregulation of phosphoinositide homoeostasis in many different disorders [157– 160]. Enzymes that regulate phosphoinositide generation and breakdown (kinases and phosphatases) are found to be mutated in a number of diseases. Of particular relevance to the PX proteins are disorders relating to enzymes regulating PtdIns3P synthesis at the early endosome, PtdIns(3,5)P2 generation from PtdIns3P at late endosomes and their respective levels during endosomal maturation (reviewed in [160]). These include disorders such as the family of Charcot–Marie–Tooth neuropathies caused by mutations in the 5-phosphatases (Sac3/Fig4 and myotubularins) regulating conversion of PtdIns(3,5)P2 into PtdIns3P, and corneal fleck dystrophy caused by mutations in the PIKfyve kinase that converts PtdIns3P into PtdIns(3,5)P2 . Further evidence for the importance of PtdIns3P levels comes from studies highlighting the role of PIKfyve in Salmonella uptake into macropinosomes and progression of the pathogen through the endosomal system [161]. Although the underlying mechanisms that lead to these different pathologies are still not clear, the PX domain proteins are extremely strong candidates for molecules whose function will be perturbed by defects in PtdIns3P metabolism. PX proteins in cancer Many PX proteins have been linked to cancers on the basis of differential expression and other criteria (Table 2). Both the unique PX protein SNX15 and the PX-PH-PLD family c The Authors Journal compilation c 2012 Biochemical Society [128] [247] [248] member PLD2 are up-regulated in metastatic mammary tumour cells [162]. SH3PXD2A (SH3 and PX domain protein 2A) is essential for the formation of podosomes, actin-rich structures required for invasion of many types of cancers. It has been shown that SH3PXD2A is required for podosome formation and invasion of several cancer cell lines in a PX domain-dependent manner [163], and that inhibiting its expression correlates with reduced tumour growth in mouse transplantation models [164]. It is hypothesized that SH3PXD2A may act to regulate the recruitment of essential factors, such as N-WASP, to podosomes via its SH3 domains. Single-nucleotide polymorphisms in the gene encoding PI3K (phosphoinositide 3-kinase)-C2β (PI3KC2β) have been associated with increased risk of prostate cancer, suggested to be due to either enhanced cell migration or increased insulin signalling [165]. There is strong evidence for a role for SNX1 as a tumour suppressor. SNX1 expression assessed by both immunohistochemistry and microarray data indicates that SNX1 is reduced in colon carcinoma cells, and, furthermore, inhibition of SNX1 expression by shRNA (small hairpin RNA) leads to increased cell proliferation and decreased apoptosis [166]. The authors provide evidence that SNX1 depletion leads to increased EGFR phosphorylation and subsequent ERK signalling from endosomes, and that this may be important for tumour progression. Further support for a role for SNX1 depletion in colorectal tumour progression comes from a study showing that a microRNA up-regulated in colon carcinomas, miR-95, specifically targets SNX1 [167]. This model fits with the original identification of the SNX1 protein as a molecule that can enhance the degradation of the EGFR [41], but it does not fit with subsequent knockdown data indicating that depletion of SNX1 by siRNA has little effect on EGFR degradation [46] and therefore will need further investigation before it can be confirmed. Two other families of PX proteins have been implicated in cancer due to their function as cell signalling modulators: the serine/threonine kinase SGK3 (serum- and glucocorticoidinduced protein kinase 3) and the phospholipases PLD1 and PLD2. Vasudevan et al. [168] established that oncogenic mutations in the PI3K subunit PIK3CA can result in cancers that are either dependent or independent of up-regulation in Akt signalling. In Akt-independent cancer cells, the key factor promoting cell survival is increased activation of SGK3 by the concerted signalling of PI3K and PDK1 (phosphoinositidedependent kinase 1). Furthermore, studies have found that SGK3 is up-regulated in ER (oestrogen receptor)-positive breast tumours, is required for oestrogen-mediated cell survival and can protect cells against apoptosis induced by chemotherapeutics [169]. Few substrates for SGK3 have been identified; however, SGK3 can attenuate the endosomal activity of ubiquitin ligases and thus impair lysosomal degradation of signalling molecules, The PX protein family such as the chemokine receptor CXCR4 involved in breast cancer [170]. Enhanced signalling due to inhibited receptor degradation is one possible mechanism by which SGK3 could promote cell survival. Perhaps the best-characterized PX proteins in terms of oncogenic potential are PLD1 and PLD2. There is not scope in the present paper to review all of the evidence for the role of PLD isoforms in cancer, so instead we refer to several previous reviews [171–173]. Very briefly, it appears that their importance in cancer is due to their role in the synthesis of PA (phosphatidic acid) from PC (phosphatidylcholine) and the effect of PA on downstream signalling, anti-apoptotic and membrane-remodelling processes. Importantly, PLD isoforms are now validated targets for the development of small-molecule inhibitors as chemotherapeutics [173–175]. PX proteins in inflammation An interesting example of how endosomal trafficking of receptors by PX proteins can affect disease-related processes is how the PX proteins SNX17 and SNX20 play complementary roles in haemopoietic cells during the inflammatory response. The importance of platelets in inflammation lies in their ability to co-operate with endothelial cells to co-ordinate the adherence and extravasation of leucocytes during the inflammatory response [176]. The release of chemokines and cytokines by macrophages within injured tissue initiates a sequence of events whereby leucocytes are attached to the endothelium, first by low-affinity interaction of adhesion molecules called selectins on endothelial and platelet cells with glycoproteins on the leucocytes (‘leucocyte rolling’), and secondly by high-affinity attachment via cellsurface integrins that are able to resist the fluid shear forces within the blood vessel. Similar mechanisms are also known to contribute to coagulation and haematogenous tumour metastasis [177]. The reciprocal arrangement of adhesion molecules on leucocytes, platelets and endothelial cells is carefully controlled by regulating their subcellular localization. PX domain proteins have been linked to the trafficking and cell-surface presentation of two key molecules in this process: P-selectin on endothelial and platelet cells by SNX17 [75,85,86], and PSGL-1 on leucocytes by SNX20 [35] (Supplementary Figure S3 at http://www.BiochemJ.org/bj/441/bj4410039add.htm). P-selectin is stored within specific intracellular compartments from which it is released to the cell surface upon stimulation: α-granules in platelets and Weibel-Palade bodies in endothelial cells. It also undergoes endocytosis, after which it is either recycled through the endocytic system or degraded within lysosomes [177,178]. SNX17 was identified as a P-selectin-interacting protein [75,85], and we have also observed binding of SNX27, suggesting that this may be a common function of the PX-FERM subfamily (R. Ghai and B.M. Collins, unpublished work). Overexpression of SNX17 was found to enhance the internalization of P-selectin from the cell surface, and to retard the degradation of P-selectin in lysosomes by restricting its transport into late endosomes/multivesicular bodies and causing an accumulation in SNX17-positive endosomes [75,86]. Within the opposing leucocyte cells, SNX20 was identified as a binder of the cytoplasmic tail of PSGL-1 [35]. Overexpression of SNX20 caused a significant redistribution of PSGL-1 from the cell surface into endosomes; however, mice deficient in SNX20 appeared to be normal and healthy and their neutrophils displayed no significant defects in PSGL-1-mediated cell adhesion or signalling, possibly due to compensation by the highly homologous SNX21. Further studies will be required to determine if this is true and whether the SNX20/SNX21 proteins control trafficking of other transmembrane cargoes. 51 PX proteins in Alzheimer’s disease AD is caused, at least in part, by the accumulation of the toxic amyloid peptide Aβ [179]. Aβ is derived from the sequential cleavage of the APP by transmembrane enzymes called secretases, first by β-secretase to release a soluble ectodomain fragment, and subsequently by γ -secretase within the transmembrane domain to form the 40–42-residue Aβ peptide. This degradation is thought to occur primarily within intracellular organelles, whereas a non-amyloidogenic pathway involving initial cleavage by the α-secretase is thought to occur primarily at the cell surface [180]. For detailed reviews of APP trafficking and processing see [181–185]. The production of Aβ is therefore dependent on a delicate balance between the trafficking of APP, the trafficking of secretase enzymes and the degree to which the molecules encounter each other within the organelles of the neuronal cell. It is increasingly apparent that many PX proteins and their partners such as the retromer complex play important roles in APP and secretase trafficking, regulating the subsequent accumulation of toxic Aβ peptide (Figure 5). This can occur in a number of ways, as described below. (i) Trafficking of β-secretase and SorLA, which is a co-receptor involved in the retrograde trafficking of APP from endosomes to the TGN, is co-ordinated by the retromer retrograde trafficking complex [186–189]. Removal of APP from the endosomal system is expected to result in less Aβ production, due to avoidance of β- and γ -secretases, which function predominantly within these organelles. SNX1, SNX2, SNX5 and SNX6 are key regulators of retromer-mediated trafficking, although a direct role for these PX-BAR proteins in APP trafficking and degradation is yet to be demonstrated. (ii) A recent study has shown an important role for SNX6 in regulating BACE1 (β-secretase 1; also called β-site APPcleaving enzyme 1) trafficking and APP degradation [190]. It was found that SNX6 forms a complex with β-secretase, and that SNX6 suppression leads to modulation of β-secretase retrograde trafficking, increased levels of β-secretase and subsequent increased production of Aβ peptide. In contrast with the normal function of the retromer complex and SNX1, this study suggests that SNX6 inhibits the retrograde traffic of BACE1 from endosomes to the Golgi. It is proposed that SNX6 may either antagonize SNX1 function in BACE1 transport or form retromer complexes with differential cargo selectivity. Indirect support for this latter hypothesis is supplied by data showing that alternative retromer-associated PX-BAR proteins can mediate trafficking of different cargo proteins [58]. (iii) SNX17 is highly expressed in neurons [83,87] and is implicated directly in the recycling of APP from endosomes to the cell surface, as it binds to the APP NPxY sorting motif and inhibition by siRNA results in an increased level of APP breakdown and Aβ peptide production within the endosomal compartment [11,87]. In addition, one of the most intriguing aspects of SNX17-receptor trafficking is the relationship between many of the identified SNX17 cargos. Many studies over the last few years have highlighted the complexity of APP processing, and in particular have shown a critical role for ApoE (apolipoprotein E) and its receptors from the LDLR family in APP processing and Aβ clearance in AD (reviewed in [181–183]). SNX17 regulates the endosome-to-cell surface recycling of many receptors of the LDLR family, including LRP1, which facilitates the transport of the APP protein through an interaction mediated by FE65 [81–85], and others implicated directly in both the clearance and generation of Aβ plaques via their role as receptors for the Aβ chaperone ApoE [181–183]. Therefore SNX17 appears to be functioning at a nexus of trafficking pathways critical for APP c The Authors Journal compilation c 2012 Biochemical Society 52 Figure 5 R. D. Teasdale and B. M. Collins Cartoon outlining potential roles of PX proteins in the trafficking and processing of APP to neurotoxic Aβ implicated in AD For reviews of APP trafficking and processing, see [181–185]. See the text for a detailed discussion. processing, not only controlling APP directly, but also affecting other receptors with important roles in the process of Aβ peptide production and clearance. It is possible that trafficking of APP and LDLR family proteins may be influenced by the other PXFERM molecules SNX27 and SNX31, but this remains to be confirmed. (iv) SNX33 and SNX9 have been shown to affect the uptake of APP from the cell surface, although the mechanism is poorly understood [66]. Increased expression of SNX33 or SNX9 was shown to significantly increase the level of soluble APP released into the extracellular space by α-secretase (sAPPα). A similar effect was seen for dominant-negative constructs of the dynamin GTPase required for scission of endocytic vesicles. SNX33 was shown to bind dynamin via its SH3 domain and its expression was able to inhibit the endocytosis of APP. Therefore the SH3-PX-BAR proteins in this system are able to affect the balance of α- and β-secretase cleavage and the level of Aβ production. The authors suggest that SNX33 and SNX9 may be working to inhibit dynamin during endocytosis. Given the role of SNX9 in promoting clathrin-dependent and -independent endocytosis, this may at first appear counterintuitive [17,21,47,191,192]. However, other studies have also shown that either overexpression or knockdown of SNX9 can effectively perturb clathrin-mediated endocytosis, indicating that altered SNX9 protein levels may impair the homoeostasis of proteininteraction networks required for endocytic vesicle formation [191–193]. One possible explanation is that overexpression of SNX33 or SNX9 results in competition for APP-specific adaptors such as Dab2 [87,194] for binding to the AP2 complex, clathrin c The Authors Journal compilation c 2012 Biochemical Society and dynamin during endocytic vesicle formation, and therefore acts to slow the dynamics of APP endocytosis. More work will be required to dissect the mechanisms by which SH3-PX-BAR proteins affect APP uptake. (v) SH3PXD2A was shown to play a role in mediating Aβinduced neurotoxicity through its interaction with members of the ADAM (a disintegrin and metalloproteinase) family of enzymes (for reviews of ADAMs, see [195,196]). SH3PXD2A can bind to (at least) ADAM12, ADAM15 and ADAM19 through association of its fifth SH3 domain with polyproline sequences in the cytosplasmic tails of the transmembrane proteases [197,198]. Exposure of neurons to cytotoxic Aβ peptide results in tyrosine phosphorylation of SH3PXD2A and relocalization of the protein, and SH3PXD2A is then able to stimulate the metalloproteinase activity of ADAM12 [199]. Treating cells with toxic Aβ peptide results in increased ADAM12 proteolytic activity, whereas expression of protease-deficient ADAM12 or an ADAM12binding mutant of SH3PXD2A blocks Aβ-induced cell death. In addition, SNPs (single nucleotide polymorphisms) in ADAM12 and SH3PXD2A may confer susceptibility to late-onset AD [200]. Although the mechanism is very poorly understood, the hypothesis is that neuronal cell death induced by toxic Aβ peptides is at least partly mediated by ADAM12 and SH3PXD2A. These proteins therefore may be potential targets for therapeutic applications. (vi) Another family of PX proteins implicated as negative regulators of Aβ production in AD are the PX-PH-PLD proteins [201]. PLD1 is significantly up-regulated in brains of AD patients, in particular within mitochondrial fractions, and is able to interact The PX protein family with the cytoplasmic tail of APP via its PH domain [202,203]. There is now strong evidence that PLD1 can influence the trafficking of both APP and the PS1 (presenilin 1) subunit of γ -secretase (and, through PS1, presumably other γ -secretase subunits), in particular promoting the formation of TGN-derived transport vesicles destined for the cell surface [204–206]. It is thought that PLD1 enhances egress of APP from the TGN and internal endosomes where Aβ production predominantly occurs, thereby reducing the level of toxic Aβ production. In addition to a role in APP trafficking, PLD1 was also found to inhibit Aβ production directly, by interfering with the assembly of γ -secretase via association with PS1 [204]. Adding to the complexity of PLD function in AD, other studies indicate that PLD activity is increased in neurons exposed to Aβ, that PLD2 is critical for mediating Aβ neuronal toxicity and that PLD2 ablation in amyloidogenic transgenic mouse models improves memory deficits and synaptic function, suggesting that PLD activity may be a valid target for therapeutic action [207]. It is becoming increasingly apparent that intracellular trafficking is central to the process of APP homoeostasis and Aβ manufacture [180,208]. Taken together, the evidence is overwhelming that PX proteins regulate various APP and secretase trafficking pathways and modulate production and toxicity of the Aβ peptide, so determining the precise roles of the PX proteins and the molecular mechanisms that underpin their function in Aβ regulation represents an important area of investigation. PX proteins in pathogen invasion Another disease process that has gained recent attention is that of pathogen invasion via corruption of the endocytic system (reviewed in [209]). The invasion of Salmonella enterica via coercion of normal cellular macropinocytic processes [161,210,211] has been found to be critically dependent on the action of PX proteins. Bujny et al. [212] found that SNX1 undergoes rapid translocation to sites of bacterial entry and SCVs (Salmonella-containing vacuoles), resulting in the formation of extensive long-range membrane tubules that are thought to mediate membrane contraction during SCV maturation. Critically, suppression of SNX1 was found to halt the progression of SCVs into the cell. Recently, Braun et al. [213], demonstrated that the PX-only protein SNX3 is also important for the process. Similarly to SNX1, SNX3 is recruited to SCVs and to the SCV membrane tubules, and its depletion leads to impaired SCV maturation. However it was found that SNX3 recruitment occurs after SNX1, and in fact SNX3 depends on SNX1 (and SNX2) for localization to these tubules. From a mechanistic standpoint, this correlates well with the known role of SNX1 and SNX2 as proteins regulating membrane tubulation. The generation of an SCV is dependent on bacterially secreted enzymes that promote the generation of PtdIns3P on its limiting membrane during the early stages of formation. Given the central role of PtdIns3P in early stages of Salmonella invasion, it is very likely that other PX proteins will also be important for the process. Indeed, SNX15 was recently found to be recruited to early SCVs (but not to tubules) [213], and given the role of PX-BAR proteins other than SNX1 in macropinocytosis [24,214], these are also likely to be involved in pathogen invasion. SNX1, SNX6, SNX9 and SNX33 have all been identified as being involved in the internalization of apoptotic cells via phagocytosis and the subsequent transport into a degradative organelle [215–217]. However, their role in phagocytosis of pathogens remains uncharacterized. Other PX family members have central roles in host defence against pathogens, with p47phox , p40phox and NOXO1 (NADPH oxidase 53 organizer 1) associated with the NADPH oxidase complex, which is responsible for the formation of superoxide anions [218,219]. Following phagocytosis of pathogens, the NADPH oxidase complex is activated, resulting in an oxidative burst that can kill the pathogen directly, or indirectly by activating other components of the innate immune system. In addition to a role in bacterial invasion, endocytosis and trafficking through the endosomal system is also critical for the uptake of bacterial toxins and the processing and assembly of viruses [209]. STx (Shiga toxin) is secreted by Shigella dysenteriae and is endocytosed by target cells by clathrinmediated uptake, bypasses the degradative pathways and is eventually translocated into the cytosol from the endoplasmic reticulum [220]. SNX1 has been found to be important for the efficient passage of STx [50,57,58,221]. SNX2 has also been implicated [50], although other studies suggest that SNX2 does not play an important role in the process [57,58]. In contrast, depletion of another PX-BAR protein SNX8 increases the rate of endosome–Golgi transport of STx, suggesting an antagonistic role in STx passage through the endosomal compartment [222]. Finally, SNX5 has been implicated in the uptake via macropinocytosis of Ebola virus [223], and the protein SNX16 has been found to be important for infection by VSV (vesicular stomatitis virus) [128]. For infection, VSV and other enveloped viruses must be endocytosed and transported to late endosomes where the acidic pH of the lumen triggers fusion of the viral envelope with the endosomal membrane and release of the nucleocapsid into the cytosol for viral replication. SNX16 was found to localize to these late endosomes and its overexpression significantly inhibited the export of the nucleocapsid. It remains to be determined, however, how SNX16 acts to regulate this process. CONCLUDING REMARKS In the last few years there has been major progress in understanding the functions of PX proteins. Of course, the diversity of these proteins makes it impossible to present a single unified picture of how they work. It is clear, however, that the coupling of the membrane-localizing PX domain with different functional modules is a mechanism by which eukaryotic cells have constructed tools to control a multitude of protein binding, membrane remodelling, signalling, motor and enzymatic functions localized to specific regions of the secretory and endocytic system. Many questions remain regarding the functional, structural and pathological roles of the PX proteins. From a cellular perspective, it will be important to develop quantitative tools for analysing the localization, interactions and, most importantly, the spatiotemporal regulation of PX proteins by different cellular signals. Structurally, although we now have a firm grasp of how the PX domain associates with PtdIns3P, there is a severe paucity of information regarding how the PX domain regulates protein– protein interactions. There is also a significant lack of knowledge regarding the structures of many of the PX-associated protein domains and the overall tertiary structures of the full-length molecules. Most importantly, it will be essential to combine these cellular and structural insights with broader systems approaches to develop an understanding of how the vast networks of interactions controlled by the PX proteins regulate cellular fate. Finally, it is imperative to determine how PX proteins contribute to different disease processes, such as pathogen invasion and amyloid production, both in order to determine whether they might be suitable therapeutic targets and also to gain insights into how such interventions might be achieved. c The Authors Journal compilation c 2012 Biochemical Society 54 R. D. Teasdale and B. M. Collins FUNDING Research in the laboratories of B.M.C. and R.D.T. is supported by funds from the Australian Research Council (ARC) and the National Health and Medical Research Council (NHMRC). B.M.C. is an ARC Future Fellow [grant number FT100100027]. R.D.T. is an NHMRC Senior Research Fellow [grant number 511042]. REFERENCES 1 Scita, G. and Di Fiore, P. P. (2010) The endocytic matrix. Nature 463, 464–473 2 Sorkin, A. and von Zastrow, M. (2009) Endocytosis and signalling: intertwining molecular networks. Nat. Rev. Mol. Cell Biol. 10, 609–622 3 Seet, L. F. and Hong, W. (2006) The Phox (PX) domain proteins and membrane traffic. Biochim. Biophys. Acta 1761, 878–896 4 Karathanassis, D., Stahelin, R. V., Bravo, J., Perisic, O., Pacold, C. M., Cho, W. and Williams, R. L. (2002) Binding of the PX domain of p47phox to phosphatidylinositol 3,4-bisphosphate and phosphatidic acid is masked by an intramolecular interaction. EMBO J. 21, 5057–5068 5 Stahelin, R. V., Ananthanarayanan, B., Blatner, N. R., Singh, S., Bruzik, K. S., Murray, D. and Cho, W. (2004) Mechanism of membrane binding of the phospholipase D1 PX domain. J. Biol. Chem. 279, 54918–54926 6 Stahelin, R. V., Burian, A., Bruzik, K. S., Murray, D. and Cho, W. (2003) Membrane binding mechanisms of the PX domains of NADPH oxidase p40phox and p47phox . J. Biol. Chem. 278, 14469–14479 7 Stahelin, R. V., Karathanassis, D., Bruzik, K. S., Waterfield, M. D., Bravo, J., Williams, R. L. and Cho, W. (2006) Structural and membrane binding analysis of the Phox homology domain of phosphoinositide 3-kinase-C2α. J. Biol. Chem. 282, 39396–39406 8 Stahelin, R. V., Karathanassis, D., Murray, D., Williams, R. L. and Cho, W. (2007) Structural and membrane binding analysis of the Phox homology domain of Bem1p: basis of phosphatidylinositol 4-phosphate specificity. J. Biol. Chem. 282, 25737–25747 9 Yu, J. W. and Lemmon, M. A. (2001) All phox homology (PX) domains from Saccharomyces cerevisiae specifically recognize phosphatidylinositol 3-phosphate. J. Biol. Chem. 276, 44179–44184 10 Falasca, M. and Maffucci, T. (2009) Rethinking phosphatidylinositol 3-monophosphate. Biochim. Biophys. Acta 1793, 1795–1803 11 Ghai, R., Mobli, M., Norwood, S. J., Bugarcic, A., Teasdale, R. D., King, G. F. and Collins, B. M. (2011) Phox homology band 4.1/ezrin/radixin/moesin-like proteins function as molecular scaffolds that interact with cargo receptors and Ras GTPases. Proc. Natl. Acad. Sci. U.S.A. 108, 7763–7768 12 Song, J., Zhao, K. Q., Newman, C. L., Vinarov, D. A. and Markley, J. L. (2007) Solution structure of human sorting nexin 22. Protein Sci. 16, 807–814 13 Carlton, J. G. and Cullen, P. J. (2005) Coincidence detection in phosphoinositide signaling. Trends Cell Biol. 15, 540–547 14 Lemmon, M. A. (2008) Membrane recognition by phospholipid-binding domains. Nat. Rev. Mol. Cell Biol. 9, 99–111 15 Narayan, K. and Lemmon, M. A. (2006) Determining selectivity of phosphoinositidebinding domains. Methods 39, 122–133 16 Badour, K., McGavin, M. K., Zhang, J., Freeman, S., Vieira, C., Filipp, D., Julius, M., Mills, G. B. and Siminovitch, K. A. (2007) Interaction of the Wiskott–Aldrich syndrome protein with sorting nexin 9 is required for CD28 endocytosis and cosignaling in T cells. Proc. Natl. Acad. Sci. U.S.A. 104, 1593–1598 17 Lundmark, R. and Carlsson, S. R. (2003) Sorting nexin 9 participates in clathrinmediated endocytosis through interactions with the core components. J. Biol. Chem. 278, 46772–46781 18 Pylypenko, O., Lundmark, R., Rasmuson, E., Carlsson, S. R. and Rak, A. (2007) The PX-BAR membrane-remodeling unit of sorting nexin 9. EMBO J. 26, 4788–4800 19 Shin, N., Ahn, N., Chang-Ileto, B., Park, J., Takei, K., Ahn, S. G., Kim, S. A., Di Paolo, G. and Chang, S. (2008) SNX9 regulates tubular invagination of the plasma membrane through interaction with actin cytoskeleton and dynamin 2. J. Cell Sci. 121, 1252–1263 20 Yarar, D., Surka, M. C., Leonard, M. C. and Schmid, S. L. (2008) SNX9 activities are regulated by multiple phosphoinositides through both PX and BAR domains. Traffic 9, 133–146 21 Yarar, D., Waterman-Storer, C. M. and Schmid, S. L. (2007) SNX9 couples actin assembly to phosphoinositide signals and is required for membrane remodeling during endocytosis. Dev. Cell 13, 43–56 22 Koharudin, L. M., Furey, W., Liu, H., Liu, Y. J. and Gronenborn, A. M. (2009) The phox domain of sorting nexin 5 lacks phosphatidylinositol 3-phosphate (PtdIns(3)P ) specificity and preferentially binds to phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P 2). J. Biol. Chem. 284, 23697–23707 23 Liu, H., Liu, Z. Q., Chen, C. X., Magill, S., Jiang, Y. and Liu, Y. J. (2006) Inhibitory regulation of EGF receptor degradation by sorting nexin 5. Biochem. Biophys. Res. Commun. 342, 537–546 c The Authors Journal compilation c 2012 Biochemical Society 24 Kerr, M. C., Lindsay, M. R., Luetterforst, R., Hamilton, N., Simpson, F., Parton, R. G., Gleeson, P. A. and Teasdale, R. D. (2006) Visualisation of macropinosome maturation by the recruitment of sorting nexins. J. Cell Sci. 119, 3967–3980 25 Hiroaki, H., Ago, T., Ito, T., Sumimoto, H. and Kohda, D. (2001) Solution structure of the PX domain, a target of the SH3 domain. Nat. Struct. Biol. 8, 526–530 26 Sumimoto, H., Kage, Y., Nunoi, H., Sasaki, H., Nose, T., Fukumaki, Y., Ohno, M., Minakami, S. and Takeshige, K. (1994) Role of Src homology 3 domains in assembly and activation of the phagocyte NADPH oxidase. Proc. Natl. Acad. Sci. U.S.A. 91, 5345–5349 27 Jang, I. H., Lee, S., Park, J. B., Kim, J. H., Lee, C. S., Hur, E. M., Kim, I. S., Kim, K. T., Yagisawa, H., Suh, P. G. and Ryu, S. H. (2003) The direct interaction of phospholipase C-γ 1 with phospholipase D2 is important for epidermal growth factor signaling. J. Biol. Chem. 278, 18184–18190 28 Prehoda, K. E. and Lim, W. A. (2001) The double life of PX domains. Nat. Struct. Biol. 8, 570–572 29 Lee, C. S., Kim, I. S., Park, J. B., Lee, M. N., Lee, H. Y., Suh, P. G. and Ryu, S. H. (2006) The phox homology domain of phospholipase D activates dynamin GTPase activity and accelerates EGFR endocytosis. Nat. Cell Biol. 8, 477–484 30 Lee, H. Y., Park, J. B., Jang, I. H., Chae, Y. C., Kim, J. H., Kim, I. S., Suh, P. G. and Ryu, S. H. (2004) Munc-18–1 inhibits phospholipase D activity by direct interaction in an epidermal growth factor-reversible manner. J. Biol. Chem. 279, 16339–16348 31 Parks, W. T., Frank, D. B., Huff, C., Renfrew Haft, C., Martin, J., Meng, X., de Caestecker, M. P., McNally, J. G., Reddi, A., Taylor, S. I. et al. (2001) Sorting nexin 6, a novel SNX, interacts with the transforming growth factor-β family of receptor serine-threonine kinases. J. Biol. Chem. 276, 19332–19339 32 Abdul-Ghani, M., Hartman, K. L. and Ngsee, J. K. (2005) Abstrakt interacts with and regulates the expression of sorting nexin-2. J. Cell. Physiol. 204, 210–218 33 Skanland, S. S., Walchli, S., Brech, A. and Sandvig, K. (2009) SNX4 in complex with clathrin and dynein: implications for endosome movement. PLoS ONE 4, e5935 34 Ishibashi, Y., Maita, H., Yano, M., Koike, N., Tamai, K., Ariga, H. and Iguchi-Ariga, S. M. (2001) Pim-1 translocates sorting nexin 6/TRAF4-associated factor 2 from cytoplasm to nucleus. FEBS Lett. 506, 33–38 35 Schaff, U. Y., Shih, H. H., Lorenz, M., Sako, D., Kriz, R., Milarski, K., Bates, B., Tchernychev, B., Shaw, G. D. and Simon, S. I. (2008) SLIC-1/sorting nexin 20: a novel sorting nexin that directs subcellular distribution of PSGL-1. Eur. J. Immunol. 38, 550–564 36 DiNitto, J. P. and Lambright, D. G. (2006) Membrane and juxtamembrane targeting by PH and PTB domains. Biochim. Biophys. Acta 1761, 850–867 37 Cullen, P. J. (2008) Endosomal sorting and signalling: an emerging role for sorting nexins. Nat. Rev. Mol. Cell Biol. 9, 574–582 38 Carlton, J., Bujny, M., Rutherford, A. and Cullen, P. (2005) Sorting nexins – unifying trends and new perspectives. Traffic 6, 75–82 39 van Weering, J. R., Verkade, P. and Cullen, P. J. (2010) SNX-BAR proteins in phosphoinositide-mediated, tubular-based endosomal sorting. Semin. Cell Dev. Biol. 21, 371–380 40 Attar, N. and Cullen, P. J. (2010) The retromer complex. Adv. Enzyme Regul. 50, 216–236 41 Kurten, R. C., Cadena, D. L. and Gill, G. N. (1996) Enhanced degradation of EGF receptors by a sorting nexin, SNX1. Science 272, 1008–1010 42 Teasdale, R. D., Loci, D., Houghton, F., Karlsson, L. and Gleeson, P. A. (2001) A large family of endosome-localized proteins related to sorting nexin 1. Biochem. J. 358, 7–16 43 Worby, C. A. and Dixon, J. E. (2002) Sorting out the cellular functions of sorting nexins. Nat. Rev. Mol. Cell Biol. 3, 919–931 44 Bhatia, V. K., Madsen, K. L., Bolinger, P. Y., Kunding, A., Hedegard, P., Gether, U. and Stamou, D. (2009) Amphipathic motifs in BAR domains are essential for membrane curvature sensing. EMBO J. 28, 3303–3314 45 Gallop, J. L. and McMahon, H. T. (2005) BAR domains and membrane curvature: bringing your curves to the BAR. Biochem. Soc. Symp. 72, 223–231 46 Carlton, J., Bujny, M., Peter, B. J., Oorschot, V. M., Rutherford, A., Mellor, H., Klumperman, J., McMahon, H. T. and Cullen, P. J. (2004) Sorting nexin-1 mediates tubular endosome-to-TGN transport through coincidence sensing of high-curvature membranes and 3-phosphoinositides. Curr. Biol. 14, 1791–1800 47 Lundmark, R. and Carlsson, S. R. (2009) SNX9 – a prelude to vesicle release. J Cell Sci. 122, 5–11 48 Bonifacino, J. S. and Hurley, J. H. (2008) Retromer. Curr. Opin. Cell Biol. 20, 427–436 49 Collins, B. M. (2008) The structure and function of the retromer protein complex. Traffic. 9, 1811–1822 50 Utskarpen, A., Slagsvold, H. H., Dyve, A. B., Skanland, S. S. and Sandvig, K. (2007) SNX1 and SNX2 mediate retrograde transport of Shiga toxin. Biochem. Biophys. Res. Commun. 358, 566–570 51 Rojas, R., Kametaka, S., Haft, C. R. and Bonifacino, J. S. (2007) Interchangeable but essential functions of SNX1 and SNX2 in the association of retromer with endosomes and the trafficking of mannose 6-phosphate receptors. Mol. Cell. Biol. 27, 1112–1124 The PX protein family 52 Gullapalli, A., Garrett, T. A., Paing, M. M., Griffin, C. T., Yang, Y. and Trejo, J. (2004) A role for sorting nexin 2 in epidermal growth factor receptor down-regulation: evidence for distinct functions of sorting nexin 1 and 2 in protein trafficking. Mol. Biol. Cell 15, 2143–2155 53 Haft, C. R., de la Luz Sierra, M., Bafford, R., Lesniak, M. A., Barr, V. A. and Taylor, S. I. (2000) Human orthologs of yeast vacuolar protein sorting proteins Vps26, 29, and 35: assembly into multimeric complexes. Mol. Biol. Cell. 11, 4105–4116 54 Haft, C. R., de la Luz Sierra, M., Barr, V. A., Haft, D. H. and Taylor, S. I. (1998) Identification of a family of sorting nexin molecules and characterization of their association with receptors. Mol. Cell. Biol 18, 7278–7287 55 Wang, Y., Zhou, Y., Szabo, K., Haft, C. R. and Trejo, J. (2002) Down-regulation of protease-activated receptor-1 is regulated by sorting nexin 1. Mol. Biol. Cell 13, 1965–1976 56 Wassmer, T., Attar, N., Harterink, M., van Weering, J. R., Traer, C. J., Oakley, J., Goud, B., Stephens, D. J., Verkade, P., Korswagen, H. C. and Cullen, P. J. (2009) The retromer coat complex coordinates endosomal sorting and dynein-mediated transport, with carrier recognition by the trans -Golgi network. Dev. Cell 17, 110–122 57 Bujny, M. V., Popoff, V., Johannes, L. and Cullen, P. J. (2007) The retromer component sorting nexin-1 is required for efficient retrograde transport of Shiga toxin from early endosome to the trans Golgi network. J. Cell Sci. 120, 2010–2021 58 Lieu, Z. Z. and Gleeson, P. A. (2010) Identification of different itineraries and retromer components for endosome-to-Golgi transport of TGN38 and Shiga toxin. Eur. J. Cell Biol. 89, 379–393 59 Gullapalli, A., Wolfe, B. L., Griffin, C. T., Magnuson, T. and Trejo, J. (2006) An essential role for SNX1 in lysosomal sorting of protease-activated receptor-1: evidence for retromer-, Hrs-, and Tsg101-independent functions of sorting nexins. Mol. Biol. Cell 17, 1228–1238 60 Schwarz, D. G., Griffin, C. T., Schneider, E. A., Yee, D. and Magnuson, T. (2002) Genetic analysis of sorting nexins 1 and 2 reveals a redundant and essential function in mice. Mol. Biol. Cell 13, 3588–3600 61 Wassmer, T., Attar, N., Bujny, M. V., Oakley, J., Traer, C. J. and Cullen, P. J. (2007) A loss-of-function screen reveals SNX5 and SNX6 as potential components of the mammalian retromer. J. Cell Sci 120, 45–54 62 Merino-Trigo, A., Kerr, M. C., Houghton, F., Lindberg, A., Mitchell, C., Teasdale, R. D. and Gleeson, P. A. (2004) Sorting nexin 5 is localized to a subdomain of the early endosomes and is recruited to the plasma membrane following EGF stimulation. J. Cell Sci. 117, 6413–6424 63 Haberg, K., Lundmark, R. and Carlsson, S. R. (2008) SNX18 is an SNX9 paralog that acts as a membrane tubulator in AP-1-positive endosomal trafficking. J. Cell Sci. 121, 1495–1505 64 Zhang, J., Zhang, X., Guo, Y., Xu, L. and Pei, D. (2009) Sorting nexin 33 induces mammalian cell micronucleated phenotype and actin polymerization by interacting with Wiskott–Aldrich syndrome protein. J. Biol. Chem. 284, 21659–21669 65 Park, S., Kim, Y., Lee, S., Park, P., Park, Z., Sun, W., Kim, H. and Chang, S. (2010) SNX18 shares a redundant role with SNX9 and modulates endoctyic trafficking at the plasma membrane. J. Cell Sci. 123, 1742–1750 66 Schobel, S., Neumann, S., Hertweck, M., Dislich, B., Kuhn, P. H., Kremmer, E., Seed, B., Baumeister, R., Haass, C. and Lichtenthaler, S. F. (2008) A novel sorting nexin modulates endocytic trafficking and α-secretase cleavage of the amyloid precursor protein. J. Biol. Chem. 283, 14257–14268 67 Wang, Q., Kaan, H. Y., Hooda, R. N., Goh, S. L. and Sondermann, H. (2008) Structure and plasticity of Endophilin and Sorting Nexin 9. Structure 16, 1574–1587 68 Frost, A., Perera, R., Roux, A., Spasov, K., Destaing, O., Egelman, E. H., De Camilli, P. and Unger, V. M. (2008) Structural basis of membrane invagination by F-BAR domains. Cell 132, 807–817 69 Chin, L. S., Raynor, M. C., Wei, X., Chen, H. Q. and Li, L. (2001) Hrs interacts with sorting nexin 1 and regulates degradation of epidermal growth factor receptor. J. Biol. Chem. 276, 7069–7078 70 Heydorn, A., Sondergaard, B. P., Ersboll, B., Holst, B., Nielsen, F. C., Haft, C. R., Whistler, J. and Schwartz, T. W. (2004) A library of 7TM receptor C-terminal tails. Interactions with the proposed post-endocytic sorting proteins ERM-binding phosphoprotein 50 (EBP50), N -ethylmaleimide-sensitive factor (NSF), sorting nexin 1 (SNX1), and G protein-coupled receptor-associated sorting protein (GASP). J. Biol. Chem. 279, 54291–54303 71 Swarbrick, J. D., Shaw, D. J., Chhabra, S., Ghai, R., Valkov, E., Norwood, S. J., Seaman, M. N. and Collins, B. M. (2011) VPS29 is not an active metallo-phosphatase but is a rigid scaffold required for retromer interaction with accessory proteins. PLoS ONE 6, e20420 72 Traer, C. J., Rutherford, A. C., Palmer, K. J., Wassmer, T., Oakley, J., Attar, N., Carlton, J. G., Kremerskothen, J., Stephens, D. J. and Cullen, P. J. (2007) SNX4 coordinates endosomal sorting of TfnR with dynein-mediated transport into the endocytic recycling compartment. Nat. Cell Biol. 9, 1370–1380 55 73 Chishti, A. H., Kim, A. C., Marfatia, S. M., Lutchman, M., Hanspal, M., Jindal, H., Liu, S. C., Low, P. S., Rouleau, G. A., Mohandas, N. et al. (1998) The FERM domain: a unique module involved in the linkage of cytoplasmic proteins to the membrane. Trends Biochem. Sci. 23, 281–282 74 Czubayko, M., Knauth, P., Schluter, T., Florian, V. and Bohnensack, R. (2006) Sorting nexin 17, a non-self-assembling and a PtdIns(3)P high class affinity protein, interacts with the cerebral cavernous malformation related protein KRIT1. Biochem. Biophys. Res. Commun. 345, 1264–1272 75 Knauth, P., Schluter, T., Czubayko, M., Kirsch, C., Florian, V., Schreckenberger, S., Hahn, H. and Bohnensack, R. (2005) Functions of sorting nexin 17 domains and recognition motif for P-selectin trafficking. J. Mol. Biol. 347, 813–825 76 van Kerkhof, P., Lee, J., McCormick, L., Tetrault, E., Lu, W., Schoenfish, M., Oorschot, V., Strous, G. J., Klumperman, J. and Bu, G. (2005) Sorting nexin 17 facilitates LRP recycling in the early endosome. EMBO J. 24, 2851–2861 77 Joubert, L., Hanson, B., Barthet, G., Sebben, M., Claeysen, S., Hong, W., Marin, P., Dumuis, A. and Bockaert, J. (2004) New sorting nexin (SNX27) and NHERF specifically interact with the 5-HT4a receptor splice variant: roles in receptor targeting. J. Cell Sci. 117, 5367–5379 78 Lunn, M. L., Nassirpour, R., Arrabit, C., Tan, J., McLeod, I., Arias, C. M., Sawchenko, P. E., Yates, III, J.R. and Slesinger, P. A. (2007) A unique sorting nexin regulates trafficking of potassium channels via a PDZ domain interaction. Nat. Neurosci. 10, 1249–1259 79 Rincon, E., de Guinoa, J. S., Gharbi, S. I., Sorzano, C. O., Carrasco, Y. R. and Merida, I. (2011) Translocation dynamics of sorting nexin 27 in activated T cells. J. Cell Sci. 124, 776–788 80 Rincon, E., Santos, T., Avila-Flores, A., Albar, J. P., Lalioti, V., Lei, C., Hong, W. and Merida, I. (2007) Proteomics identification of sorting nexin 27 as a diacylglycerol kinase ζ -associated protein: new diacylglycerol kinase roles in endocytic recycling. Mol. Cell. Proteomics 6, 1073–1087 81 Betts, G. N., van der Geer, P. and Komives, E. A. (2008) Structural and functional consequences of tyrosine phosphorylation in the LRP1 cytoplasmic domain. J. Biol. Chem. 283, 15656–15664 82 Donoso, M., Cancino, J., Lee, J., van Kerkhof, P., Retamal, C., Bu, G., Gonzalez, A., Caceres, A. and Marzolo, M. P. (2009) Polarized traffic of LRP1 involves AP1B and SNX17 operating on Y-dependent sorting motifs in different pathways. Mol. Biol. Cell 20, 481–497 83 Stockinger, W., Sailler, B., Strasser, V., Recheis, B., Fasching, D., Kahr, L., Schneider, W. J. and Nimpf, J. (2002) The PX-domain protein SNX17 interacts with members of the LDL receptor family and modulates endocytosis of the LDL receptor. EMBO J. 21, 4259–4267 84 Burden, J. J., Sun, X. M., Garcia, A. B. and Soutar, A. K. (2004) Sorting motifs in the intracellular domain of the low density lipoprotein receptor interact with a novel domain of sorting nexin-17. J. Biol. Chem. 279, 16237–16245 85 Florian, V., Schluter, T. and Bohnensack, R. (2001) A new member of the sorting nexin family interacts with the C-terminus of P-selectin. Biochem. Biophys. Res. Commun. 281, 1045–1050 86 Williams, R., Schluter, T., Roberts, M. S., Knauth, P., Bohnensack, R. and Cutler, D. F. (2004) Sorting nexin 17 accelerates internalization yet retards degradation of P-selectin. Mol. Biol. Cell 15, 3095–3105 87 Lee, J., Retamal, C., Cuitino, L., Caruano-Yzermans, A., Shin, J. E., van Kerkhof, P., Marzolo, M. P. and Bu, G. (2008) Adaptor protein sorting nexin 17 regulates amyloid precursor protein trafficking and processing in the early endosomes. J. Biol. Chem. 283, 11501–11508 88 Wu, C., Ma, M. H., Brown, K. R., Geisler, M., Li, L., Tzeng, E., Jia, C. Y., Jurisica, I. and Li, S. S. (2007) Systematic identification of SH3 domain-mediated human protein– protein interactions by peptide array target screening. Proteomics 7, 1775–1785 89 Ghai, R. and Collins, B. M. (2011) PX-FERM proteins: a link between endosomal trafficking and signaling? Small GTPases 2, 1–5 90 Balana, B., Maslennikov, I., Kwiatkowski, W., Stern, K. M., Bahima, L., Choe, S. and Slesinger, P. A. (2011) Mechanism underlying selective regulation of G protein-gated inwardly rectifying potassium channels by the psychostimulant-sensitive sorting nexin 27. Proc. Natl. Acad. Sci. U.S.A. 108, 5831–5836 91 Fujiyama, K., Kajii, Y., Hiraoka, S. and Nishikawa, T. (2003) Differential regulation by stimulants of neocortical expression of mrt1, arc , and homer1a mRNA in the rats treated with repeated methamphetamine. Synapse 49, 143–149 92 Kajii, Y., Muraoka, S., Hiraoka, S., Fujiyama, K., Umino, A. and Nishikawa, T. (2003) A developmentally regulated and psychostimulant-inducible novel rat gene mrt1 encoding PDZ-PX proteins isolated in the neocortex. Mol. Psychiatry 8, 434–444 93 MacNeil, A. J., Mansour, M. and Pohajdak, B. (2007) Sorting nexin 27 interacts with the Cytohesin associated scaffolding protein (CASP) in lymphocytes. Biochem. Biophys. Res. Commun. 359, 848–853 c The Authors Journal compilation c 2012 Biochemical Society 56 R. D. Teasdale and B. M. Collins 94 MacNeil, A. J. and Pohajdak, B. (2009) Getting a GRASP on CASP: properties and role of the cytohesin-associated scaffolding protein in immunity. Immunol. Cell Biol. 87, 72–80 95 Nassirpour, R. and Slesinger, P. A. (2007) Subunit-specific regulation of Kir3 channels by sorting nexin 27. Channels 1, 331–333 96 Cai, L., Loo, L. S., Atlashkin, V., Hanson, B. J. and Hong, W. (2011) Deficiency of sorting nexin 27 (SNX27) leads to growth retardation and elevated levels of N-methylD-aspartate receptor 2C (NR2C).. Mol. Cell. Biol. 31, 1734–1747 97 Lauffer, B. E., Melero, C., Temkin, P., Lei, C., Hong, W., Kortemme, T. and von Zastrow, M. (2010) SNX27 mediates PDZ-directed sorting from endosomes to the plasma membrane. J. Cell Biol. 190, 565–574 98 Temkin, P., Lauffer, B., Jager, S., Cimermancic, P., Krogan, N. J. and von Zastrow, M. (2011) SNX27 mediates retromer tubule entry and endosome-to-plasma membrane trafficking of signalling receptors. Nat. Cell Biol. 13, 715–721 99 Rottner, K., Hanisch, J. and Campellone, K. G. (2010) WASH, WHAMM and JMY: regulation of Arp2/3 complex and beyond. Trends Cell Biol. 20, 650–661 100 Gomez, T. S. and Billadeau, D. D. (2009) A FAM21-containing WASH complex regulates retromer-dependent sorting. Dev. Cell 17, 699–711 101 Harbour, M. E., Breusegem, S. Y., Antrobus, R., Freeman, C., Reid, E. and Seaman, M. N. (2010) The cargo-selective retromer complex is a recruiting hub for protein complexes that regulate endosomal tubule dynamics. J. Cell Sci. 123, 3703–3717 102 Zheng, B., Lavoie, C., Tang, T. D., Ma, P., Meerloo, T., Beas, A. and Farquhar, M. G. (2004) Regulation of epidermal growth factor receptor degradation by heterotrimeric Gα s protein. Mol. Biol. Cell 15, 5538–5550 103 Zheng, B., Ma, Y. C., Ostrom, R. S., Lavoie, C., Gill, G. N., Insel, P. A., Huang, X. Y. and Farquhar, M. G. (2001) RGS-PX1, a GAP for Gα s and sorting nexin in vesicular trafficking. Science 294, 1939–1942 104 Zheng, B., Tang, T., Tang, N., Kudlicka, K., Ohtsubo, K., Ma, P., Marth, J. D., Farquhar, M. G. and Lehtonen, E. (2006) Essential role of RGS-PX1/sorting nexin 13 in mouse development and regulation of endocytosis dynamics. Proc. Natl. Acad. Sci. U.S.A. 103, 16776–16781 105 Hao, X., Wang, Y., Ren, F., Zhu, S., Ren, Y., Jia, B., Li, Y. P., Shi, Y. and Chang, Z. (2011) SNX25 regulates TGF-β signaling by enhancing the receptor degradation. Cell. Signal. 23, 935–946 106 Jean-Baptiste, G., Yang, Z. and Greenwood, M. T. (2006) Regulatory mechanisms involved in modulating RGS function. Cell. Mol. Life Sci. 63, 1969–1985 107 Willars, G. B. (2006) Mammalian RGS proteins: multifunctional regulators of cellular signalling. Semin. Cell Dev. Biol. 17, 363–376 108 Sadowski, L., Pilecka, I. and Miaczynska, M. (2009) Signaling from endosomes: location makes a difference. Exp. Cell Res. 315, 1601–1609 109 Kan, A., Ikeda, T., Saito, T., Yano, F., Fukai, A., Hojo, H., Ogasawara, T., Ogata, N., Nakamura, K., Chung, U. I. and Kawaguchi, H. (2009) Screening of chondrogenic factors with a real-time fluorescence-monitoring cell line ATDC5-C2ER: identification of sorting nexin 19 as a novel factor. Arthritis Rheum. 60, 3314–3323 110 Suh, J. M., Stenesen, D., Peters, J. M., Inoue, A., Cade, A. and Graff, J. M. (2008) An RGS-containing sorting nexin controls Drosophila lifespan. PLoS ONE 3, e2152 111 Qin, B., He, M., Chen, X. and Pei, D. (2006) Sorting nexin 10 induces giant vacuoles in mammalian cells. J. Biol. Chem. 281, 36891–36896 112 Mor, A., Wynne, J. P., Ahearn, I. M., Dustin, M. L., Du, G. and Philips, M. R. (2009) Phospholipase D1 regulates lymphocyte adhesion via upregulation of Rap1 at the plasma membrane. Mol. Cell. Biol. 29, 3297–3306 113 Xu, Y., Hortsman, H., Seet, L., Wong, S. H. and Hong, W. (2001) SNX3 regulates endosomal function through its PX-domain-mediated interaction with PtdIns(3)P . Nat. Cell Biol. 3, 658–666 114 Pons, V., Luyet, P. P., Morel, E., Abrami, L., van der Goot, F. G., Parton, R. G. and Gruenberg, J. (2008) Hrs and SNX3 functions in sorting and membrane invagination within multivesicular bodies. PLoS Biol. 6, e214 115 Boulkroun, S., Ruffieux-Daidie, D., Vitagliano, J. J., Poirot, O., Charles, R. P., Lagnaz, D., Firsov, D., Kellenberger, S. and Staub, O. (2008) Vasopressin-inducible ubiquitin-specific protease 10 increases ENaC cell surface expression by deubiquitylating and stabilizing sorting nexin 3. Am. J. Physiol. Renal Physiol. 295, F889–F900 116 Mizutani, R., Yamauchi, J., Kusakawa, S., Nakamura, K., Sanbe, A., Torii, T., Miyamoto, Y. and Tanoue, A. (2009) Sorting nexin 3, a protein upregulated by lithium, contains a novel phosphatidylinositol-binding sequence and mediates neurite outgrowth in N1E-115 cells. Cell. Signal. 21, 1586–1594 117 Strochlic, T. I., Schmiedekamp, B. C., Lee, J., Katzmann, D. J. and Burd, C. G. (2008) Opposing activities of the Snx3-retromer complex and ESCRT proteins mediate regulated cargo sorting at a common endosome. Mol. Biol. Cell 19, 4694–4706 118 Strochlic, T. I., Setty, T. G., Sitaram, A. and Burd, C. G. (2007) Grd19/Snx3p functions as a cargo-specific adapter for retromer-dependent endocytic recycling. J. Cell Biol. 177, 115–125 c The Authors Journal compilation c 2012 Biochemical Society 119 Voos, W. and Stevens, T. H. (1998) Retrieval of resident late-Golgi membrane proteins from the prevacuolar compartment of Saccharomyces cerevisiae is dependent on the function of Grd19p. J. Cell Biol. 140, 577–590 120 Harterink, M., Port, F., Lorenowicz, M. J., McGough, I. J., Silhankova, M., Betist, M. C., van Weering, J. R., van Heesbeen, R. G., Middelkoop, T. C., Basler, K. et al. (2011) A SNX3-dependent retromer pathway mediates retrograde transport of the Wnt sorting receptor Wntless and is required for Wnt secretion. Nat. Cell Biol. 13, 914–923 121 Takemoto, Y., Furuta, M., Sato, M., Kubo, M. and Hashimoto, Y. (1999) Isolation and characterization of a novel HS1 SH3 domain binding protein, HS1BP3. Int. Immunol. 11, 1957–1964 122 Higgins, J. J., Lombardi, R. Q., Pucilowska, J., Jankovic, J., Golbe, L. I. and Verhagen, L. (2006) HS1-BP3 gene variant is common in familial essential tremor. Mov. Disord. 21, 306–309 123 Higgins, J. J., Lombardi, R. Q., Pucilowska, J., Jankovic, J., Tan, E. K. and Rooney, J. P. (2005) A variant in the HS1-BP3 gene is associated with familial essential tremor. Neurology 64, 417–421 124 Shatunov, A., Jankovic, J., Elble, R., Sambuughin, N., Singleton, A., Hallett, M. and Goldfarb, L. (2005) A variant in the HS1-BP3 gene is associated with familial essential tremor. Neurology 65, 1995 125 Karpenahalli, M. R., Lupas, A. N. and Soding, J. (2007) TPRpred: a tool for prediction of TPR-, PPR- and SEL1-like repeats from protein sequences. BMC Bioinformatics 8, 2 126 Choi, J. H., Hong, W. P., Kim, M. J., Kim, J. H., Ryu, S. H. and Suh, P. G. (2004) Sorting nexin 16 regulates EGF receptor trafficking by phosphatidylinositol-3-phosphate interaction with the Phox domain. J. Cell Sci. 117, 4209–4218 127 Hanson, B. J. and Hong, W. (2003) Evidence for a role of SNX16 in regulating traffic between the early and later endosomal compartments. J. Biol. Chem. 278, 34617–34630 128 Le Blanc, I., Luyet, P. P., Pons, V., Ferguson, C., Emans, N., Petiot, A., Mayran, N., Demaurex, N., Faure, J., Sadoul, R. et al. (2005) Endosome-to-cytosol transport of viral nucleocapsids. Nat. Cell Biol. 7, 653–664 129 Rodal, A. A., Blunk, A. D., Akbergenova, Y., Jorquera, R. A., Buhl, L. K. and Littleton, J. T. (2011) A presynaptic endosomal trafficking pathway controls synaptic growth signaling. J. Cell. Biol. 193, 201–217 130 Buchanan, S. G. and Gay, N. J. (1996) Structural and functional diversity in the leucine-rich repeat family of proteins. Prog. Biophys. Mol. Biol. 65, 1–44 131 Piletz, J. E., Ivanov, T. R., Sharp, J. D., Ernsberger, P., Chang, C. H., Pickard, R. T., Gold, G., Roth, B., Zhu, H., Jones, J. C. et al. (2000) Imidazoline receptor antisera-selected (IRAS) cDNA: cloning and characterization. DNA Cell Biol. 19, 319–329 132 Lim, K. P. and Hong, W. (2004) Human Nischarin/imidazoline receptor antisera-selected protein is targeted to the endosomes by a combined action of a PX domain and a coiled-coil region. J. Biol. Chem. 279, 54770–54782 133 Sano, H., Liu, S. C., Lane, W. S., Piletz, J. E. and Lienhard, G. E. (2002) Insulin receptor substrate 4 associates with the protein IRAS. J. Biol. Chem. 277, 19439–19447 134 Alahari, S. K. (2003) Nischarin inhibits Rac induced migration and invasion of epithelial cells by affecting signaling cascades involving PAK. Exp. Cell Res. 288, 415–424 135 Alahari, S. K., Lee, J. W. and Juliano, R. L. (2000) Nischarin, a novel protein that interacts with the integrin α5 subunit and inhibits cell migration. J. Cell Biol. 151, 1141–1154 136 Alahari, S. K. and Nasrallah, H. (2004) A membrane proximal region of the integrin α5 subunit is important for its interaction with nischarin. Biochem. J. 377, 449–457 137 Alahari, S. K., Reddig, P. J. and Juliano, R. L. (2004) The integrin-binding protein Nischarin regulates cell migration by inhibiting PAK. EMBO J. 23, 2777–2788 138 Reddig, P. J., Xu, D. and Juliano, R. L. (2005) Regulation of p21-activated kinaseindependent Rac1 signal transduction by nischarin. J. Biol. Chem. 280, 30994–31002 139 Phillips, S. A., Barr, V. A., Haft, D. H., Taylor, S. I. and Haft, C. R. (2001) Identification and characterization of SNX15, a novel sorting nexin involved in protein trafficking. J. Biol. Chem. 276, 5074–5084 140 Barr, V. A., Phillips, S. A., Taylor, S. I. and Haft, C. R. (2000) Overexpression of a novel sorting nexin, SNX15, affects endosome morphology and protein trafficking. Traffic 1, 904–916 141 Henne, W. M., Buchkovich, N. J. and Emr, S. D. (2011) The ESCRT pathway. Dev. Cell 21, 77–91 142 Ciccarelli, F. D., Proukakis, C., Patel, H., Cross, H., Azam, S., Patton, M. A., Bork, P. and Crosby, A. H. (2003) The identification of a conserved domain in both spartin and spastin, mutated in hereditary spastic paraplegia. Genomics 81, 437–441 143 Hurley, J. H. and Yang, D. (2008) MIT domainia. Dev. Cell 14, 6–8 144 Scott, A., Gaspar, J., Stuchell-Brereton, M. D., Alam, S. L., Skalicky, J. J. and Sundquist, W. I. (2005) Structure and ESCRT-III protein interactions of the MIT domain of human VPS4A. Proc. Natl. Acad. Sci. U.S.A. 102, 13813–13818 145 Hoepfner, S., Severin, F., Cabezas, A., Habermann, B., Runge, A., Gillooly, D., Stenmark, H. and Zerial, M. (2005) Modulation of receptor recycling and degradation by the endosomal kinesin KIF16B. Cell 121, 437–450 146 Miki, H., Okada, Y. and Hirokawa, N. (2005) Analysis of the kinesin superfamily: insights into structure and function. Trends Cell Biol. 15, 467–476 The PX protein family 147 Hirokawa, N., Noda, Y., Tanaka, Y. and Niwa, S. (2009) Kinesin superfamily motor proteins and intracellular transport. Nat. Rev. Mol. Cell Biol. 10, 682–696 148 Blatner, N. R., Wilson, M. I., Lei, C., Hong, W., Murray, D., Williams, R. L. and Cho, W. (2007) The structural basis of novel endosome anchoring activity of KIF16B kinesin. EMBO J. 26, 3709–3719 149 Casimir, C. M., Bu-Ghanim, H. N., Rodaway, A. R., Bentley, D. L., Rowe, P. and Segal, A. W. (1991) Autosomal recessive chronic granulomatous disease caused by deletion at a dinucleotide repeat. Proc. Natl. Acad. Sci. U.S.A. 88, 2753–2757 150 Goldblatt, D. and Thrasher, A. J. (2000) Chronic granulomatous disease. Clin. Exp. Immunol. 122, 1–9 151 Noack, D., Rae, J., Cross, A. R., Ellis, B. A., Newburger, P. E., Curnutte, J. T. and Heyworth, P. G. (2001) Autosomal recessive chronic granulomatous disease caused by defects in NCF-1, the gene encoding the phagocyte p47-phox: mutations not arising in the NCF-1 pseudogenes. Blood 97, 305–311 152 Kanai, F., Liu, H., Field, S. J., Akbary, H., Matsuo, T., Brown, G. E., Cantley, L. C. and Yaffe, M. B. (2001) The PX domains of p47phox and p40phox bind to lipid products of PI(3)K. Nat. Cell Biol. 3, 675–678 153 Vervoort, V. S., Viljoen, D., Smart, R., Suthers, G., DuPont, B. R., Abbott, A. and Schwartz, C. E. (2002) Sorting nexin 3 (SNX3) is disrupted in a patient with a translocation t(6;13)(q21;q12) and microcephaly, microphthalmia, ectrodactyly, prognathism (MMEP) phenotype. J. Med. Genet. 39, 893–899 154 Kumar, R. A., Everman, D. B., Morgan, C. T., Slavotinek, A., Schwartz, C. E. and Simpson, E. M. (2007) Absence of mutations in NR2E1 and SNX3 in five patients with MMEP (microcephaly, microphthalmia, ectrodactyly, and prognathism) and related phenotypes. BMC Med. Genet. 8, 48 155 Harley, J. B., Alarcon-Riquelme, M. E., Criswell, L. A., Jacob, C. O., Kimberly, R. P., Moser, K. L., Tsao, B. P., Vyse, T. J., Langefeld, C. D., Nath, S. K. et al. (2008) Genome-wide association scan in women with systemic lupus erythematosus identifies susceptibility variants in ITGAM , PXK , KIAA1542 and other loci. Nat. Genet. 40, 204–210 156 Stefaniuk, M. and Lukasiuk, K. (2010) Cloning of expressed sequence tags (ESTs) representing putative epileptogenesis-related genes and the localization of their expression in the normal brain. Neurosci. Lett. 482, 230–234 157 Vicinanza, M., D’Angelo, G., Di Campli, A. and De Matteis, M. A. (2008) Function and dysfunction of the PI system in membrane trafficking. EMBO J. 27, 2457–2470 158 Vicinanza, M., D’Angelo, G., Di Campli, A. and De Matteis, M. A. (2008) Phosphoinositides as regulators of membrane trafficking in health and disease. Cell. Mol. Life Sci. 65, 2833–2841 159 Vanhaesebroeck, B., Guillermet-Guibert, J., Graupera, M. and Bilanges, B. (2010) The emerging mechanisms of isoform-specific PI3K signalling. Nat. Rev. Mol. Cell Biol. 11, 329–341 160 Nicot, A. S. and Laporte, J. (2008) Endosomal phosphoinositides and human diseases. Traffic 9, 1240–1249 161 Kerr, M. C., Wang, J. T., Castro, N. A., Hamilton, N. A., Town, L., Brown, D. L., Meunier, F. A., Brown, N. F., Stow, J. L. and Teasdale, R. D. (2010) Inhibition of the PtdIns(5) kinase PIKfyve disrupts intracellular replication of Salmonella . EMBO J. 29, 1331–1347 162 Krol, M., Polanska, J., Pawlowski, K. M., Turowski, P., Skierski, J., Majewska, A., Ugorski, M., Morty, R. E. and Motyl, T. (2010) Transcriptomic signature of cell lines isolated from canine mammary adenocarcinoma metastases to lungs. J. Appl. Genet. 51, 37–50 163 Seals, D. F., Azucena, Jr, E.F., Pass, I., Tesfay, L., Gordon, R., Woodrow, M., Resau, J. H. and Courtneidge, S. A. (2005) The adaptor protein Tks5/Fish is required for podosome formation and function, and for the protease-driven invasion of cancer cells. Cancer Cell 7, 155–165 164 Blouw, B., Seals, D. F., Pass, I., Diaz, B. and Courtneidge, S. A. (2008) A role for the podosome/invadopodia scaffold protein Tks5 in tumor growth in vivo . Eur. J. Cell Biol. 87, 555–567 165 Koutros, S., Schumacher, F. R., Hayes, R. B., Ma, J., Huang, W. Y., Albanes, D., Canzian, F., Chanock, S. J., Crawford, E. D., Diver, W. R. et al. (2010) Pooled analysis of phosphatidylinositol 3-kinase pathway variants and risk of prostate cancer. Cancer Res. 70, 2389–2396 166 Nguyen, L. N., Holdren, M. S., Nguyen, A. P., Furuya, M. H., Bianchini, M., Levy, E., Mordoh, J., Liu, A., Guncay, G. D., Campbell, J. S. and Parks, W. T. (2006) Sorting nexin 1 down-regulation promotes colon tumorigenesis. Clin. Cancer Res. 12, 6952–6959 167 Huang, Z., Huang, S., Wang, Q., Liang, L., Ni, S., Wang, L., Sheng, W., He, X. and Du, X. (2011) MicroRNA-95 promotes cell proliferation and targets sorting nexin 1 in human colorectal carcinoma. Cancer Res. 71, 2582–2589 168 Vasudevan, K. M., Barbie, D. A., Davies, M. A., Rabinovsky, R., McNear, C. J., Kim, J. J., Hennessy, B. T., Tseng, H., Pochanard, P., Kim, S. Y. et al. (2009) AKT-independent signaling downstream of oncogenic PIK3CA mutations in human cancer. Cancer Cell 16, 21–32 57 169 Wang, Y., Zhou, D., Phung, S., Masri, S., Smith, D. and Chen, S. (2011) SGK3 is an estrogen-inducible kinase promoting estrogen-mediated survival of breast cancer cells. Mol. Endocrinol. 25, 72–82 170 Slagsvold, T., Marchese, A., Brech, A. and Stenmark, H. (2006) CISK attenuates degradation of the chemokine receptor CXCR4 via the ubiquitin ligase AIP4. EMBO J. 25, 3738–3749 171 Foster, D. A. and Xu, L. (2003) Phospholipase D in cell proliferation and cancer. Mol. Cancer Res. 1, 789–800 172 Peng, X. and Frohman, M. A. (2011) Mammalian phospholipase D physiological and pathological roles. Acta Physiol., doi: 10.1111/j.1748-1716.2011.02298.x 173 Su, W., Chen, Q. and Frohman, M. A. (2009) Targeting phospholipase D with small-molecule inhibitors as a potential therapeutic approach for cancer metastasis. Future Oncol. 5, 1477–1486 174 Lavieri, R. R., Scott, S. A., Selvy, P. E., Kim, K., Jadhav, S., Morrison, R. D., Daniels, J. S., Brown, H. A. and Lindsley, C. W. (2010) Design, synthesis, and biological evaluation of halogenated N -(2-(4-oxo-1-phenyl-1,3,8-triazaspiro[4.5]decan-8-yl)ethyl)benzamides: discovery of an isoform-selective small molecule phospholipase D2 inhibitor. J. Med. Chem. 53, 6706–6719 175 Scott, S. A., Selvy, P. E., Buck, J. R., Cho, H. P., Criswell, T. L., Thomas, A. L., Armstrong, M. D., Arteaga, C. L., Lindsley, C. W. and Brown, H. A. (2009) Design of isoform-selective phospholipase D inhibitors that modulate cancer cell invasiveness. Nat. Chem. Biol. 5, 108–117 176 Tabuchi, A. and Kuebler, W. M. (2008) Endothelium-platelet interactions in inflammatory lung disease. Vascul. Pharmacol. 49, 141–150 177 Ludwig, R. J., Schon, M. P. and Boehncke, W. H. (2007) P-selectin: a common therapeutic target for cardiovascular disorders, inflammation and tumour metastasis. Expert Opin. Ther. Targets 11, 1103–1117 178 Chen, M. and Geng, J. G. (2006) P-selectin mediates adhesion of leukocytes, platelets, and cancer cells in inflammation, thrombosis, and cancer growth and metastasis. Arch. Immunol. Ther. Exp. 54, 75–84 179 Querfurth, H. W. and LaFerla, F. M. (2010) Alzheimer’s disease. N. Engl. J. Med. 362, 329–344 180 Sannerud, R. and Annaert, W. (2009) Trafficking, a key player in regulated intramembrane proteolysis. Semin. Cell Dev. Biol. 20, 183–190 181 Andersen, O. M. and Willnow, T. E. (2006) Lipoprotein receptors in Alzheimer’s disease. Trends Neurosci. 29, 687–694 182 Bu, G. (2009) Apolipoprotein E and its receptors in Alzheimer’s disease: pathways, pathogenesis and therapy. Nat. Rev. Neurosci. 10, 333–344 183 Bu, G., Cam, J. and Zerbinatti, C. (2006) LRP in amyloid-β production and metabolism. Ann. N. Y. Acad. Sci. 1086, 35–53 184 Thinakaran, G. and Koo, E. H. (2008) Amyloid precursor protein trafficking, processing, and function. J. Biol. Chem. 283, 29615–29619 185 Wolfe, M. S. and Guenette, S. Y. (2007) APP at a glance. J. Cell Sci. 120, 3157–3161 186 Nielsen, M. S., Gustafsen, C., Madsen, P., Nyengaard, J. R., Hermey, G., Bakke, O., Mari, M., Schu, P., Pohlmann, R., Dennes, A. and Petersen, C. M. (2007) Sorting by the cytoplasmic domain of the amyloid precursor protein binding receptor SorLA. Mol. Cell. Biol. 27, 6842–6851 187 Rogaeva, E., Meng, Y., Lee, J. H., Gu, Y., Kawarai, T., Zou, F., Katayama, T., Baldwin, C. T., Cheng, R., Hasegawa, H. et al. (2007) The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease. Nat. Genet. 39, 168–177 188 He, X., Li, F., Chang, W. P. and Tang, J. (2005) GGA proteins mediate the recycling pathway of memapsin 2 (BACE). J. Biol. Chem. 280, 11696–11703 189 Muhammad, A., Flores, I., Zhang, H., Yu, R., Staniszewski, A., Planel, E., Herman, M., Ho, L., Kreber, R., Honig, L. S. et al. (2008) Retromer deficiency observed in Alzheimer’s disease causes hippocampal dysfunction, neurodegeneration, and Aβ accumulation. Proc. Natl. Acad. Sci. U.S.A. 105, 7327–7332 190 Okada, H., Zhang, W., Peterhoff, C., Hwang, J. C., Nixon, R. A., Ryu, S. H. and Kim, T. W. (2010) Proteomic identification of sorting nexin 6 as a negative regulator of BACE1mediated APP processing. FASEB J. 24, 2783–2794 191 Shin, N., Lee, S., Ahn, N., Kim, S. A., Ahn, S. G., YongPark, Z. and Chang, S. (2007) Sorting nexin 9 interacts with dynamin 1 and N-WASP and coordinates synaptic vesicle endocytosis. J. Biol. Chem. 282, 28939–28950 192 Soulet, F., Yarar, D., Leonard, M. and Schmid, S. L. (2005) SNX9 regulates dynamin assembly and is required for efficient clathrin-mediated endocytosis. Mol. Biol. Cell 16, 2058–2067 193 Lundmark, R. and Carlsson, S. R. (2002) The β-appendages of the four adaptor-protein (AP) complexes: structure and binding properties, and identification of sorting nexin 9 as an accessory protein to AP-2. Biochem J. 362, 597–607 194 Yun, M., Keshvara, L., Park, C. G., Zhang, Y. M., Dickerson, J. B., Zheng, J., Rock, C. O., Curran, T. and Park, H. W. (2003) Crystal structures of the Dab homology domains of mouse disabled 1 and 2. J. Biol. Chem. 278, 36572–36581 c The Authors Journal compilation c 2012 Biochemical Society 58 R. D. Teasdale and B. M. Collins 195 Duffy, M. J., McKiernan, E., O’Donovan, N. and McGowan, P. M. (2009) The role of ADAMs in disease pathophysiology. Clin. Chim. Acta 403, 31–36 196 Kveiborg, M., Albrechtsen, R., Couchman, J. R. and Wewer, U. M. (2008) Cellular roles of ADAM12 in health and disease. Int. J. Biochem. Cell. Biol. 40, 1685–1702 197 Abram, C. L., Seals, D. F., Pass, I., Salinsky, D., Maurer, L., Roth, T. M. and Courtneidge, S. A. (2003) The adaptor protein fish associates with members of the ADAMs family and localizes to podosomes of Src-transformed cells. J. Biol. Chem. 278, 16844–16851 198 Zhong, J. L., Poghosyan, Z., Pennington, C. J., Scott, X., Handsley, M. M., Warn, A., Gavrilovic, J., Honert, K., Kruger, A., Span, P. N. et al. (2008) Distinct functions of natural ADAM-15 cytoplasmic domain variants in human mammary carcinoma. Mol. Cancer Res. 6, 383–394 199 Malinin, N. L., Wright, S., Seubert, P., Schenk, D. and Griswold-Prenner, I. (2005) Amyloid-β neurotoxicity is mediated by FISH adapter protein and ADAM12 metalloprotease activity. Proc. Natl. Acad. Sci. U.S.A. 102, 3058–3063 200 Harold, D., Jehu, L., Turic, D., Hollingworth, P., Moore, P., Summerhayes, P., Moskvina, V., Foy, C., Archer, N., Hamilton, B. A. et al. (2007) Interaction between the ADAM12 and SH3MD1 genes may confer susceptibility to late-onset Alzheimer’s disease. Am. J. Med. Genet. B Neuropsychiatr. Genet. 144B, 448–452 201 Oliveira, T. G. and Di Paolo, G. (2010) Phospholipase D in brain function and Alzheimer’s disease. Biochim. Biophys. Acta 1801, 799–805 202 Jin, J. K., Ahn, B. H., Na, Y. J., Kim, J. I., Kim, Y. S., Choi, E. K., Ko, Y. G., Chung, K. C., Kozlowski, P. B. and Min do, S. (2007) Phospholipase D1 is associated with amyloid precursor protein in Alzheimer’s disease. Neurobiol. Aging 28, 1015–1027 203 Jin, J. K., Kim, N. H., Lee, Y. J., Kim, Y. S., Choi, E. K., Kozlowski, P. B., Park, M. H., Kim, H. S. and Min do, S. (2006) Phospholipase D1 is up-regulated in the mitochondrial fraction from the brains of Alzheimer’s disease patients. Neurosci. Lett. 407, 263–267 204 Cai, D., Netzer, W. J., Zhong, M., Lin, Y., Du, G., Frohman, M., Foster, D. A., Sisodia, S. S., Xu, H., Gorelick, F. S. and Greengard, P. (2006) Presenilin-1 uses phospholipase D1 as a negative regulator of β-amyloid formation. Proc. Natl. Acad. Sci. U.S.A. 103, 1941–1946 205 Cai, D., Zhong, M., Wang, R., Netzer, W. J., Shields, D., Zheng, H., Sisodia, S. S., Foster, D. A., Gorelick, F. S., Xu, H. and Greengard, P. (2006) Phospholipase D1 corrects impaired βAPP trafficking and neurite outgrowth in familial Alzheimer’s disease-linked presenilin-1 mutant neurons. Proc. Natl. Acad. Sci. U.S.A. 103, 1936–1940 206 Liu, Y., Zhang, Y. W., Wang, X., Zhang, H., You, X., Liao, F. F. and Xu, H. (2009) Intracellular trafficking of presenilin 1 is regulated by β-amyloid precursor protein and phospholipase D1. J. Biol. Chem. 284, 12145–12152 207 Oliveira, T. G., Chan, R. B., Tian, H., Laredo, M., Shui, G., Staniszewski, A., Zhang, H., Wang, L., Kim, T. W., Duff, K. E. et al. (2010) Phospholipase D2 ablation ameliorates Alzheimer’s disease-linked synaptic dysfunction and cognitive deficits. J. Neurosci. 30, 16419–16428 208 Owen, D. J. and Collins, B. M. (2010) Vesicle transport: a new player in APP trafficking. Curr. Biol. 20, R413–R415 209 Gruenberg, J. and van der Goot, F. G. (2006) Mechanisms of pathogen entry through the endosomal compartments. Nat. Rev. Mol. Cell Biol. 7, 495–504 210 Kerr, M. C. and Teasdale, R. D. (2009) Defining macropinocytosis. Traffic 10, 364–371 211 Schroeder, N., Mota, L. J. and Meresse, S. (2011) Salmonella -induced tubular networks. Trends Microbiol. 19, 268–277 212 Bujny, M. V., Ewels, P. A., Humphrey, S., Attar, N., Jepson, M. A. and Cullen, P. J. (2008) Sorting nexin-1 defines an early phase of Salmonella -containing vacuole-remodeling during Salmonella infection. J. Cell Sci. 121, 2027–2036 213 Braun, V., Wong, A., Landekic, M., Hong, W. J., Grinstein, S. and Brumell, J. H. (2010) Sorting nexin 3 (SNX3) is a component of a tubular endosomal network induced by Salmonella and involved in maturation of the Salmonella -containing vacuole. Cell. Microbiol. 12, 1352–1367 214 Wang, J. T., Kerr, M. C., Karunaratne, S., Jeanes, A., Yap, A. S. and Teasdale, R. D. (2010) The SNX-PX-BAR family in macropinocytosis: the regulation of macropinosome formation by SNX-PX-BAR proteins. PLoS ONE 5, e13763 215 Almendinger, J., Doukoumetzidis, K., Kinchen, J. M., Kaech, A., Ravichandran, K. S. and Hengartner, M. O. (2011) A conserved role for SNX9-family members in the regulation of phagosome maturation during engulfment of apoptotic cells. PLoS ONE 6, e18325 216 Chen, D., Xiao, H., Zhang, K., Wang, B., Gao, Z., Jian, Y., Qi, X., Sun, J., Miao, L. and Yang, C. (2010) Retromer is required for apoptotic cell clearance by phagocytic receptor recycling. Science 327, 1261–1264 217 Lu, N., Shen, Q., Mahoney, T. R., Liu, X. and Zhou, Z. (2011) Three sorting nexins drive the degradation of apoptotic cells in response to PtdIns(3)P signaling. Mol. Biol. Cell 22, 354–374 218 Cheng, G. and Lambeth, J. D. (2004) NOXO1, regulation of lipid binding, localization, and activation of Nox1 by the Phox homology (PX) domain. J. Biol. Chem. 279, 4737–4742 c The Authors Journal compilation c 2012 Biochemical Society 219 Park, J. B. (2003) Phagocytosis induces superoxide formation and apoptosis in macrophages. Exp. Mol. Med. 35, 325–335 220 Sandvig, K., Grimmer, S., Lauvrak, S. U., Torgersen, M. L., Skretting, G., van Deurs, B. and Iversen, T. G. (2002) Pathways followed by ricin and Shiga toxin into cells. Histochem. Cell Biol. 117, 131–141 221 Popoff, V., Mardones, G. A., Bai, S. K., Chambon, V., Tenza, D., Burgos, P. V., Shi, A., Benaroch, P., Urbe, S., Lamaze, C. et al. (2009) Analysis of articulation between clathrin and retromer in retrograde sorting on early endosomes. Traffic 10, 1868–1880 222 Dyve, A. B., Bergan, J., Utskarpen, A. and Sandvig, K. (2009) Sorting nexin 8 regulates endosome-to-Golgi transport. Biochem. Biophys. Res. Commun. 390, 109–114 223 Nanbo, A., Imai, M., Watanabe, S., Noda, T., Takahashi, K., Neumann, G., Halfmann, P. and Kawaoka, Y. (2010) Ebolavirus is internalized into host cells via macropinocytosis in a viral glycoprotein-dependent manner. PLoS Pathog. 6, e1001121 224 Ju, W., Yoo, B. C., Kim, I. J., Kim, J. W., Kim, S. C. and Lee, H. P. (2009) Identification of genes with differential expression in chemoresistant epithelial ovarian cancer using high-density oligonucleotide microarrays. Oncol. Res. 18, 47–56 225 Nishimura, Y., Yoshioka, K., Bereczky, B. and Itoh, K. (2008) Evidence for efficient phosphorylation of EGFR and rapid endocytosis of phosphorylated EGFR via the early/late endocytic pathway in a gefitinib-sensitive non-small cell lung cancer cell line. Mol. Cancer. 7, 42 226 Alto, N. M., Weflen, A. W., Rardin, M. J., Yarar, D., Lazar, C. S., Tonikian, R., Koller, A., Taylor, S. S., Boone, C., Sidhu, S. S. et al. (2007) The type III effector EspF coordinates membrane trafficking by the spatiotemporal activation of two eukaryotic signaling pathways. J. Cell Biol. 178, 1265–1278 227 Marches, O., Batchelor, M., Shaw, R. K., Patel, A., Cummings, N., Nagai, T., Sasakawa, C., Carlsson, S. R., Lundmark, R., Cougoule, C. et al. (2006) EspF of enteropathogenic Escherichia coli binds sorting nexin 9. J. Bacteriol. 188, 3110–3115 228 Heiseke, A., Schobel, S., Lichtenthaler, S. F., Vorberg, I., Groschup, M. H., Kretzschmar, H., Schatzl, H. M. and Nunziante, M. (2008) The novel sorting nexin SNX33 interferes with cellular PrP formation by modulation of PrP shedding. Traffic 9, 1116–1129 229 Ji, T., Wu, Y., Wang, H., Wang, J. and Jiang, Y. (2010) Diagnosis and fine mapping of a deletion in distal 11q in two Chinese patients with developmental delay. J. Hum. Genet. 55, 486–489 230 Bare, L. A., Morrison, A. C., Rowland, C. M., Shiffman, D., Luke, M. M., Iakoubova, O. A., Kane, J. P., Malloy, M. J., Ellis, S. G., Pankow, J. S. et al. (2007) Five common gene variants identify elevated genetic risk for coronary heart disease. Genet. Med. 9, 682–689 231 Jacques, C., Baris, O., Prunier-Mirebeau, D., Savagner, F., Rodien, P., Rohmer, V., Franc, B., Guyetant, S., Malthiery, Y. and Reynier, P. (2005) Two-step differential expression analysis reveals a new set of genes involved in thyroid oncocytic tumors. J. Clin. Endocrinol. Metab. 90, 2314–2320 232 Tyybakinoja, A., Saarinen-Pihkala, U., Elonen, E. and Knuutila, S. (2006) Amplified, lost, and fused genes in 11q23–25 amplicon in acute myeloid leukemia, an array-CGH study. Genes Chromosomes Cancer 45, 257–264 233 Mestre-Escorihuela, C., Rubio-Moscardo, F., Richter, J. A., Siebert, R., Climent, J., Fresquet, V., Beltran, E., Agirre, X., Marugan, I., Marin, M. et al. (2007) Homozygous deletions localize novel tumor suppressor genes in B-cell lymphomas. Blood 109, 271–280 234 Knobbe, C. B. and Reifenberger, G. (2003) Genetic alterations and aberrant expression of genes related to the phosphatidyl-inositol-3 -kinase/protein kinase B (Akt) signal transduction pathway in glioblastomas. Brain Pathol. 13, 507–518 235 Traer, C. J., Foster, F. M., Abraham, S. M. and Fry, M. J. (2006) Are class II phosphoinositide 3-kinases potential targets for anticancer therapies? Bull. Cancer 93, E53–E58 236 Shen, Q., Stanton, M. L., Feng, W., Rodriguez, M. E., Ramondetta, L., Chen, L., Brown, R. E. and Duan, X. (2010) Morphoproteomic analysis reveals an overexpressed and constitutively activated phospholipase D1–mTORC2 pathway in endometrial carcinoma. Int. J. Clin. Exp. Pathol. 4, 13–21 237 Kang, D. W., Park, M. H., Lee, Y. J., Kim, H. S., Lindsley, C. W., Alex Brown, H. and Min do, S. (2011) Autoregulation of phospholipase D activity is coupled to selective induction of phospholipase D1 expression to promote invasion of breast cancer cells. Int. J. Cancer 128, 805–816 238 Kang, D. W. and Min do, S. (2010) Platelet derived growth factor increases phospholipase D1 but not phospholipase D2 expression via NFκB signaling pathway and enhances invasion of breast cancer cells. Cancer Lett. 294, 125–133 239 Kang, D. W., Lee, J. Y., Oh, D. H., Park, S. Y., Woo, T. M., Kim, M. K., Park, M. H., Jang, Y. H. and Min do, S. (2009) Triptolide-induced suppression of phospholipase D expression inhibits proliferation of MDA-MB-231 breast cancer cells. Exp. Mol. Med. 41, 678–685 The PX protein family 240 Zhong, M., Shen, Y., Zheng, Y., Joseph, T., Jackson, D. and Foster, D. A. (2003) Phospholipase D prevents apoptosis in v-Src-transformed rat fibroblasts and MDA-MB-231 breast cancer cells. Biochem. Biophys. Res. Commun. 302, 615–619 241 Kang, D. W., Park, M. H., Lee, Y. J., Kim, H. S., Kwon, T. K., Park, W. S. and Min do, S. (2008) Phorbol ester up-regulates phospholipase D1 but not phospholipase D2 expression through a PKC/Ras/ERK/NFκB-dependent pathway and enhances matrix metalloproteinase-9 secretion in colon cancer cells. J. Biol. Chem. 283, 4094–4104 242 Henkels, K. M., Farkaly, T., Mahankali, M., Segall, J. E. and Gomez-Cambronero, J. (2011) Cell invasion of highly metastatic MTLn3 cancer cells is dependent on phospholipase D2 (PLD2) and Janus kinase 3 (JAK3). J. Mol. Biol. 408, 850–862 243 Knoepp, S. M., Chahal, M. S., Xie, Y., Zhang, Z., Brauner, D. J., Hallman, M. A., Robinson, S. A., Han, S., Imai, M., Tomlinson, S. and Meier, K. E. (2008) Effects of active and inactive phospholipase D2 on signal transduction, adhesion, migration, invasion, and metastasis in EL4 lymphoma cells. Mol. Pharmacol. 74, 574–584 59 244 Saito, M., Iwadate, M., Higashimoto, M., Ono, K., Takebayashi, Y. and Takenoshita, S. (2007) Expression of phospholipase D2 in human colorectal carcinoma. Oncol. Rep. 18, 1329–1334 245 Oshimoto, H., Okamura, S., Yoshida, M. and Mori, M. (2003) Increased activity and expression of phospholipase D2 in human colorectal cancer. Oncol. Res. 14, 31–37 246 Wright, P. K., May, F. E., Darby, S., Saif, R., Lennard, T. W. and Westley, B. R. (2009) Estrogen regulates vesicle trafficking gene expression in EFF-3, EFM-19 and MCF-7 breast cancer cells. Int. J. Clin. Exp. Pathol. 2, 463–475 247 Osman, I., Bajorin, D. F., Sun, T. T., Zhong, H., Douglas, D., Scattergood, J., Zheng, R., Han, M., Marshall, K. W. and Liew, C. C. (2006) Novel blood biomarkers of human urinary bladder cancer. Clin. Cancer Res. 12, 3374–3380 248 Watahiki, A., Waki, K., Hayatsu, N., Shiraki, T., Kondo, S., Nakamura, M., Sasaki, D., Arakawa, T., Kawai, J., Harbers, M. et al. (2004) Libraries enriched for alternatively spliced exons reveal splicing patterns in melanocytes and melanomas. Nat. Methods 1, 233–239 Received 11 July 2011/26 July 2011; accepted 27 July 2011 Published on the Internet 14 December 2011, doi:10.1042/BJ20111226 c The Authors Journal compilation c 2012 Biochemical Society Biochem. J. (2012) 441, 39–59 (Printed in Great Britain) doi:10.1042/BJ20111226 SUPPLEMENTARY ONLINE DATA Insights into the PX (phox-homology) domain and SNX (sorting nexin) protein families: structures, functions and roles in disease Rohan D. TEASDALE and Brett M. COLLINS1 Institute for Molecular Bioscience, University of Queensland, St. Lucia, Queensland 4072, Australia See the following pages for Supplementary Figures S1–S3 and Supplementary Tables S1–S3. 1 To whom correspondence should be addressed (email [email protected]). c The Authors Journal compilation c 2012 Biochemical Society R. D. Teasdale and B. M. Collins Figure S1 Known structures of mammalian PX domains, including yeast Grd19p/Snx3p There are three known structures of PX domains in complex with PtdIns3P : SNX9, p40phox and yeast Grd19p/Snx3p. These structures show identical binding modes for PtdIns3P . In many other structures, SO4 ions are found in the putative binding site, usually mimicking the location of the 3-phosphate group (associated with the conserved arginine side chain). In this Figure, all 15 known mammalian PX domain structures are shown in surface representation (PDB codes are given below each structure). Only one representative structure is shown for each protein, and for proteins c The Authors Journal compilation c 2012 Biochemical Society The PX protein family Figure S2 Sequence alignment of human SNX20 and SNX21 The PX domain and the C-terminal domain of unknown structure and function are indicated. We refer to this domain as the PXB domain (PX-associated domain B) to fit with terminology adopted for the PXA-RGS-PX-PXC proteins. Secondary-structure predictions calculated with JPRED [1] (http://www.compbio.dundee.ac.uk/www-jpred/) are indicated for SNX20 (above) and SNX21 (below). The alignment was generated using ESPript 2.2 [2] (http://espript.ibcp.fr/ESPript/ESPript/). where more than one structure is available, only PtdIns3P or SO4 -bound structures are shown. Surfaces of the PX domains are coloured according to electrostatic potential red to blue from − 0.5 to + 0.5 V. An Ins(1,3,4,5)P 4 molecule has been positioned in the putative binding site of each structure, based on the known binding orientation of PtdIns3P , to provide an indication of whether 3-, 4- and 5-phosphates of phosphoinositides are likely to fit within the binding pocket. In all cases, steric clashes are observed with at least one of the modelled 4- or 5-phosphate groups, and in some instances the residues required for 3-phosphate co-ordination are missing. Phosphate positions are labelled (3, 4 and 5), and whether the phosphate groups are able to associate is indicated. Reported lipid-binding specificities and key side chains of the binding site are given in Supplementary Table S2. There are no clear cases where structures support the contention that a 4- or 5-phosphorylated phosphoinositide would be able to actively engage the PX domain, without binding in a significantly different orientation to PtdIns3P . In the cases of SNX5, PI3K-C2α and PI3K-C2γ , there appears to be little possibility that any phosphoinositide (including PtdIns3P ) will bind in the preferred conformation. Note 1: structures marked with asterisks (*) are NMR structures, and in these examples the polyproline loop is highly dynamic in conformation and often obscures the binding pocket. This highlights the flexible nature of the polyproline loop (and to a lesser extent the loop between the β1 and β2 strands) and the importance of its conformation for phosphoinositide binding. Note 2: in the IRAS crystal structure, both the flexible polyproline loop and the loop between strands β1 and β2 are in conformations that obscure the binding pocket, but the binding determinants for the PtdIns3P are present. c The Authors Journal compilation c 2012 Biochemical Society R. D. Teasdale and B. M. Collins Figure S3 Cartoon outlining a potential role for PX proteins in inflammation (A) Inflammatory signals trigger the attachment of leucocytes to blood-vessel endothelial cells mediated by interactions of the leucocyte PSGL-1 glycoprotein with endothelial and platelet P-selectin adhesion proteins. The P-selectin protein is stored within α-granules in platelets and Weibel-Palade bodies (WPBs) in endothelial cells. (B) Secretion of P-selectin from Weibel-Palade bodies, along with von Willebrand factor, is promoted by secretagogue signals. In endothelial cells, SNX17 interacts with P-selectin on early endosomes (EE). Overexpression of SNX17 promotes recruitment of P-selectin to endosomes, and retards its degradation in lysosomes. Whether SNX17 is able to promote cell-surface recycling as for members of the LDLR family is unknown. (C) In leucocytes, the glycoprotein PSGL-1 interacts with P-selectin via its glycans containing sialyl Lewis X. Its localization to endosomes is strongly enhanced by SNX20, but it still remains unknown exactly what role SNX20–PSGL-1 interaction plays in PSGL-1 trafficking and signalling. It is also unknown whether the close homologue SNX21 is able to perform similar functions to SNX20 in this process. LE, late endosome; RE, recycling endosome. c The Authors Journal compilation c 2012 Biochemical Society Table S1 Critical phosphoinositide binding determinants in human PX proteins, and structurally predicted phosphoinositide specificities n.d., not determined. P3 Tyrinositol (aromatic stacking with inositol ring†) LysP1 (binds to 1phosphate‡) Arg4,5-hydroxy (binds to 4- and 5-hydroxy groups of inositol ring§) Polyproline loop (potentially clashes with 5-phosphate groups) Reported phosphoinositide preferences PI3P PI3P > PI(3,5)P 2 None PI(4,5)P 2 None n.d. PI3P PI3P PI3P PI3P None n.d. n.d. n.d. PI3P PI(3,4)P 2 ≈ PI(4,5)P 2 ≈ PI(3,5)P 2 ≈ PI(3,4,5)P 3 > PI3P ≈ PI4P PI(4,5)P 2 > PI(3,4)P 2 > PI(3,5)P 2 n.d. Subfamily Protein PDB code (method, ligand) Arg (binds to 3-phosphate*) PX-BAR SNX1 2I4K (NMR, apo) R186 F187 K214 R239 Polyproline loop occludes 5-phosphate SNX2 SNX4 SNX5 R183 R106 3HPC (X-ray, apo), 3HPB (X-ray, apo) F184 Y107 Q68 K211 K132 H69 R236 R155 K96 ? ? T139 PI3P PI3P Polyproline loop occludes 5-phosphate SNX6 Q68 H69 R96 T139 ? SNX7 3IQ2 (X-ray, SO4 ) R73 Y74 K99 R117 R109 R132 Q63 Y110 Y133 H64 K135 K158 R91 R150 R177 T134 The structure of homologue SNX5 suggests that the normal PtdIns-binding pocket is simply not present Polyproline loop is open ? ? ? R286 Y287 K313 R327 Polyproline loop occludes 5-phosphate SNX18 R312 Y313 K338 R353 ? PI3P SNX33 R266 Y267 K292 R308 ? PI3P SNX8 SNX30 SNX32 SH3-PX-BAR SNX9 2RAJ, (X-ray, SO4 ), 2RAK (X-ray, PI3P ), 2RAI (X-ray, apo), 3DYU (X-ray, apo), 3DYT (X-ray, apo) Comments In the reported NMR structure there are significant local conformational differences that occlude the binding site. Nevertheless specific side chains are present in approximately the correct positions compared with known co-crystal structures of other PX domains PI3P > PI(3,5)P 2 n.d. The structure of SNX5 suggests that the normal PtdIns-binding pocket is simply not present K99 will need to reorient to bind the 1-phosphate The structure of homologue SNX5 suggests that the normal PtdInsP -binding pocket is simply not present Note: although the reported specificity is PI(4,5)P 2 for liposomes, PI3P is also bound and only PI3P [not PI(4,5)P 2 ] is able to bind in soaked crystals The PX protein family c The Authors Journal compilation c 2012 Biochemical Society Structurally predicted phosphoinositide specificity¶ Continued PDB code (method, ligand) ArgP3 (binds to 3-phosphate*) Tyrinositol (aromatic stacking with inositol ring†) LysP1 (binds to 1phosphate‡) Arg4,5-hydroxy (binds to 4- and 5-hydroxy groups of inositol ring§) Polyproline loop (potentially clashes with 5-phosphate groups) Structurally predicted phosphoinositide specificity¶ Subfamily Protein PX-SH3 SH3PXD2A R43 Y44 K78 R93 ? PI3P SH3PXD2B SNX28 (NOXO1) R44 S41 Y45 W42 K78 K71 R94 R90 ? ? PI3P None PI3P p40phox 2DYB (X-ray, apo) 1H6H (X-ray, PI3P ) R58 Y59 K92 (plus R60) R105 p47phox 1KQ6 (X-ray, SO4 ), 1O7K (X-ray, SO4 ), 1GD5 (NMR, apo) 3LUI (X-ray, SO4 ), 3FOG (X-ray, apo) R43 F44 K73 R90 R36 Y37 K62 R75 PX-FERM SNX17 PXA-RGS-PX-PXC SNX27 SNX31 SNX13 R196 R37 R601 Y197 Y38 Y602 K222 K62 K628 R236 R76 R642 Polyproline loop occludes 5-phosphate Polyproline loop occludes 5-phosphate Polyproline loop occludes 5-phosphate ? ? ? SNX14 SNX19 SNX25 R616 R582 R552 Y617 Y583 L553 K642 K611 K580 R657 R629 K595 ? ? ? SNX3 SNX10 SNX11 SNX12 SNX22 SNX24 HS1BP3 R70 R53 R59 R71 R43 R38 K70 Y71 Y54 Y60 Y72 Y44 Y39 Y71 K95 K79 K85 K96 K66 K61 K96 R118 R95 R100 R119 R79 R75 R109 ? ? ? ? ? ? ? PX-only 2CSK (NMR, apo) 2ETT (NMR, apo) Comments The SNX25 PX domain has conservative alterations in two of the four canonical binding residues (Tyr/Phe to Leu at inositol-binding site, and Arg to Lys at 4-phosphate-binding site). It remains to be seen if these substitutions can support PtdIns binding HS1BP3 has a conservative substitution (Arg to Lys in P3-binding site). It remains to be seen if such a substitution can support binding to PtdIns3P Reported phosphoinositide preferences PI3P ≈ PI(3,4)P 2 > PI(4,5)P 2 PI3P ≈ PI(3,4)P2 PI4P ≈ PI5P ≈ PI(3,5)P 2 > PI(3,4)P 2 PI3P PI3P PI(3,4)P 2 > PI3 P > PI(3,5)P 2 PI3P PI3P PI3P PI3P PI3P PI3P PI3P PI3P ? PI3P n.d. PI3P ≈ PI5P > PI(3,5)P 2 n.d. n.d. n.d. PI3P PI3P PI3P PI3P PI3P PI3P PI3P ? PI3P n.d. n.d. n.d. PI3P n.d. n.d. R. D. Teasdale and B. M. Collins c The Authors Journal compilation c 2012 Biochemical Society Table S1 Table S1 Continued Tyrinositol (aromatic stacking with inositol ring†) LysP1 (binds to 1phosphate‡) R55 R53 R44 Y56 Y54 Y45 K79 K83 K69 R92 R99 R84 S99 Y100 – – S172 T1462 Y173 F1463 R200 R1488 – R1503 ? Polyproline loop is open T1405 S1237 F1406 F1238 R1431 W1263 R1446 R1275 ? Polyproline loop occludes 5-phosphate PLD1 K119 F120 R165 R180 ? PLD2 K103 Y104 R149 K164 ? Protein PX-S/T-kinase PXK RPK118 SGK3 (CISK) PX-SH3-GAP SNX26 (TCGAP) PX-PI3K PX-RICS (GCGAP) PI3K-C2α PI3K-C2β PI3K-C2γ PDB code (method, ligand) 1XTN (X-ray, SO4 ), 1XTE (X-ray, apo) 2IWL (X-ray, SO4 ), 2AR5 (X-ray, apo), 2REA (X-ray, apo), 2RED (X-ray, apo) 2WWE (X-ray, apo) Comments Structurally predicted phosphoinositide specificity¶ PI3P PI3P PI3P ? ? Polyproline loop occludes 5-phosphate ? c The Authors Journal compilation c 2012 Biochemical Society None None PI3P PI3P PI3P > PI(3,5)P 2 ≈ PI(4,5)P 2 ≈ PI(3,4,5)P 3 PI(4,5)P 2 ≈ PI(3,4)P 2 > PI3P ≈ PI4P PI3P ≈ PI4P ≈ PI5P PI(4,5)P 2 None None n.d. n.d. PI3P ? PI(3,4,5)P 3 PI3P > PI5P > PI(4,5)P 2 > PI4P > PI(3,5)P 2 > PI(3,4)P 2 PI3P ? n.d. None D1464 occludes the normal 1-phosphate-binding site. No density is observed in the structure for R1488 or the polyproline loop. T1462 in the ArgP3 position will preclude 3-phosphate interaction E1239 occludes the normal 1-phosphate-binding site. W1263 will preclude electrostatic contact with the 1-phosphate. S1237 in the ArgP3 position will preclude 3-phosphate interaction The PLD1 PX domain has conservative alterations in two of the four canonical binding residues (Arg to Lys at 3-phosphate-binding site, and Lys to Arg at 1-phosphate-binding site) The PLD2 PX domain has conservative alterations in three of the four canonical binding residues (Arg to Lys at 3-phosphate-binding site, Lys to Arg at 1-phosphate-binding site and Arg to Lys at the 4-hydroxy-binding site) Reported phosphoinositide preferences The PX protein family ArgP3 (binds to 3-phosphate*) Subfamily PX-PH-PLD Polyproline loop (potentially clashes with 5-phosphate groups) Arg4,5-hydroxy (binds to 4- and 5-hydroxy groups of inositol ring§) Continued Subfamily Protein PX-PXB SNX20 Kinesin-PX SNX21 Kif16B PX-MIT SNX15 PX-LRR-IRAS IRAS PX-SNX16 SNX29-PX PX-SNX34 Yeast PX protein SNX16 SNX29 SNX34 Snx3p Polyproline loop (potentially clashes with 5-phosphate groups) ArgP3 (binds to 3-phosphate*) Tyrinositol (aromatic stacking with inositol ring†) LysP1 (binds to 1-phosphate‡) Arg4,5-hydroxy (binds to 4- and 5-hydroxy groups of inositol ring§) R116 Y117 K143 R158 ? R171 R1220 Y172 Y1221 K198 K1246 R212 R1260 R51 Y52 R81 R97 ? Polyproline loop occludes 5-phosphate ? 3P0C (X-ray, apo) R49 Y50 K75 R88 1OCU (X-ray, PI3P) R144 R309 R55 R81 Y145 Y310 S56 Y82 K168 K336 R84 K112 R184 R350 R99 R127 PDB code (method, ligand) 2V14 (X-ray, apo) Polyproline loop and loop between strands β1 and β2 occlude the binding site ? ? ? Polyproline loop occludes 5-phosphate Comments K1246 will need to reorient to bind the 1-phosphate SNX15 has a conservative substitution (Lys to Arg in 1-phosphate-binding site) The occluding loops will need to alter their conformations to accommodate binding Structurally predicted phosphoinositide specificity¶ Reported phosphoinositide preferences PI3P PI3P ? PI4P ≈ PI5P ≈ PI(3,5)P 2 ≈ PI(3,4)P 2 n.d. PI3P > PI(3,4)P 2 > PI(3,4,5)P 3 > PI(3,5)P 2 n.d. PI3P PI3P PI3P PI3P PI3P ? PI3P PI3P n.d. n.d. PI3P PI3P PI3P *The Arg side chain makes specific electrostatic contact with the 3-phosphate group and is therefore required for binding of 3-phosphorylated PtdIns. †The aromatic groups of Tyr or Phe make stacking interations with the inositol ring and are therefore required for binding of PtdIns. ‡The lysine side chain at the end of the polyproline loop is involved in electrostatic contact with the 1-phosphate group and is thus important for PtdIns orientation and binding. §The Arg side chain makes specific H-bonds to the 4- and 5-hydroxy groups of PtdIns3P and therefore precludes binding of 4- and 5-phosphorylated PtdIns. The polyproline loop generally orients in such a way as to sterically preclude binding of 5-phosphates. However this loop is relatively flexible and therefore might accommodate binding via conformational changes. For all proteins without a known structure we have used ‘?’ to indicate that their orientation is unknown. ¶The phosphoinositide preferences are based on a critical evaluation of binding experiments reported in the literature. R. D. Teasdale and B. M. Collins c The Authors Journal compilation c 2012 Biochemical Society Table S1 The PX protein family Table S2 Identified interacting molecules for the mammalian PX protein subfamilies Interactions between PX proteins are in bold. Ligands have been classified into functional families: PX protein, transmembrane cargo, trafficking, cytoplasmic signalling protein, cytoskeleton and other. Transmembrane signalling receptors (e.g. EGFR) have been designated as transmembrane cargo for the purposes of this Table. PX sub-family PX protein Ligand Ligand class Domain from PX protein Domain from ligand protein Reference(s) PX-BAR SNX1 SNX1 SNX2 SNX5 SNX6 Tyrosine kinase receptors (EGFR, IR, PDGFR) Type I and II TGF-β receptors Transferrin receptor Dopamine receptors (D1, D5) GPCRs (M1, NK1, NK2, DOP, US28, GLP) P-selectin Hrs Retromer Clathrin heavy chain (CHC17) PX protein PX protein PX protein PX protein Cargo Cargo Cargo Cargo Cargo Cargo Trafficking Trafficking Trafficking – – BAR domain – Cytoplasmic domain Intracellular domain – C-terminal tails C-terminal tails – C-terminus excluding the VHS domain VPS35 and VPS29 subunits – [3–5] [6–8] [5,8–10] [5,8,11,12] [6,13] [11] [6] [14] [15] [16] [17,18] [3,5,7,19,20] [21] Enterophilin Rab6IP1 RME-8 J-domain protein Trafficking Trafficking Trafficking – C-terminus IWN3 and ARM-like domains [22] [5] [18,23] Arf6-interacting protein 1 Ypt Interacting protein 1 Ras-like protein (RLP) Trafficking Trafficking Signalling – – GTPase domain [5] [5] [24] Kalirin-7 (RhoGEF) WASH complex Rho GTPase (inactive form) SNX2 SNX1 SNX5 SNX6 Tyrosine kinase receptors (EGFR, IR, PDGFR) Type I and II TGF-β receptors TPR/MET tyrosine kinase Retromer Clathrin heavy chain (CHC17) Cytoskeleton Cytoskeleton Cytoskeleton PX protein PX protein PX protein PX protein Cargo Cargo Cargo Trafficking Trafficking – Strumpellin subunit of WASH complex – – – – – Cytoplasmic domain Intracellular domain – VPS35 and VPS29 subunits – [25] [26,27] [25] [3,6,7] [6–8] [5,8] [5,8,11] [6] [11] [28] [3,5,7] [21] Enterophilin Archain Kalirin7 (RhoGEF) WASH complex Rho GTPase (inactive form) Formin-binding protein 17 (FBP17) Zinc finger CDGSH Abstrakt (DEAD-box helicase) SNX4 SNX2 SNX6 Tyrosine kinase receptors (EGFR, PDGFR) Type I and II TGF-β receptors CFTR Clathrin heavy chain (CHC17) Trafficking Trafficking Cytoskeleton Cytoskeleton Cytoskeleton Cytoskeleton Other Other PX protein PX protein PX protein Cargo Cargo Cargo Trafficking – – – – – – Residues 1–162 – – – Cytoplasmic domains Intracellular domain – – [22] [5] [25] [26] [25] [29] [5] [30] [31] [6] [11] [6] [11] [32] [21] Amphiphysin 2 KIBRA (Dynein interactor) Dynein Trafficking Cytoskeleton Cytoskeleton Residues 1–304 (BAR domain) C-terminal domain – [33] [31] [21,31] SNX1 SNX2 SNX6 Retromer Clathrin heavy chain (CHC22) Dynactin DOCK180 Stathmin-2 CREB bZIP transcription factor CSE1 Fanconi anemia proteins (FANCs) Mind bomb 1 (Mib1) PX protein PX protein PX protein Trafficking Trafficking Cytoskeleton Cytoskeleton Cytoskeleton Other Other Other Other – – BAR domain – BAR domain PX domain – – – – BAR domain N-terminus Inverted clathrin box within PX domain (DFLGL) BAR domain – C-terminal segment of BAR domain – – Linker between PX and BAR domain – – BAR domain – – – – – PX domain PX + BAR domains – Inverted clathrin box within PX domain (DFLGL) BAR domain – – – BAR domain – – PX domain – – – – PX domain – Inverted clathrin box within PX domain (EFELL) Helix 3 of BAR domain BAR domain Inverted clathrin box within PX domain (EFELL) BAR domain – – – Helix 2 of BAR domain – Helix 1 of BAR domain – – – BAR domain – BAR domain – – – Hub domain (residues 1074–1640) C-terminus of p150glued (DCTN1) DHR1 domain – – – – – [8–10] [5,8] [5,8] [5,20] [34] [5] [35] [5] [5] [5] [36,37] [38] SNX2 SNX4 SNX5 c The Authors Journal compilation c 2012 Biochemical Society R. D. Teasdale and B. M. Collins Table S2 Continued PX sub-family PX protein Ligand Ligand class Domain from PX protein Domain from ligand protein Reference(s) SNX6 SNX6 SNX1 SNX2 SNX4 SNX5 Tyrosine kinase receptors (EGFR, IR, PDGFR, LR) Type I and II TGF-β receptors BACE-1 Retromer Arf6 Interacting protein 5 GIT1 Dynactin PX protein PX protein PX protein PX protein PX protein Cargo – – – – – – – – – – – – [11] [8,11,12] [5,8,11] [11] [5,8] [11] Cargo Cargo Trafficking Trafficking Signalling Cytoskeleton PX domain PX domain – – BAR domain Residues 71–140 [11,39] [40] [5] [5] [41] [5,8] NudE-like 1 MLLT10 CREB bZIP transcription factor Pim1 kinase Translationally controlled tumour protein (TCTP) p27Kip1 BRMS-1 Cytoskeleton Other Other Other Other – – – PX domain – Intracellular domains – – – Coiled-coil domain 2 C-terminus of p150glued (DCTN1); residues 1061–1282 – – – – – Other Other Residues 300–406 [43] [44] USP16 PLCγ 1 Other Signalling – Polyproline loop of PX domain (residues 90–103; TLIIPPLPEKFIVK) Proline-rich domain Residues 1–88 (coiled-coil domain) – SH3 domain – – – SNX9 SNX18 SNX33 Metalloprotease disintegrins MDC9 and MDC15 (ADAM9 and ADAM15) Insulin receptor AP2 complex PX protein PX protein PX protein Cargo BAR domain BAR domain – SH3 domain [47–50] [51] [52] [53] Cargo Trafficking Clathrin Trafficking Dynamin 1 and dynamin 2 Synaptojanin-1 ITCH ubiquitin ligase Son of sevenless 1 (Sos1) and Sos2 PI3K p85 PtdIns(4)P -5-kinases Iα, Iβ and Iγ Cdc-associated kinase 1 (ACK1) and ACK2 WASP and N-WASP Arp2/3 complex Trafficking Trafficking Trafficking Signalling Signalling Signalling Signalling Cytoskeleton Cytoskeleton Proline-rich domain Proline-rich domain Proline-rich domain Proline-rich domain – – Proline-rich domain Proline-rich domain – [50,56–59] [60,61] [62] [63] [64] [59] [60,65] [50,59,64,66] [59] Aldolase Other Near aldolase active site [57,67] USP7 EspF USP25 PARP11 TL132L SNX18 SNX9 Dynamin2 AP1 complex Other Other Other Other Other PX protein PX protein Trafficking Trafficking Trafficking Trafficking Signalling Cytoskeleton – Proline-rich domain – – – BAR domain BAR domain Proline-rich domain γ 1 subunit appendage domains Proline-rich domain Proline-rich domain Proline-rich domain Proline-rich domain [45] [68,69] [45] [45] [45] [51,70] [51] [51,71] [71] Synaptojanin-1 ITCH ubiquitin ligase Son of sevenless 1 (Sos1) and Sos2 WASP and N-WASP – Low complexity region between SH3 and PX domains Low complexity region between SH3 and PX domains SH3 domain SH3 domain SH3 domain SH3 domain PX domain PX domain SH3 domain SH3 domain Low complexity region between SH3 and PX domains Low complexity region between SH3 and PX domains. Minimal sequence WDEDWDG – SH3 domain – – – BAR domain BAR domain SH3 domain Low complexity region between SH3 and PX domains SH3 domain SH3 domain SH3 domain SH3 domain BAR domain BAR domain – PARPAPAPP polyproline sequence – α and β2 subunits appendage domains – SNX7 SH3-PX-BAR SNX8 SNX30 SNX32 SNX9 SNX18 c The Authors Journal compilation c 2012 Biochemical Society [5] [5] [5] [42] [12] [44] [46] [54] [55,56] [56] [51] [62] [63] [51,72] The PX protein family Table S2 Continued PX sub-family PX-SH3 PX protein Ligand Ligand class Domain from PX protein Domain from ligand protein Reference(s) SNX33 SNX33 SNX9 ADAM15 Dynamin 1 and dynamin 2 WASP ADAM proteases (ADAM12, ADAM15, ADAM19) PX protein PX protein Cargo Trafficking Cytoskeleton Cargo – – SH3 domain SH3 domain SH3 domain SH3 domain (number 5) – – Proline-rich domain Proline-rich domain Proline-rich domain C-terminal cytoplasmic tail polyproline sequences [52] [52] [73] [71,74] [52] [75,76] Dynamin 2 Grb2 Sos1 Nck1 and Nck2 WIP N-WASP Dystroglycan – p22phox Trafficking Signalling Signalling Signalling Cytoskeleton Cytoskeleton Other SH3 domains (1,2 and 5) Polyproline sequences SH3 domains (1–2) Src-phosphorylated Tyr557 SH3 domains (3 and 5) SH3 domains (1-5) SH3 domain (number 3) Polyproline N-terminal SH3 domain Polyproline SH2 domain – – Polyproline [77,78] [77] [78] [79] [77] [77] [80] Other Tandem SH3 domains Polyproline sequence [81–83] p67phox NOX1 NOXA1 p47phox Other Other Other PX protein Polyproline sequence – Polyproline sequence SH3 domain SH3 domain NADPH-binding domain SH3 domain Polyproline sequence [81] [84] [83,85,86] [87–95] Ku70 Signalling N- and C-terminal regions [96] Coronin Moesin p67phox Thioredoxin p47phox Cytoskeleton Cytoskeleton Other Other PX protein p40phox cPLA2 RelA Akt kinase Cortactin Actin Moesin p67phox p22phox Fyn tyrosine kinase CrkII Rho GTPases (Cdc42, TC10β) TrkA receptor tyrosine kinase PX protein Signalling Signalling Signalling Cytoskeleton Cytoskeleton Cytoskeleton Other Other Signalling Signalling Cytoskeleton Cargo C-terminus PX domain C-terminal PB1 domain – PX domain polyproline loop Polyproline sequence PX domain SH3 domain C-terminal regions (last 186 residues) – FERM domain PB1 domain – SH3 domain – Residues 319–337 PX domain Polyproline sequence SH3 domain Polyproline sequences Polyproline sequences GAP domain C-terminal domain – – FERM domain C-terminal SH3 domain Polyproline sequence SH3 domain SH3 domain (N-terminal) GTPase fold Cytoplasmic domain NR2B (GluRε2) NMDA receptor Gab1, Gab2 PSD-95 Crk and CrkL Nck p130CAS p120 Ras GAP 14-3-3 proteins GABARAP and GABARAPL1 N-Shc β-Catenin N-Cadherin Dynein/dynactin complex P-selectin Cargo Signalling Signalling Signalling Signalling Signalling Signalling Signalling Signalling Signalling Cytoskeleton Cytoskeleton Cytoskeleton Cargo C-terminal domain C-terminal domain – Polyproline sequences Polyproline sequences C-terminal domain Polyproline sequences RSKSDP sequence (Ser1796 ) – C-terminal domain C-terminal domain – – FERM domain Cytoplasmic domain – – SH3 domain SH3 domain – SH3 domain LRP1 LDLR VLDLR ApoER2 (LRP8) APP FEEL-1/stabilin-1 Cargo Cargo Cargo Cargo Cargo Cargo FERM domain FERM domain FERM domain FERM domain FERM domain FERM domain (F3 module) SH3PXD2A (SH3MD1, FISH, Tks5) SH3PXD2B (Tks4) SNX28 (NOXO1, p41NOX, SH3PXD5) p40phox (NCF4, SH3PXD4) p47phox (NCF1, SH3PXD1A) PX-SH3-RhoGAP SNX26 (TC-GAP) PX-RICS (GC-GAP, p250GAP, p200GAP, Grit) PX-FERM SNX17 SH3 domain C2 domain Polyproline sequence – SH2 domain – – – FTNAAFDPSP peptide motif IGNPTY peptide motif FDNPVY peptide motif FDNPVY peptide motif FDNPVY peptide motif YENPTY peptide motif NPVF peptide motif [97] [98,99] [87,88,90,91,94,100–103] [104] [105] [87–95] [106] [107] [108] [109] [110] [98,99] [89–92,95,111–116] [92,115,117,118] [119] [120] [120] [121] [122–124] [125] [122,123] [121,125] [125] [121,125] [126] [127] [128] [121] [122,123] [122,123] [127] [16,129,130] [131–134] [133,135] [133] [133] [136,137] [138] c The Authors Journal compilation c 2012 Biochemical Society R. D. Teasdale and B. M. Collins Table S2 Continued PX sub-family PX protein SNX27 (Mrt1) SNX31 Ligand Ligand class Domain from PX protein Domain from ligand protein Reference(s) PTCH1 H-Ras Krit1 Abl1 Cargo Signalling Signalling Signalling – – – SH3 domain [130] [137] [139] [46] Crk Signalling SH3 domain [46] Src Signalling SH3 domain [46] Fyn Signalling SH3 domain [46] Grb2 Signalling SH3 domain [46] Nck1 Signalling SH3 domain [46] p85A Signalling SH3 domain [46] PLCγ 1 Signalling SH3 domain [46] KIF1Bβ Cytoskeleton FERM domain FERM domain FERM domain Polyproline loop of PX domain (residues 55–66; ANVLPAFPPKKLFS) Polyproline loop of PX domain (residues 55–66; ANVLPAFPPKKLFS) Polyproline loop of PX domain (residues 55–66; ANVLPAFPPKKLFS) Polyproline loop of PX domain (residues 55–66; ANVLPAFPPKKLFS) Polyproline loop of PX domain (residues 55–66; ANVLPAFPPKKLFS) Polyproline loop of PX domain (residues 55–66; ANVLPAFPPKKLFS) Polyproline loop of PX domain (residues 55–66; ANVLPAFPPKKLFS) Polyproline loop of PX domain (residues 55–66; ANVLPAFPPKKLFS) FERM domain (F3 module) [140] 5-HT4 R Cargo PDZ domain Kir3.3/GIRK3 Cargo PDZ domain NMDA receptor 2C (NR2C) Cargo PDZ domain Very large GPCR Cargo PDZ domain β 2 -Adrenergic receptor Cargo PDZ domain APP CASP Cargo Trafficking FERM domain PDZ domain DGKζ Signalling PDZ domain Connector enhancer of kinase suppressor of Ras 2 (CNKSR2) Ribosomal protein S6 kinase, 90 kDa, polypeptide 3 (RPS6KA3) Protein tyrosine phosphatase, non-receptor type (PTPN2) H-Ras Dynactin 1 Trichoplein, keratin filament-binding protein (TCHP) Microtubule-associated protein 1A (MAP1A) Rho GDP dissociation inhibitor (GDI) α WASH complex Liver mitochondrial glutaminase (GA) E2a-Pbx1-associated protein (EB-1) ATPase, Na + /K + transporting, β2 polypeptide (ATP1B2) Component 1, q subcomponent-binding protein (C1QBP) RAN-binding protein 9 G3BP1 H-Ras Signalling PDZ domain Signalling PDZ domain Signalling – C-terminal residues 813–916 ESCF; C-terminal Type I PDZ binding motif ESESKV; C-terminal Type I PDZ binding motif ESEV; C-terminal Type I PDZ binding motif ETHL; C-terminal Type I PDZ binding motif DSLL; C-terminal Type I PDZ binding motif YENPTY peptide motif ESRF; C-terminal Type I PDZ binding motif ETAV; C-terminal Type I PDZ binding motif ETHV; C-terminal Type I PDZ binding motif ETAL; C-terminal Type I PDZ binding motif – Signalling Cytoskeleton Cytoskeleton FERM domain – – – – – [137] [145] [145] Cytoskeleton – – [145] Cytoskeleton – – [145] Cytoskeleton Other – PDZ domain [151] [145] Other Other – – – ESMV; C-terminal Type I PDZ binding motif – – Other – – [145] Other Other Signalling – – FERM domain – – – [145] [45] [137] c The Authors Journal compilation c 2012 Biochemical Society [141] [142–144] [145] [145] [146] [137] [147,148] [145,149,150] [145] [145] [145] [145] [145] The PX protein family Table S2 Continued PX sub-family PX protein Ligand Ligand class Domain from PX protein Domain from ligand protein Reference(s) PXA-RGS-PX-PXC SNX13 Hrs Gα s – IA-2 (islet antigen-2) Trafficking Signalling PX + PXC domains RGS domain – – [152] [153] Other PX domain (residues 494–676) [154] SNX25 PXK (MONaKA) TGF-β receptor I Na,K-ATPase subunits β1 and β3 β-Actin Cargo Cargo Cytoskeleton [39] [155] [156] RPS6KC1 (RPK118, humS6PKh1) Sphingosine kinase (SPHK) Signalling PXA and PX domains – WASP-homology domain (residues 531–578) PSK2 domain Residues 744–979 (C-terminal protein tyrosine phosphatase domain) – – – – [157] Peroxiredoxin-3 AIP4 E3 ubiquitin ligase Other Trafficking – WW domains [158] [159] GSK-3β Clathrin Signalling Trafficking – Clathrin terminal domain [160] [161,162] Dynactin subunits (p50 and p150) EGFR Cytoskeleton Cargo Pseudo-kinase domain Kinase domain (possible PPFY motif) – N-terminal clathrin-binding sequence – N-terminal domain – VENPE(pY)L sequence [163] [164] Clathrin Trafficking Clathrin terminal domain [165] Shc Eps8 Rac GEF complex (Eps8–Abi1–Sos1) Grb2 Signalling Signalling – Eps8 SH3 domain [166] [166] SH3 domains [166,167] – [168] SNX14 SNX19 PX-S/T kinase SGK3 (CISK, SGKL) PI3K-PX PIK3C2A (PI3K-C2α) PIK3C2B (PI3K-C2β) PX-PH-PLD PIK3C2G (PI3K-C2γ ) PLD1 PLD2 Tyrosine phosphorylated proteins (p70 and p110) – Other N-terminal clathrin-binding sequence – N-terminal polyproline sequences N-terminal polyproline sequences – PLD1 PLD2 EGFR APP (Amyloid precursor protein) Presenilin 1 Arf1 RalA Secretory carrier membrane protein (SCAMP)2 Caveolin1 and Caveolin 3 CtBP1/BARS Munc18-1 Dynamin Amphyphysin I and II AP180 PKCα PLC-γ 1 Type Iα PIP kinase PEA-15 CKII c-Src tyrosine kinase Cdc42 RhoA Actin α-Synuclein PLD1 PLD2 EGFR Munc18-1 Dynamin Caveolin 1 Amphyphysin I and II Type Iα PIP kinase mTor/Raptor complex Syk kinase PLC-γ 1 PX protein PX protein Cargo Cargo Cargo Trafficking Trafficking Trafficking – – – PH domain – – – – – – – Cytoplasmic domain Intracellular loop – – – [169] [169] [170] [171] [172] [173] [174,175] [176] Trafficking Trafficking Trafficking Trafficking Trafficking Trafficking Signalling Signalling Signalling Signalling Signalling Signalling Signalling Cytoskeleton Cytoskeleton Other PX protein PX protein Cargo Trafficking Trafficking Trafficking Trafficking Signalling Signalling Signalling Signalling – – PX domain PX domain – C-terminal residues 820–1036 C-terminal domain PX domain – C-terminus – PH domain – C-terminal domain – PX + PH domains – – – PX domain PX domain – – – TOS motif in PH domain PX domain PX domain Scaffolding domain – – GTPase domain N-terminal domain Residues 312–314 (TSP) C-terminal peptide SH3 domain N-terminal domain Death-effector domain – Kinase domain – – – N-terminal domain – – – – GTPase domain – N-terminal domain N-terminal domain Raptor WD40 domain – SH3 domain [171,177] [178] [179] [180] [181] [182] [177,183–186] [187] [188] [189] [190] [191] [192] [189,192–194] [195] [196] [169] [169] [170] [179] [180,197] [198] [181] [188] [199] [200] [187] Signalling c The Authors Journal compilation c 2012 Biochemical Society R. D. Teasdale and B. M. Collins Table S2 Continued PX sub-family PX-PXB PX-only PX protein SNX20 SNX21 SNX3 SNX10 SNX11 SNX12 SNX22 SNX24 HS1BP3 Kinesin-PX PX-MIT SNX23 (KIF16B) SNX15 PX-LRR-IRAS IRAS (nischarin) PX-SNX16 SNX29-PX PX-SNX34 SNX16 SNX29 SNX34 (C6ORF145) Ligand Ligand class Domain from PX protein Domain from ligand protein Reference(s) PEA-15 c-Src tyrosine kinase PTP1B PKC-ζ Signalling Signalling Signalling Signalling C-terminus PH domain – PX domain (lysines 101–103) [189] [191] [201] [202] Grb2 Signalling Rac2 Cytoskeleton Small GTPase domain [205,206] β-Actin RhoA Cytoskeleton Cytoskeleton YLNR and YRNY phosphotyrosine motifs in PX domain PH domain (including two CRIB sequence motifs) Residues 613–723 PX domain Death-effector domain Kinase domain – Kinase domain (residues 348–370 required) SH2 domain [207] [208] Aldolase α-Synuclein PSGL-1 – ENaC Clathrin heavy chain (CHC17) Other Other Cargo PH domain PX + PH domains PX domain – Small GTPase domain nucleotide free – N-terminal domain C-terminal cytoplasmic tail [209] [196] [210] Cargo Trafficking – – [211] [21] Trafficking Other – Inverted clathrin box within PX domain (DFEWL) – – – – [20] [211] Signalling – SH3 domain [212] PX protein PX protein PX protein PX protein Cargo PX protein Cargo PX domain PX domain PX domain PX domain PX domain C-terminal region Residues 710–810 (human numbering) [213] [213] [213] [213] [213] [214] [214–216] Signalling C-terminus PX domain – – – – C-terminal region Cytoplasmic domain peptide IYILYKLGFFKRSL C-terminus Signalling Signalling Cytoskeleton PX protein N-terminus N-terminus N-terminus C-terminal coiled-coil Kinase domain PDZ and kinase domains – C-terminal coiled-coil [218] [219] [220] [221,222] Retromer Usp10 – – – – – Haemopoietic cell-specific Lyn substrate 1 (HS1) – SNX15 SNX1 SNX2 SNX4 PDGFR IRAS α5 Integrin Insulin receptor substrates (IRS-4, IRS-1, IRS-2, IRS-3) PAK1, PAK4, PAK5 kinases LIM kinase Rac1 SNX16 – – c The Authors Journal compilation c 2012 Biochemical Society [201,203–205] [217] The PX protein family Table S3 Yeast PX proteins and their closest mammalian homologues PX family Yeast protein Yeast name Aliases Closest human homologue* PX-BAR YOR069W YOR132W YMR004W YJL036W YDR425W YDL113C YKR078W YML104C YHR105W YOR357C YKR031C YPL115C GRD2, PEP10, VPT5 PEP21, VPT3 SNX8 CVT13, ATG24 SNX1 None PX-PH-PLD PX-PH-GAP Vps5p Vps17p Mvp1p Snx4p Snx41p Snx42p YKR078Wp Mdm1p Ypt35p Snx3p Pld1p Bem3p PX-SNARE Vam7p SH3-PX-CAD PX-unknown PXA -RGS-PX-PXA PX-only GRD19 SPO14 YJL036W SNX30 SNX4 SNX4 None SNX13 SNX13, SNX11 SNX3 PLD2 None YGL212W VPS43, VPL24 KIF16B Bem1p YBR200W SRO1 SH3PXD2B YPR097Wp YPR097W CVT20, ATG20 None Notes Has extended loop between helices H2 and H3 of BAR domain Has extended loop between helices H2 and H3 of BAR domain Has a large insert between helices α1 and α2 of the PX domain Mammalian PX-SH3-GAP proteins have an SH3 domain, not a PH domain. They have extended regions C-terminal to the PX and GAP domains, whereas the yeast proteins have extended regions N-terminal to the PX and GAP domains The Vam7p SNARE is a clear invention of fungi, on the basis of detailed phylogenetic analysis [223]. Has two N-terminal SH3 domains upstream of the PX domain and a CAD/PB1 domain C-terminal to the PX domain. The CAD/PB1 domain is from the ubiquitin superfamily The PX domain of YPR097Wp is highly degenerate, with a predicted insert of ∼130 amino acids between α1 and α2. YPR097Wp is large (1073 residues) and overall is predicted to be highly α-helical in structure. However, there are no other known domains identified outside the PX domain *The closest human PX protein homologue is listed, although this may not necessarily mean the proteins are orthologues. Where ‘none’ is stated this means that, on default settings, BLAST searches did not retrieve any known PX domain proteins encoded in the human genome. REFERENCES 1 Cole, C., Barber, J. D. and Barton, G. J. (2008) The Jpred 3 secondary structure prediction server. Nucleic Acids Res. 36, W197–W201 2 Gouet, P., Courcelle, E., Stuart, D. I. and Metoz, F. (1999) ESPript: analysis of multiple sequence alignments in PostScript. Bioinformatics 15, 305–308 3 Haft, C. R., de la Luz Sierra, M., Bafford, R., Lesniak, M. A., Barr, V. A. and Taylor, S. I. (2000) Human orthologs of yeast vacuolar protein sorting proteins Vps26, 29, and 35: assembly into multimeric complexes. Mol. Biol. Cell 11, 4105–4116 4 Kurten, R. C., Eddington, A. D., Chowdhury, P., Smith, R. D., Davidson, A. D. and Shank, B. B. (2001) Self-assembly and binding of a sorting nexin to sorting endosomes. J. Cell Sci. 114, 1743–1756 5 Wassmer, T., Attar, N., Harterink, M., Weering, J. R., Traer, C. J., Oakley, J., Goud, B., Stephens, D. J., Korswagen, H. C. and Cullen, P. J. (2009) The retromer coat complex coordinates endosomal sorting and dynein-mediated transport, with carrier recognition by the trans -Golgi network. Dev. Cell 17, 110–122 6 Haft, C. R., de la Luz Sierra, M., Barr, V. A., Haft, D. H. and Taylor, S. I. (1998) Identification of a family of sorting nexin molecules and characterization of their association with receptors. Mol. Cell. Biol. 18, 7278–7287 7 Rojas, R., Kametaka, S., Haft, C. R. and Bonifacino, J. S. (2007) Interchangeable but essential functions of SNX1 and SNX2 in the association of retromer with endosomes and the trafficking of mannose 6-phosphate receptors. Mol. Cell. Biol. 27, 1112–1124 8 Hong, Z., Yang, Y., Zhang, C., Niu, Y., Li, K., Zhao, X. and Liu, J. J. (2009) The retromer component SNX6 interacts with dynactin p150Glued and mediates endosome-to-TGN transport. Cell Res. 19, 1334–1349 9 Kerr, M. C., Lindsay, M. R., Luetterforst, R., Hamilton, N., Simpson, F., Parton, R. G., Gleeson, P. A. and Teasdale, R. D. (2006) Visualisation of macropinosome maturation by the recruitment of sorting nexins. J. Cell Sci. 119, 3967–3980 10 Liu, H., Liu, Z. Q., Chen, C. X., Magill, S., Jiang, Y. and Liu, Y. J. (2006) Inhibitory regulation of EGF receptor degradation by sorting nexin 5. Biochem. Biophys. Res. Commun. 342, 537–546 11 Parks, W. T., Frank, D. B., Huff, C., Renfrew Haft, C., Martin, J., Meng, X., de Caestecker, M. P., McNally, J. G., Reddi, A., Taylor, S. I. et al. (2001) Sorting nexin 6, a novel SNX, interacts with the transforming growth factor-β family of receptor serine-threonine kinases. J. Biol. Chem. 276, 19332–19339 12 Yoon, T., Kim, M. and Lee, K. (2006) Inhibition of Na,K-ATPase-suppressive activity of translationally controlled tumor protein by sorting nexin 6. FEBS Lett. 580, 3558–3564 13 Kurten, R. C., Cadena, D. L. and Gill, G. N. (1996) Enhanced degradation of EGF receptors by a sorting nexin, SNX1. Science 272, 1008–1010 14 Heydorn, A., Sondergaard, B. P., Hadrup, N., Holst, B., Haft, C. R. and Schwartz, T. W. (2004) Distinct in vitro interaction pattern of dopamine receptor subtypes with adaptor proteins involved in post-endocytotic receptor targeting. FEBS Lett. 556, 276–280 15 Heydorn, A., Sondergaard, B. P., Ersboll, B., Holst, B., Nielsen, F. C., Haft, C. R., Whistler, J. and Schwartz, T. W. (2004) A library of 7TM receptor C-terminal tails. Interactions with the proposed post-endocytic sorting proteins ERM-binding phosphoprotein 50 (EBP50), N -ethylmaleimide-sensitive factor (NSF), sorting nexin 1 (SNX1), and G protein-coupled receptor-associated sorting protein (GASP). J. Biol. Chem. 279, 54291–54303 16 Williams, R., Schluter, T., Roberts, M. S., Knauth, P., Bohnensack, R. and Cutler, D. F. (2004) Sorting nexin 17 accelerates internalization yet retards degradation of P-selectin. Mol. Biol. Cell 15, 3095–3105 17 Chin, L. S., Raynor, M. C., Wei, X., Chen, H. Q. and Li, L. (2001) Hrs interacts with sorting nexin 1 and regulates degradation of epidermal growth factor receptor. J. Biol. Chem. 276, 7069–7078 18 Popoff, V., Mardones, G. A., Bai, S. K., Chambon, V., Tenza, D., Burgos, P. V., Shi, A., Benaroch, P., Urbé, S., Lamaze, C. et al. (2009) Analysis of articulation between clathrin and retromer in retrograde sorting on early endosomes. Traffic 10, 1868–1880 19 Gullapalli, A., Garrett, T. A., Paing, M. M., Griffin, C. T., Yang, Y. and Trejo, J. (2004) A role for sorting nexin 2 in epidermal growth factor receptor down-regulation: evidence for distinct functions of sorting nexin 1 and 2 in protein trafficking. Mol. Biol. Cell 15, 2143–2155 20 Harterink, M., Port, F., Lorenowicz, M. J., McGough, I. J., Silhankova, M., Betist, M. C., van Weering, J. R., van Heesbeen, R. G., Middelkoop, T. C., Basler, K. et al. (2011) A SNX3-dependent retromer pathway mediates retrograde transport of the Wnt sorting receptor Wntless and is required for Wnt secretion. Nat. Cell Biol. 13, 914–923 21 Skanland, S. S., Walchli, S., Brech, A. and Sandvig, K. (2009) SNX4 in complex with clathrin and dynein: implications for endosome movement. PLoS ONE 4, e5935 22 Pons, V., Hullin-Matsuda, F., Nauze, M., Barbaras, R., Peres, C., Collet, X., Perret, B., Chap, H. and Gassama-Diagne, A. (2003) Enterophilin-1, a new partner of sorting nexin 1, decreases cell surface epidermal growth factor receptor. J. Biol. Chem. 278, 21155–21161 23 Shi, A., Sun, L., Banerjee, R., Tobin, M., Zhang, Y. and Grant, B. D. (2009) Regulation of endosomal clathrin and retromer-mediated endosome to Golgi retrograde transport by the J-domain protein RME-8. EMBO J. 28, 3290–3302 c The Authors Journal compilation c 2012 Biochemical Society R. D. Teasdale and B. M. Collins 24 Piek, E., Van Dinther, M., Parks, W. T., Sallee, J. M., Bottinger, E. P., Roberts, A. B. and Ten Dijke, P. (2004) RLP, a novel Ras-like protein, is an immediate-early transforming growth factor-β (TGF-β) target gene that negatively regulates transcriptional activity induced by TGF-β. Biochem. J. 383, 187–199 25 Prosser, D. C., Tran, D., Schooley, A., Wendland, B. and Ngsee, J. K. (2010) A novel, retromer-independent role for sorting nexins 1 and 2 in RhoG-dependent membrane remodeling. Traffic 11, 1347–1362 26 Gomez, T. S. and Billadeau, D. D. (2009) A FAM21-containing WASH complex regulates retromer-dependent sorting. Dev. Cell 17, 699–711 27 Harbour, M. E., Breusegem, S. Y., Antrobus, R., Freeman, C., Reid, E. and Seaman, M. N. (2010) The cargo-selective retromer complex is a recruiting hub for protein complexes that regulate endosomal tubule dynamics. J. Cell Sci. 123, 3703–3717 28 Schaaf, C. P., Benzing, J., Schmitt, T., Erz, D. H., Tewes, M., Bartram, C. R. and Janssen, J. W. (2005) Novel interaction partners of the TPR/MET tyrosine kinase. FASEB J. 19, 267–269 29 Fuchs, U., Rehkamp, G., Haas, O. A., Slany, R., Konig, M., Bojesen, S., Bohle, R. M., Damm-Welk, C., Ludwig, W. D., Harbott, J. and Borkhardt, A. (2001) The human formin-binding protein 17 (FBP17) interacts with sorting nexin, SNX2, and is an MLL-fusion partner in acute myelogeneous leukemia. Proc. Natl. Acad. Sci. U.S.A. 98, 8756–8761 30 Abdul-Ghani, M., Hartman, K. L. and Ngsee, J. K. (2005) Abstrakt interacts with and regulates the expression of sorting nexin-2. J. Cell. Physiol. 204, 210–218 31 Traer, C. J., Rutherford, A. C., Palmer, K. J., Wassmer, T., Oakley, J., Attar, N., Carlton, J. G., Kremerskothen, J., Stephens, D. J. and Cullen, P. J. (2007) SNX4 coordinates endosomal sorting of TfnR with dynein-mediated transport into the endocytic recycling compartment. Nat. Cell Biol. 9, 1370–1380 32 Wang, X., Venable, J., LaPointe, P., Hutt, D. M., Koulov, A. V., Coppinger, J., Gurkan, C., Kellner, W., Matteson, J., Plutner, H. et al. (2006) Hsp90 cochaperone Aha1 downregulation rescues misfolding of CFTR in cystic fibrosis. Cell 127, 803–815 33 Leprince, C., Le Scolan, E., Meunier, B., Fraisier, V., Brandon, N., De Gunzburg, J. and Camonis, J. (2003) Sorting nexin 4 and amphiphysin 2, a new partnership between endocytosis and intracellular trafficking. J. Cell Sci. 116, 1937–1948 34 Towler, M. C., Gleeson, P. A., Hoshino, S., Rahkila, P., Manalo, V., Ohkoshi, N., Ordahl, C., Parton, R. G. and Brodsky, F. M. (2004) Clathrin isoform CHC22, a component of neuromuscular and myotendinous junctions, binds sorting nexin 5 and has increased expression during myogenesis and muscle regeneration. Mol. Biol. Cell 15, 3181–3195 35 Hara, S., Kiyokawa, E., Iemura, S., Natsume, T., Wassmer, T., Cullen, P. J., Hiai, H. and Matsuda, M. (2008) The DHR1 domain of DOCK180 binds to SNX5 and regulates cation-independent mannose 6-phosphate receptor transport. Mol. Biol. Cell 19, 3823–3835 36 Otsuki, T., Kajigaya, S., Ozawa, K. and Liu, J. M. (1999) SNX5, a new member of the sorting nexin family, binds to the Fanconi anemia complementation group A protein. Biochem. Biophys. Res. Commun. 265, 630–635 37 Reuter, T. Y., Medhurst, A. L., Waisfisz, Q., Zhi, Y., Herterich, S., Hoehn, H., Gross, H. J., Joenje, H., Hoatlin, M. E., Mathew, C. G. and Huber, P. A. (2003) Yeast two-hybrid screens imply involvement of Fanconi anemia proteins in transcription regulation, cell signaling, oxidative metabolism, and cellular transport. Exp. Cell Res. 289, 211–221 38 Yoo, K. W., Kim, E. H., Jung, S. H., Rhee, M., Koo, B. K., Yoon, K. J., Kong, Y. Y. and Kim, C. H. (2006) Snx5, as a Mind bomb-binding protein, is expressed in hematopoietic and endothelial precursor cells in zebrafish. FEBS Lett. 580, 4409–4416 39 Hao, X., Wang, Y., Ren, F., Zhu, S., Ren, Y., Jia, B., Li, Y. P., Shi, Y. and Chang, Z. (2011) SNX25 regulates TGF-β signaling by enhancing the receptor degradation. Cell. Signal. 23, 935–946 40 Okada, H., Zhang, W., Peterhoff, C., Hwang, J. C., Nixon, R. A., Ryu, S. H. and Kim, T. W. (2010) Proteomic identification of sorting nexin 6 as a negative regulator of BACE1mediated APP processing. FASEB J. 24, 2783–2794 41 Cavet, M. E., Pang, J., Yin, G. and Berk, B. C. (2008) An epidermal growth factor (EGF)dependent interaction between GIT1 and sorting nexin 6 promotes degradation of the EGF receptor. FASEB J. 22, 3607–3616 42 Ishibashi, Y., Maita, H., Yano, M., Koike, N., Tamai, K., Ariga, H. and Iguchi-Ariga, S. M. (2001) Pim-1 translocates sorting nexin 6/TRAF4-associated factor 2 from cytoplasm to nucleus. FEBS Lett. 506, 33–38 43 Fuster, J. J., Gonzalez, J. M., Edo, M. D., Viana, R., Boya, P., Cervera, J., Verges, M., Rivera, J. and Andres, V. (2010) Tumor suppressor p27Kip1 undergoes endolysosomal degradation through its interaction with sorting nexin 6. FASEB J. 24, 2998–3009 44 Rivera, J., Megı́as, D. and Bravo, J. (2010) Sorting nexin 6 interacts with breast cancer metastasis suppressor-1 and promotes transcriptional repression. J. Cell. Biochem. 111, 1464–1472 45 Sowa, M. E., Bennett, E. J., Gygi, S. P. and Harper, J. W. (2009) Defining the human deubiquitinating enzyme interaction landscape. Cell 138, 389–403 46 Wu, C., Ma, M. H., Brown, K. R., Geisler, M., Li, L., Tzeng, E., Jia, C. Y., Jurisica, I. and Li, S. S. (2007) Systematic identification of SH3 domain-mediated human protein– protein interactions by peptide array target screening. Proteomics 7, 1775–1785 c The Authors Journal compilation c 2012 Biochemical Society 47 Pylypenko, O., Lundmark, R., Rasmuson, E., Carlsson, S. R. and Rak, A. (2007) The PX-BAR membrane-remodeling unit of sorting nexin 9. EMBO J. 26, 4788–4800 48 Wang, Q., Kaan, H. Y., Hooda, R. N., Goh, S. L. and Sondermann, H. (2008) Structure and plasticity of endophilin and sorting nexin 9. Structure 16, 1574–1587 49 Childress, C., Lin, Q. and Yang, W. (2006) Dimerization is required for SH3PX1 tyrosine phosphorylation in response to epidermal growth factor signalling and interaction with ACK2. Biochem. J. 394, 693–698 50 Shin, N., Lee, S., Ahn, N., Kim, S. A., Ahn, S. G., YongPark, Z. and Chang, S. (2007) Sorting nexin 9 interacts with dynamin 1 and N-WASP and coordinates synaptic vesicle endocytosis. J. Biol. Chem. 282, 28939–28950 51 Park, S., Kim, Y., Lee, S., Park, P., Park, Z., Sun, W., Kim, H. and Chang, S. (2010) SNX18 shares a redundant role with SNX9 and modulates endoctyic trafficking at the plasma membrane. J. Cell Sci. 123, 1742–1750 52 Zhang, J., Zhang, X., Guo, Y., Xu, L. and Pei, D. (2009) Sorting nexin 33 induces mammalian cell micronucleated phenotype and actin polymerization by interacting with Wiskott–Aldrich syndrome protein. J. Biol. Chem. 284, 21659–21669 53 Howard, L., Nelson, K. K., Maciewicz, R. A. and Blobel, C. P. (1999) Interaction of the metalloprotease disintegrins MDC9 and MDC15 with two SH3 domain-containing proteins, endophilin I and SH3PX1. J. Biol. Chem. 274, 31693–31699 54 Macaulay, S. L., Stoichevska, V., Grusovin, J., Gough, K. H., Castelli, L. A. and Ward, C. W. (2003) Insulin stimulates movement of sorting nexin 9 between cellular compartments: a putative role mediating cell surface receptor expression and insulin action. Biochem. J. 376, 123–134 55 Lundmark, R. and Carlsson, S. R. (2002) The β-appendages of the four adaptor-protein (AP) complexes: structure and binding properties, and identification of sorting nexin 9 as an accessory protein to AP-2. Biochem. J. 362, 597–607 56 Lundmark, R. and Carlsson, S. R. (2003) Sorting nexin 9 participates in clathrinmediated endocytosis through interactions with the core components. J. Biol. Chem. 278, 46772–46781 57 Lundmark, R. and Carlsson, S. R. (2004) Regulated membrane recruitment of dynamin-2 mediated by sorting nexin 9. J. Biol. Chem. 279, 42694–42702 58 Soulet, F., Yarar, D., Leonard, M. and Schmid, S. L. (2005) SNX9 regulates dynamin assembly and is required for efficient clathrin-mediated endocytosis. Mol. Biol. Cell 16, 2058–2067 59 Shin, N., Ahn, N., Chang-Ileto, B., Park, J., Takei, K., Ahn, S. G., Kim, S. A., Di Paolo, G. and Chang, S. (2008) SNX9 regulates tubular invagination of the plasma membrane through interaction with actin cytoskeleton and dynamin 2. J. Cell Sci. 121, 1252–1263 60 Yeow-Fong, L., Lim, L. and Manser, E. (2005) SNX9 as an adaptor for linking synaptojanin-1 to the Cdc42 effector ACK1. FEBS Lett. 579, 5040–5048 61 Miele, A. E., Watson, P. J., Evans, P. R., Traub, L. M. and Owen, D. J. (2004) Two distinct interaction motifs in amphiphysin bind two independent sites on the clathrin terminal domain β-propeller. Nat. Struct. Mol. Biol. 11, 242–248 62 Baumann, C., Lindholm, C. K., Rimoldi, D. and Levy, F. (2010) The E3 ubiquitin ligase Itch regulates sorting nexin 9 through an unconventional substrate recognition domain. FEBS J. 277, 2803–2814 63 Schulze, W. X. and Mann, M. (2004) A novel proteomic screen for peptide–protein interactions. J. Biol. Chem. 279, 10756–10764 64 Badour, K., McGavin, M. K., Zhang, J., Freeman, S., Vieira, C., Filipp, D., Julius, M., Mills, G. B. and Siminovitch, K. A. (2007) Interaction of the Wiskott–Aldrich syndrome protein with sorting nexin 9 is required for CD28 endocytosis and cosignaling in T cells. Proc. Natl. Acad. Sci. U.S.A. 104, 1593–1598 65 Lin, Q., Lo, C. G., Cerione, R. A. and Yang, W. (2002) The Cdc42 target ACK2 interacts with sorting nexin 9 (SH3PX1) to regulate epidermal growth factor receptor degradation. J. Biol. Chem. 277, 10134–10138 66 Yarar, D., Waterman-Storer, C. M. and Schmid, S. L. (2007) SNX9 couples actin assembly to phosphoinositide signals and is required for membrane remodeling during endocytosis. Dev. Cell 13, 43–56 67 Rangarajan, E. S., Park, H., Fortin, E., Sygusch, J. and Izard, T. (2010) Mechanism of aldolase control of sorting nexin 9 function in endocytosis. J. Biol. Chem. 285, 11983–11990 68 Marches, O., Batchelor, M., Shaw, R. K., Patel, A., Cummings, N., Nagai, T., Sasakawa, C., Carlsson, S. R., Lundmark, R., Cougoule, C. et al. (2006) EspF of enteropathogenic Escherichia coli binds sorting nexin 9. J. Bacteriol. 188, 3110–3115 69 Alto, N. M., Weflen, A. W., Rardin, M. J., Yarar, D., Lazar, C. S., Tonikian, R., Koller, A., Taylor, S. S., Boone, C., Sidhu, S. S. et al. (2007) The type III effector EspF coordinates membrane trafficking by the spatiotemporal activation of two eukaryotic signaling pathways. J. Cell Biol. 178, 1265–1278 70 Dislich, B., Than, M. E. and Lichtenthaler, S. F. (2010) Specific amino acids in the BAR domain allow homodimerization and prevent heterodimerization of sorting nexin 33. Biochem. J. 433, 75–83 71 Haberg, K., Lundmark, R. and Carlsson, S. R. (2008) SNX18 is an SNX9 paralog that acts as a membrane tubulator in AP-1-positive endosomal trafficking. J. Cell Sci. 121, 1495–1505 The PX protein family 72 Nakazawa, S., Gotoh, N., Matsumoto, H., Murayama, C., Suzuki, T. and Yamamoto, T. (2011) Expression of sorting nexin 18 (SNX18) is dynamically regulated in developing spinal motor neurons. J. Histochem. Cytochem. 59, 202–213 73 Karkkainen, S., Hiipakka, M., Wang, J. H., Kleino, I., Vaha-Jaakkola, M., Renkema, G. H., Liss, M., Wagner, R. and Saksela, K. (2006) Identification of preferred protein interactions by phage-display of the human Src homology-3 proteome. EMBO Rep. 7, 186–191 74 Schobel, S., Neumann, S., Hertweck, M., Dislich, B., Kuhn, P. H., Kremmer, E., Seed, B., Baumeister, R., Haass, C. and Lichtenthaler, S. F. (2008) A novel sorting nexin modulates endocytic trafficking and α-secretase cleavage of the amyloid precursor protein. J. Biol. Chem. 283, 14257–14268 75 Abram, C. L., Seals, D. F., Pass, I., Salinsky, D., Maurer, L., Roth, T. M. and Courtneidge, S. A. (2003) The adaptor protein fish associates with members of the ADAMs family and localizes to podosomes of Src-transformed cells. J. Biol. Chem. 278, 16844–16851 76 Zhong, J. L., Poghosyan, Z., Pennington, C. J., Scott, X., Handsley, M. M., Warn, A., Gavrilovic, J., Honert, K., Kruger, A., Span, P. N. et al. (2008) Distinct functions of natural ADAM-15 cytoplasmic domain variants in human mammary carcinoma. Mol. Cancer Res. 6, 383–394 77 Oikawa, T., Itoh, T. and Takenawa, T. (2008) Sequential signals toward podosome formation in NIH-src cells. J. Cell Biol. 182, 157–169 78 Rufer, A. C., Rumpf, J., von Holleben, M., Beer, S., Rittinger, K. and Groemping, Y. (2009) Isoform-selective interaction of the adaptor protein Tks5/FISH with Sos1 and dynamins. J. Mol. Biol. 390, 939–950 79 Stylli, S. S., Stacey, T. T., Verhagen, A. M., Xu, S. S., Pass, I., Courtneidge, S. A. and Lock, P. (2009) Nck adaptor proteins link Tks5 to invadopodia actin regulation and ECM degradation. J. Cell Sci. 122, 2727–2740 80 Thompson, O., Kleino, I., Crimaldi, L., Gimona, M., Saksela, K. and Winder, S. J. (2008) Dystroglycan, Tks5 and Src mediated assembly of podosomes in myoblasts. PLoS ONE 3, e3638 81 Takeya, R., Ueno, N., Kami, K., Taura, M., Kohjima, M., Izaki, T., Nunoi, H. and Sumimoto, H. (2003) Novel human homologues of p47phox and p67phox participate in activation of superoxide-producing NADPH oxidases. J. Biol. Chem. 278, 25234–25246 82 Nakano, Y., Banfi, B., Jesaitis, A. J., Dinauer, M. C., Allen, L. A. and Nauseef, W. M. (2007) Critical roles for p22phox in the structural maturation and subcellular targeting of Nox3. Biochem. J. 403, 97–108 83 Dutta, S. and Rittinger, K. (2010) Regulation of NOXO1 activity through reversible interactions with p22phox and NOXA1. PLoS ONE 5, e10478 84 Park, H. S., Park, D. and Bae, Y. S. (2006) Molecular interaction of NADPH oxidase 1 with βPix and Nox organizer 1. Biochem. Biophys. Res. Commun. 339, 985–990 85 Yamamoto, A., Kami, K., Takeya, R. and Sumimoto, H. (2007) Interaction between the SH3 domains and C-terminal proline-rich region in NADPH oxidase organizer 1 (Noxo1). Biochem. Biophys. Res. Commun. 352, 560–565 86 Valente, A. J., El Jamali, A., Epperson, T. K., Gamez, M. J., Pearson, D. W. and Clark, R. A. (2007) NOX1 NADPH oxidase regulation by the NOXA1 SH3 domain. Free Radic. Biol. Med. 43, 384–396 87 Fuchs, A., Dagher, M. C. and Vignais, P. V. (1995) Mapping the domains of interaction of p40phox with both p47phox and p67phox of the neutrophil oxidase complex using the two-hybrid system. J. Biol. Chem. 270, 5695–5697 88 Wientjes, F. B., Hsuan, J. J., Totty, N. F. and Segal, A. W. (1993) p40phox , a third cytosolic component of the activation complex of the NADPH oxidase to contain src homology 3 domains. Biochem. J. 296, 557–561 89 Ito, T., Nakamura, R., Sumimoto, H., Takeshige, K. and Sakaki, Y. (1996) An SH3 domain-mediated interaction between the phagocyte NADPH oxidase factors p40phox and p47phox . FEBS Lett. 385, 229–232 90 Fuchs, A., Dagher, M. C., Faure, J. and Vignais, P. V. (1996) Topological organization of the cytosolic activating complex of the superoxide-generating NADPH-oxidase. Pinpointing the sites of interaction between p47phoz , p67phox and p40phox using the two-hybrid system. Biochim. Biophys. Acta 1312, 39–47 91 Wientjes, F. B., Panayotou, G., Reeves, E. and Segal, A. W. (1996) Interactions between cytosolic components of the NADPH oxidase: p40phox interacts with both p67phox and p47phox . Biochem. J. 317, 919–924 92 Wilson, L., Butcher, C., Finan, P. and Kellie, S. (1997) SH3 domain-mediated interactions involving the phox components of the NADPH oxidase. Inflamm. Res. 46, 265–271 93 Grizot, S., Grandvaux, N., Fieschi, F., Faure, J., Massenet, C., Andrieu, J. P., Fuchs, A., Vignais, P. V., Timmins, P. A., Dagher, M. C. and Pebay-Peyroula, E. (2001) Small angle neutron scattering and gel filtration analyses of neutrophil NADPH oxidase cytosolic factors highlight the role of the C-terminal end of p47phox in the association with p40phox . Biochemistry 40, 3127–3133 94 Lapouge, K., Smith, S. J., Groemping, Y. and Rittinger, K. (2002) Architecture of the p40–p47–p67phox complex in the resting state of the NADPH oxidase. A central role for p67phox . J. Biol. Chem. 277, 10121–10128 95 Massenet, C., Chenavas, S., Cohen-Addad, C., Dagher, M. C., Brandolin, G., Pebay-Peyroula, E. and Fieschi, F. (2005) Effects of p47phox C terminus phosphorylations on binding interactions with p40phox and p67phox . Structural and functional comparison of p40phox and p67phox SH3 domains. J. Biol. Chem. 280, 13752–13761 96 Grandvaux, N., Grizot, S., Vignais, P. V. and Dagher, M. C. (1999) The Ku70 autoantigen interacts with p40phox in B lymphocytes. J. Cell Sci. 112, 503–513 97 Grogan, A., Reeves, E., Keep, N., Wientjes, F., Totty, N. F., Burlingame, A. L., Hsuan, J. J. and Segal, A. W. (1997) Cytosolic phox proteins interact with and regulate the assembly of coronin in neutrophils. J. Cell Sci. 110, 3071–3081 98 Wientjes, F. B., Reeves, E. P., Soskic, V., Furthmayr, H. and Segal, A. W. (2001) The NADPH oxidase components p47phox and p40phox bind to moesin through their PX domain. Biochem. Biophys. Res. Commun. 289, 382–388 99 Zhan, Y., He, D., Newburger, P. E. and Zhou, G. W. (2004) p47phox PX domain of NADPH oxidase targets cell membrane via moesin-mediated association with the actin cytoskeleton. J. Cell. Biochem. 92, 795–809 100 Tsunawaki, S., Mizunari, H., Nagata, M., Tatsuzawa, O. and Kuratsuji, T. (1994) A novel cytosolic component, p40phox , of respiratory burst oxidase associates with p67phox and is absent in patients with chronic granulomatous disease who lack p67phox . Biochem. Biophys. Res. Commun. 199, 1378–1387 101 Tsunawaki, S., Kagara, S., Yoshikawa, K., Yoshida, L. S., Kuratsuji, T. and Namiki, H. (1996) Involvement of p40phox in activation of phagocyte NADPH oxidase through association of its carboxyl-terminal, but not its amino-terminal, with p67phox . J. Exp. Med. 184, 893–902 102 Nakamura, R., Sumimoto, H., Mizuki, K., Hata, K., Ago, T., Kitajima, S., Takeshige, K., Sakaki, Y. and Ito, T. (1998) The PC motif: a novel and evolutionarily conserved sequence involved in interaction between p40phox and p67phox , SH3 domain-containing cytosolic factors of the phagocyte NADPH oxidase. Eur. J. Biochem. 251, 583–589 103 Wilson, M. I., Gill, D. J., Perisic, O., Quinn, M. T. and Williams, R. L. (2003) PB1 domain-mediated heterodimerization in NADPH oxidase and signaling complexes of atypical protein kinase C with Par6 and p62. Mol. Cell 12, 39–50 104 Nishiyama, A., Ohno, T., Iwata, S., Matsui, M., Hirota, K., Masutani, H., Nakamura, H. and Yodoi, J. (1999) Demonstration of the interaction of thioredoxin with p40phox , a phagocyte oxidase component, using a yeast two-hybrid system. Immunol. Lett. 68, 155–159 105 Hiroaki, H., Ago, T., Ito, T., Sumimoto, H. and Kohda, D. (2001) Solution structure of the PX domain, a target of the SH3 domain. Nat. Struct. Biol. 8, 526–530 106 Shmelzer, Z., Karter, M., Eisenstein, M., Leto, T. L., Hadad, N., Ben-Menahem, D., Gitler, D., Banani, S., Wolach, B., Rotem, M. and Levy, R. (2008) Cytosolic phospholipase A2α is targeted to the p47phox -PX domain of the assembled NADPH oxidase via a novel binding site in its C2 domain. J. Biol. Chem. 283, 31898–31908 107 Gu, Y., Xu, Y. C., Wu, R. F., Nwariaku, F. E., Souza, R. F., Flores, S. C. and Terada, L. S. (2003) p47phox participates in activation of RelA in endothelial cells. J. Biol. Chem. 278, 17210–17217 108 Chen, Q., Powell, D. W., Rane, M. J., Singh, S., Butt, W., Klein, J. B. and McLeish, K. R. (2003) Akt phosphorylates p47phox and mediates respiratory burst activity in human neutrophils. J. Immunol. 170, 5302–5308 109 Touyz, R. M., Yao, G., Quinn, M. T., Pagano, P. J. and Schiffrin, E. L. (2005) p47phox associates with the cytoskeleton through cortactin in human vascular smooth muscle cells: role in NAD(P)H oxidase regulation by angiotensin II. Arterioscler. Thromb. Vasc. Biol. 25, 512–518 110 Tamura, M., Itoh, K., Akita, H., Takano, K. and Oku, S. (2006) Identification of an actin-binding site in p47phox an organizer protein of NADPH oxidase. FEBS Lett. 580, 261–267 111 Finan, P., Shimizu, Y., Gout, I., Hsuan, J., Truong, O., Butcher, C., Bennett, P., Waterfield, M. D. and Kellie, S. (1994) An SH3 domain and proline-rich sequence mediate an interaction between two components of the phagocyte NADPH oxidase complex. J. Biol. Chem. 269, 13752–13755 112 Iyer, S. S., Pearson, D. W., Nauseef, W. M. and Clark, R. A. (1994) Evidence for a readily dissociable complex of p47phox and p67phox in cytosol of unstimulated human neutrophils. J. Biol. Chem. 269, 22405–22411 113 Leusen, J. H., Fluiter, K., Hilarius, P. M., Roos, D., Verhoeven, A. J. and Bolscher, B. G. (1995) Interactions between the cytosolic components p47phox and p67phox of the human neutrophil NADPH oxidase that are not required for activation in the cell-free system. J. Biol. Chem. 270, 11216–11221 114 De Leo, F. R., Ulman, K. V., Davis, A. R., Jutila, K. L. and Quinn, M. T. (1996) Assembly of the human neutrophil NADPH oxidase involves binding of p67phox and flavocytochrome b to a common functional domain in p47phox . J. Biol. Chem. 271, 17013–17020 115 de Mendez, I., Homayounpour, N. and Leto, T. L. (1997) Specificity of p47phox SH3 domain interactions in NADPH oxidase assembly and activation. Mol. Cell. Biol. 17, 2177–2185 c The Authors Journal compilation c 2012 Biochemical Society R. D. Teasdale and B. M. Collins 116 Mizuki, K., Takeya, R., Kuribayashi, F., Nobuhisa, I., Kohda, D., Nunoi, H., Takeshige, K. and Sumimoto, H. (2005) A region C-terminal to the proline-rich core of p47phox regulates activation of the phagocyte NADPH oxidase by interacting with the C-terminal SH3 domain of p67phox . Arch. Biochem. Biophys. 444, 185–194 117 Sumimoto, H., Kage, Y., Nunoi, H., Sasaki, H., Nose, T., Fukumaki, Y., Ohno, M., Minakami, S. and Takeshige, K. (1994) Role of Src homology 3 domains in assembly and activation of the phagocyte NADPH oxidase. Proc. Natl. Acad. Sci. U.S.A. 91, 5345–5349 118 Sumimoto, H., Hata, K., Mizuki, K., Ito, T., Kage, Y., Sakaki, Y., Fukumaki, Y., Nakamura, M. and Takeshige, K. (1996) Assembly and activation of the phagocyte NADPH oxidase. Specific interaction of the N-terminal Src homology 3 domain of p47phox with p22phox is required for activation of the NADPH oxidase. J. Biol. Chem. 271, 22152–22158 119 Liu, H., Nakazawa, T., Tezuka, T. and Yamamoto, T. (2006) Physical and functional interaction of Fyn tyrosine kinase with a brain-enriched Rho GTPase-activating protein TCGAP. J. Biol. Chem. 281, 23611–23619 120 Chiang, S. H., Hwang, J., Legendre, M., Zhang, M., Kimura, A. and Saltiel, A. R. (2003) TCGAP, a multidomain Rho GTPase-activating protein involved in insulin-stimulated glucose transport. EMBO J. 22, 2679–2691 121 Nakamura, T., Komiya, M., Sone, K., Hirose, E., Gotoh, N., Morii, H., Ohta, Y. and Mori, N. (2002) Grit, a GTPase-activating protein for the Rho family, regulates neurite extension through association with the TrkA receptor and N-Shc and CrkL/Crk adapter molecules. Mol. Cell. Biol. 22, 8721–8734 122 Hayashi, T., Okabe, T., Nasu-Nishimura, Y., Sakaue, F., Ohwada, S., Matsuura, K., Akiyama, T. and Nakamura, T. (2007) PX-RICS, a novel splicing variant of RICS, is a main isoform expressed during neural development. Genes Cells 12, 929–939 123 Okabe, T., Nakamura, T., Nishimura, Y. N., Kohu, K., Ohwada, S., Morishita, Y. and Akiyama, T. (2003) RICS, a novel GTPase-activating protein for Cdc42 and Rac1, is involved in the β-catenin-N-cadherin and N -methyl-D-aspartate receptor signaling. J. Biol. Chem. 278, 9920–9927 124 Nakazawa, T., Watabe, A. M., Tezuka, T., Yoshida, Y., Yokoyama, K., Umemori, H., Inoue, A., Okabe, S., Manabe, T. and Yamamoto, T. (2003) p250GAP, a novel brain-enriched GTPase-activating protein for Rho family GTPases, is involved in the N -methylD-aspartate receptor signaling. Mol. Biol. Cell 14, 2921–2934 125 Zhao, C., Ma, H., Bossy-Wetzel, E., Lipton, S. A., Zhang, Z. and Feng, G. S. (2003) GC-GAP, a Rho family GTPase-activating protein that interacts with signaling adapters Gab1 and Gab2. J. Biol. Chem. 278, 34641–34653 126 Shang, X., Moon, S. Y. and Zheng, Y. (2007) p200 RhoGAP promotes cell proliferation by mediating cross-talk between Ras and Rho signaling pathways. J. Biol. Chem. 282, 8801–8811 127 Nakamura, T., Hayashi, T., Mimori-Kiyosue, Y., Sakaue, F., Matsuura, K., Iemura, S., Natsume, T. and Akiyama, T. (2010) The PX-RICS-14–3–3ζ /θ complex couples N-cadherin–β-catenin with dynein–dynactin to mediate its export from the endoplasmic reticulum. J. Biol. Chem. 285, 16145–16154 128 Nakamura, T., Hayashi, T., Nasu-Nishimura, Y., Sakaue, F., Morishita, Y., Okabe, T., Ohwada, S., Matsuura, K. and Akiyama, T. (2008) PX-RICS mediates ER-to-Golgi transport of the N-cadherin/β-catenin complex. Genes Dev. 22, 1244–1256 129 Florian, V., Schluter, T. and Bohnensack, R. (2001) A new member of the sorting nexin family interacts with the C-terminus of P-selectin. Biochem. Biophys. Res. Commun. 281, 1045–1050 130 Knauth, P., Schluter, T., Czubayko, M., Kirsch, C., Florian, V., Schreckenberger, S., Hahn, H. and Bohnensack, R. (2005) Functions of sorting nexin 17 domains and recognition motif for P-selectin trafficking. J. Mol. Biol. 347, 813–825 131 Betts, G. N., van der Geer, P. and Komives, E. A. (2008) Structural and functional consequences of tyrosine phosphorylation in the LRP1 cytoplasmic domain. J. Biol. Chem. 283, 15656–15664 132 Donoso, M., Cancino, J., Lee, J., van Kerkhof, P., Retamal, C., Bu, G., Gonzalez, A., Caceres, A. and Marzolo, M. P. (2009) Polarized traffic of LRP1 involves AP1B and SNX17 operating on Y-dependent sorting motifs in different pathways. Mol. Biol. Cell 20, 481–497 133 Stockinger, W., Sailler, B., Strasser, V., Recheis, B., Fasching, D., Kahr, L., Schneider, W. J. and Nimpf, J. (2002) The PX-domain protein SNX17 interacts with members of the LDL receptor family and modulates endocytosis of the LDL receptor. EMBO J. 21, 4259–4267 134 van Kerkhof, P., Lee, J., McCormick, L., Tetrault, E., Lu, W., Schoenfish, M., Oorschot, V., Strous, G. J., Klumperman, J. and Bu, G. (2005) Sorting nexin 17 facilitates LRP recycling in the early endosome. EMBO J. 24, 2851–2861 135 Burden, J. J., Sun, X. M., Garcia, A. B. and Soutar, A. K. (2004) Sorting motifs in the intracellular domain of the low density lipoprotein receptor interact with a novel domain of sorting nexin-17. J. Biol. Chem. 279, 16237–16245 136 Lee, J., Retamal, C., Cuitino, L., Caruano-Yzermans, A., Shin, J. E., van Kerkhof, P., Marzolo, M. P. and Bu, G. (2008) Adaptor protein sorting nexin 17 regulates amyloid precursor protein trafficking and processing in the early endosomes. J. Biol. Chem. 283, 11501–11508 c The Authors Journal compilation c 2012 Biochemical Society 137 Ghai, R., Mobli, M., Norwood, S. J., Bugarcic, A., Teasdale, R. D., King, G. F. and Collins, B. M. (2011) Phox homology band 4.1/ezrin/radixin/moesin-like proteins function as molecular scaffolds that interact with cargo receptors and Ras GTPases. Proc. Natl. Acad. Sci. U.S.A 108, 7763–7768 138 Adachi, H. and Tsujimoto, M. (2010) Adaptor protein sorting nexin 17 interacts with the scavenger receptor FEEL-1/stabilin-1 and modulates its expression on the cell surface. Biochim. Biophys. Acta 1803, 553–563 139 Czubayko, M., Knauth, P., Schluter, T., Florian, V. and Bohnensack, R. (2006) Sorting nexin 17, a non-self-assembling and a PtdIns(3)P high class affinity protein, interacts with the cerebral cavernous malformation related protein KRIT1. Biochem. Biophys. Res. Commun. 345, 1264–1272 140 Seog, D. H. and Han, J. (2008) Sorting nexin 17 interacts directly with kinesin superfamily KIF1Bβ protein. Korean J. Physiol. Pharmacol. 12, 199–204 141 Joubert, L., Hanson, B., Barthet, G., Sebben, M., Claeysen, S., Hong, W., Marin, P., Dumuis, A. and Bockaert, J. (2004) New sorting nexin (SNX27) and NHERF specifically interact with the 5-HT4a receptor splice variant: roles in receptor targeting. J. Cell Sci. 117, 5367–5379 142 Lunn, M. L., Nassirpour, R., Arrabit, C., Tan, J., McLeod, I., Arias, C. M., Sawchenko, P. E., Yates, 3rd, J.R. and Slesinger, P. A. (2007) A unique sorting nexin regulates trafficking of potassium channels via a PDZ domain interaction. Nat. Neurosci. 10, 1249–1259 143 Nassirpour, R. and Slesinger, P. A. (2007) Subunit-specific regulation of Kir3 channels by sorting nexin 27. Channels (Austin) 1, 331–333 144 Balana, B., Maslennikov, I., Kwiatkowski, W., Stern, K. M., Bahima, L., Choe, S. and Slesinger, P. A. (2011) Mechanism underlying selective regulation of G protein-gated inwardly rectifying potassium channels by the psychostimulant-sensitive sorting nexin 27. Proc. Natl. Acad. Sci. U.S.A. 108, 5831–5836 145 Cai, L., Loo, L. S., Atlashkin, V., Hanson, B. J. and Hong, W. (2011) Deficiency of sorting nexin 27 (SNX27) leads to growth retardation and elevated levels of N -methylD-aspartate (NMDA) receptor 2C (NR2C). Mol. Cell. Biol. 31, 1734–1747 146 Lauffer, B. E., Melero, C., Temkin, P., Lei, C., Hong, W., Kortemme, T. and von Zastrow, M. (2010) SNX27 mediates PDZ-directed sorting from endosomes to the plasma membrane. J. Cell Biol. 190, 565–574 147 MacNeil, A. J., Mansour, M. and Pohajdak, B. (2007) Sorting nexin 27 interacts with the cytohesin associated scaffolding protein (CASP) in lymphocytes. Biochem. Biophys. Res. Commun. 359, 848–853 148 MacNeil, A. J. and Pohajdak, B. (2009) Getting a GRASP on CASP: properties and role of the cytohesin-associated scaffolding protein in immunity. Immunol. Cell Biol. 87, 72–80 149 Rincon, E., Santos, T., Avila-Flores, A., Albar, J. P., Lalioti, V., Lei, C., Hong, W. and Merida, I. (2007) Proteomics identification of sorting nexin 27 as a diacylglycerol kinase ζ -associated protein: new diacylglycerol kinase roles in endocytic recycling. Mol. Cell. Proteomics 6, 1073–1087 150 Rincon, E., de Guinoa, J. S., Gharbi, S. I., Sorzano, C. O., Carrasco, Y. R. and Merida, I. (2011) Translocation dynamics of sorting nexin 27 in activated T cells. J. Cell Sci. 124, 776–788 151 Temkin, P., Lauffer, B., Jager, S., Cimermancic, P., Krogan, N. J. and von Zastrow, M. (2011) SNX27 mediates retromer tubule entry and endosome-to-plasma membrane trafficking of signalling receptors. Nat. Cell Biol. 13, 715–721 152 Zheng, B., Lavoie, C., Tang, T. D., Ma, P., Meerloo, T., Beas, A. and Farquhar, M. G. (2004) Regulation of epidermal growth factor receptor degradation by heterotrimeric Gαs protein. Mol. Biol. Cell 15, 5538–5550 153 Zheng, B., Ma, Y. C., Ostrom, R. S., Lavoie, C., Gill, G. N., Insel, P. A., Huang, X. Y. and Farquhar, M. G. (2001) RGS-PX1, a GAP for Gαs and sorting nexin in vesicular trafficking. Science 294, 1939–1942 154 Hu, Y. F., Zhang, H. L., Cai, T., Harashima, S. and Notkins, A. L. (2005) The IA-2 interactome. Diabetologia 48, 2576–2581 155 Mao, H., Ferguson, T. S., Cibulsky, S. M., Holmqvist, M., Ding, C., Fei, H. and Levitan, I. B. (2005) MONaKA, a novel modulator of the plasma membrane Na,K-ATPase. J. Neurosci. 25, 7934–7943 156 Takeuchi, H., Takeuchi, T., Gao, J., Cantley, L. C. and Hirata, M. (2010) Characterization of PXK as a protein involved in epidermal growth factor receptor trafficking. Mol. Cell. Biol. 30, 1689–1702 157 Hayashi, S., Okada, T., Igarashi, N., Fujita, T., Jahangeer, S. and Nakamura, S. (2002) Identification and characterization of RPK118, a novel sphingosine kinase-1-binding protein. J. Biol. Chem. 277, 33319–33324 158 Liu, L., Yang, C., Yuan, J., Chen, X., Xu, J., Wei, Y., Yang, J., Lin, G. and Yu, L. (2005) RPK118, a PX domain-containing protein, interacts with peroxiredoxin-3 through pseudo-kinase domains. Mol. Cells 19, 39–45 159 Slagsvold, T., Marchese, A., Brech, A. and Stenmark, H. (2006) CISK attenuates degradation of the chemokine receptor CXCR4 via the ubiquitin ligase AIP4. EMBO J. 25, 3738–3749 The PX protein family 160 Dai, F., Yu, L., He, H., Chen, Y., Yu, J., Yang, Y., Xu, Y., Ling, W. and Zhao, S. (2002) Human serum and glucocorticoid-inducible kinase-like kinase (SGKL) phosphorylates glycogen syntheses kinase 3 β (GSK-3β) at serine-9 through direct interaction. Biochem. Biophys. Res. Commun. 293, 1191–1196 161 Gaidarov, I., Smith, M. E., Domin, J. and Keen, J. H. (2001) The class II phosphoinositide 3-kinase C2α is activated by clathrin and regulates clathrin-mediated membrane trafficking. Mol. Cell 7, 443–449 162 Gaidarov, I., Zhao, Y. and Keen, J. H. (2005) Individual phosphoinositide 3-kinase C2α domain activities independently regulate clathrin function. J. Biol. Chem. 280, 40766–40772 163 Zhao, Y., Gaidarov, I. and Keen, J. H. (2007) Phosphoinositide 3-kinase C2α links clathrin to microtubule-dependent movement. J. Biol. Chem. 282, 1249–1256 164 Arcaro, A., Zvelebil, M. J., Wallasch, C., Ullrich, A., Waterfield, M. D. and Domin, J. (2000) Class II phosphoinositide 3-kinases are downstream targets of activated polypeptide growth factor receptors. Mol. Cell. Biol. 20, 3817–3830 165 Wheeler, M. and Domin, J. (2006) The N-terminus of phosphoinositide 3-kinase-C2β regulates lipid kinase activity and binding to clathrin. J. Cell. Physiol. 206, 586–593 166 Katso, R. M., Pardo, O. E., Palamidessi, A., Franz, C. M., Marinov, M., De Laurentiis, A., Downward, J., Scita, G., Ridley, A. J., Waterfield, M. D. and Arcaro, A. (2006) Phosphoinositide 3-kinase C2β regulates cytoskeletal organization and cell migration via Rac-dependent mechanisms. Mol. Biol. Cell 17, 3729–3744 167 Wheeler, M. and Domin, J. (2001) Recruitment of the class II phosphoinositide 3-kinase C2β to the epidermal growth factor receptor: role of Grb2. Mol. Cell. Biol. 21, 6660–6667 168 Paulhe, F., Perret, B., Chap, H., Iberg, N., Morand, O. and Racaud-Sultan, C. (2002) Phosphoinositide 3-kinase C2α is activated upon smooth muscle cell migration and regulated by α v β 3 integrin engagement. Biochem. Biophys. Res. Commun. 297, 261–266 169 Kam, Y. and Exton, J. H. (2002) Dimerization of phospholipase D isozymes. Biochem. Biophys. Res. Commun. 290, 375–380 170 Slaaby, R., Jensen, T., Hansen, H. S., Frohman, M. A. and Seedorf, K. (1998) PLD2 complexes with the EGF receptor and undergoes tyrosine phosphorylation at a single site upon agonist stimulation. J. Biol. Chem. 273, 33722–33727 171 Jin, J. K., Ahn, B. H., Na, Y. J., Kim, J. I., Kim, Y. S., Choi, E. K., Ko, Y. G., Chung, K. C., Kozlowski, P. B. and Min, D. S. (2007) Phospholipase D1 is associated with amyloid precursor protein in Alzheimer’s disease. Neurobiol. Aging 28, 1015–1027 172 Cai, D., Netzer, W. J., Zhong, M., Lin, Y., Du, G., Frohman, M., Foster, D. A., Sisodia, S. S., Xu, H., Gorelick, F. S. and Greengard, P. (2006) Presenilin-1 uses phospholipase D1 as a negative regulator of β-amyloid formation. Proc. Natl. Acad. Sci. U.S.A. 103, 1941–1946 173 Kim, J. H., Lee, S. D., Han, J. M., Lee, T. G., Kim, Y., Park, J. B., Lambeth, J. D., Suh, P. G. and Ryu, S. H. (1998) Activation of phospholipase D1 by direct interaction with ADP-ribosylation factor 1 and RalA. FEBS Lett. 430, 231–235 174 Luo, J. Q., Liu, X., Hammond, S. M., Colley, W. C., Feig, L. A., Frohman, M. A., Morris, A. J. and Foster, D. A. (1997) RalA interacts directly with the Arf-responsive, PIP2dependent phospholipase D1. Biochem. Biophys. Res. Commun. 235, 854–859 175 Vitale, N., Mawet, J., Camonis, J., Regazzi, R., Bader, M. F. and Chasserot-Golaz, S. (2005) The Small GTPase RalA controls exocytosis of large dense core secretory granules by interacting with ARF6-dependent phospholipase D1. J. Biol. Chem. 280, 29921–29928 176 Liu, L., Liao, H., Castle, A., Zhang, J., Casanova, J., Szabo, G. and Castle, D. (2005) SCAMP2 interacts with Arf6 and phospholipase D1 and links their function to exocytotic fusion pore formation in PC12 cells. Mol. Biol. Cell 16, 4463–4472 177 Kim, J. H., Han, J. M., Lee, S., Kim, Y., Lee, T. G., Park, J. B., Lee, S. D., Suh, P. G. and Ryu, S. H. (1999) Phospholipase D1 in caveolae: regulation by protein kinase Cα and caveolin-1. Biochemistry 38, 3763–3769 178 Haga, Y., Miwa, N., Jahangeer, S., Okada, T. and Nakamura, S. (2009) CtBP1/BARS is an activator of phospholipase D1 necessary for agonist-induced macropinocytosis. EMBO J. 28, 1197–1207 179 Lee, H. Y., Park, J. B., Jang, I. H., Chae, Y. C., Kim, J. H., Kim, I. S., Suh, P. G. and Ryu, S. H. (2004) Munc-18–1 inhibits phospholipase D activity by direct interaction in an epidermal growth factor-reversible manner. J. Biol. Chem. 279, 16339–16348 180 Lee, C. S., Kim, I. S., Park, J. B., Lee, M. N., Lee, H. Y., Suh, P. G. and Ryu, S. H. (2006) The phox homology domain of phospholipase D activates dynamin GTPase activity and accelerates EGFR endocytosis. Nat. Cell Biol. 8, 477–484 181 Lee, C., Kim, S. R., Chung, J. K., Frohman, M. A., Kilimann, M. W. and Rhee, S. G. (2000) Inhibition of phospholipase D by amphiphysins. J. Biol. Chem. 275, 18751–18758 182 Cho, J. H., Oh, D. Y., Kim, H. J., Park, S. Y., Choi, H. J., Kwon, S. J., Lee, K. S. and Han, J. S. (2011) The TSP motif in AP180 inhibits phospholipase D1 activity resulting in increased efficacy of anticancer drug via its direct binding to carboxyl terminal of phospholipase D1. Cancer Lett. 302, 144–154 183 Sung, T. C., Zhang, Y., Morris, A. J. and Frohman, M. A. (1999) Structural analysis of human phospholipase D1. J. Biol. Chem. 274, 3659–3666 184 Hu, T. and Exton, J. H. (2003) Mechanisms of regulation of phospholipase D1 by protein kinase Cα. J. Biol. Chem. 278, 2348–2355 185 Hu, T. and Exton, J. H. (2004) Protein kinase Cα translocates to the perinuclear region to activate phospholipase D1. J. Biol. Chem. 279, 35702–35708 186 Jin, J. K., Kim, N. H., Lee, Y. J., Kim, Y. S., Choi, E. K., Kozlowski, P. B., Park, M. H., Kim, H. S. and Min, D. S. (2006) Phospholipase D1 is up-regulated in the mitochondrial fraction from the brains of Alzheimer’s disease patients. Neurosci. Lett. 407, 263–267 187 Jang, I. H., Lee, S., Park, J. B., Kim, J. H., Lee, C. S., Hur, E. M., Kim, I. S., Kim, K. T., Yagisawa, H., Suh, P. G. and Ryu, S. H. (2003) The direct interaction of phospholipase C-γ 1 with phospholipase D2 is important for epidermal growth factor signaling. J. Biol. Chem. 278, 18184–18190 188 Divecha, N., Roefs, M., Halstead, J. R., D’Andrea, S., Fernandez-Borga, M., Oomen, L., Saqib, K. M., Wakelam, M. J. and D’Santos, C. (2000) Interaction of the type Iα PIP kinase with phospholipase D: a role for the local generation of phosphatidylinositol 4,5-bisphosphate in the regulation of PLD2 activity. EMBO J. 19, 5440–5449 189 Zhang, Y., Redina, O., Altshuller, Y. M., Yamazaki, M., Ramos, J., Chneiweiss, H., Kanaho, Y. and Frohman, M. A. (2000) Regulation of expression of phospholipase D1 and D2 by PEA-15, a novel protein that interacts with them. J. Biol. Chem. 275, 35224–35232 190 Ahn, B. H., Min, G., Bae, Y. S. and Min, D. S. (2006) Phospholipase D is activated and phosphorylated by casein kinase-II in human U87 astroglioma cells. Exp. Mol. Med. 38, 55–62 191 Ahn, B. H., Kim, S. Y., Kim, E. H., Choi, K. S., Kwon, T. K., Lee, Y. H., Chang, J. S., Kim, M. S., Jo, Y. H. and Min, D. S. (2003) Transmodulation between phospholipase D and c-Src enhances cell proliferation. Mol. Cell. Biol. 23, 3103–3115 192 Yoon, M. S., Cho, C. H., Lee, K. S. and Han, J. S. (2006) Binding of Cdc42 to phospholipase D1 is important in neurite outgrowth of neural stem cells. Biochem. Biophys. Res. Commun. 347, 594–600 193 Yamazaki, M., Zhang, Y., Watanabe, H., Yokozeki, T., Ohno, S., Kaibuchi, K., Shibata, H., Mukai, H., Ono, Y., Frohman, M. A. and Kanaho, Y. (1999) Interaction of the small G protein RhoA with the C terminus of human phospholipase D1. J. Biol. Chem. 274, 6035–6038 194 Jang, M. J., Lee, M. J., Park, H. Y., Bae, Y. S., Min, D. S., Ryu, S. H. and Kwak, J. Y. (2004) Phosphorylation of phospholipase D1 and the modulation of its interaction with RhoA by cAMP-dependent protein kinase. Exp. Mol. Med. 36, 172–178 195 Kusner, D. J., Barton, J. A., Wen, K. K., Wang, X., Rubenstein, P. A. and Iyer, S. S. (2002) Regulation of phospholipase D activity by actin. Actin exerts bidirectional modulation of mammalian phospholipase D activity in a polymerization-dependent, isoform-specific manner. J. Biol. Chem. 277, 50683–50692 196 Ahn, B. H., Rhim, H., Kim, S. Y., Sung, Y. M., Lee, M. Y., Choi, J. Y., Wolozin, B., Chang, J. S., Lee, Y. H., Kwon, T. K. et al. (2002) α-Synuclein interacts with phospholipase D isozymes and inhibits pervanadate-induced phospholipase D activation in human embryonic kidney-293 cells. J. Biol. Chem. 277, 12334–12342 197 Park, J. B., Lee, C. S., Lee, H. Y., Kim, I. S., Lee, B. D., Jang, I. H., Jung, Y. W., Oh, Y. S., Han, M. Y., Jensen, O. N. et al. (2004) Regulation of phospholipase D2 by GTPdependent interaction with dynamin. Adv. Enzyme Regul. 44, 249–264 198 Czarny, M., Fiucci, G., Lavie, Y., Banno, Y., Nozawa, Y. and Liscovitch, M. (2000) Phospholipase D2: functional interaction with caveolin in low-density membrane microdomains. FEBS Lett. 467, 326–332 199 Ha, S. H., Kim, D. H., Kim, I. S., Kim, J. H., Lee, M. N., Lee, H. J., Jang, S. K., Suh, P. G. and Ryu, S. H. (2006) PLD2 forms a functional complex with mTOR/raptor to transduce mitogenic signals. Cell. Signal. 18, 2283–2291 200 Lee, J. H., Kim, Y. M., Kim, N. W., Kim, J. W., Her, E., Kim, B. K., Kim, J. H., Ryu, S. H., Park, J. W., Seo, D. W. et al. (2006) Phospholipase D2 acts as an essential adaptor protein in the activation of Syk in antigen-stimulated mast cells. Blood 108, 956–964 201 Horn, J., Lopez, I., Miller, M. W. and Gomez-Cambronero, J. (2005) The uncovering of a novel regulatory mechanism for PLD2: formation of a ternary complex with protein tyrosine phosphatase PTP1B and growth factor receptor-bound protein GRB2. Biochem. Biophys. Res. Commun. 332, 58–67 202 Kim, J. H., Ohba, M., Suh, P. G. and Ryu, S. H. (2005) Novel functions of the phospholipase D2-Phox homology domain in protein kinase Cζ activation. Mol. Cell. Biol. 25, 3194–3208 203 Di Fulvio, M., Frondorf, K., Henkels, K. M., Lehman, N. and Gomez-Cambronero, J. (2007) The Grb2/PLD2 interaction is essential for lipase activity, intracellular localization and signaling in response to EGF. J. Mol. Biol. 367, 814–824 204 Di Fulvio, M., Lehman, N., Lin, X., Lopez, I. and Gomez-Cambronero, J. (2006) The elucidation of novel SH2 binding sites on PLD2. Oncogene 25, 3032–3040 205 Mahankali, M., Peng, H. J., Cox, D. and Gomez-Cambronero, J. (2011) The mechanism of cell membrane ruffling relies on a phospholipase D2 (PLD2), Grb2 and Rac2 association. Cell. Signal. 23, 1291–1298 c The Authors Journal compilation c 2012 Biochemical Society R. D. Teasdale and B. M. Collins 206 Peng, H. J., Henkels, K. M., Mahankali, M., Dinauer, M. C. and Gomez-Cambronero, J. (2011) Evidence for two CRIB domains in phospholipase D2 (PLD2) that the enzyme uses to specifically bind to the small GTPase Rac2. J. Biol. Chem. 286, 16308–16320 207 Lee, S., Park, J. B., Kim, J. H., Kim, Y., Shin, K. J., Lee, J. S., Ha, S. H., Suh, P. G. and Ryu, S. H. (2001) Actin directly interacts with phospholipase D, inhibiting its activity. J. Biol. Chem. 276, 28252–28260 208 Jeon, H., Kwak, D., Noh, J., Lee, M. N., Lee, C. S., Suh, P. G. and Ryu, S. H. (2011) Phospholipase D2 induces stress fiber formation through mediating nucleotide exchange for RhoA. Cell. Signal. 23, 1320–1326 209 Kim, J. H., Lee, S., Lee, T. G., Hirata, M., Suh, P. G. and Ryu, S. H. (2002) Phospholipase D2 directly interacts with aldolase via Its PH domain. Biochemistry 41, 3414–3421 210 Schaff, U. Y., Shih, H. H., Lorenz, M., Sako, D., Kriz, R., Milarski, K., Bates, B., Tchernychev, B., Shaw, G. D. and Simon, S. I. (2008) SLIC-1/sorting nexin 20: a novel sorting nexin that directs subcellular distribution of PSGL-1. Eur. J. Immunol. 38, 550–564 211 Boulkroun, S., Ruffieux-Daidie, D., Vitagliano, J. J., Poirot, O., Charles, R. P., Lagnaz, D., Firsov, D., Kellenberger, S. and Staub, O. (2008) Vasopressin-inducible ubiquitin-specific protease 10 increases ENaC cell surface expression by deubiquitylating and stabilizing sorting nexin 3. Am. J. Physiol. Renal Physiol. 295, F889–F900 212 Takemoto, Y., Furuta, M., Sato, M., Kubo, M. and Hashimoto, Y. (1999) Isolation and characterization of a novel HS1 SH3 domain binding protein, HS1BP3. Int. Immunol. 11, 1957–1964 213 Phillips, S. A., Barr, V. A., Haft, D. H., Taylor, S. I. and Haft, C. R. (2001) Identification and characterization of SNX15, a novel sorting nexin involved in protein trafficking. J. Biol. Chem. 276, 5074–5084 Received 11 July 2011/26 July 2011; accepted 27 July 2011 Published on the Internet 14 December 2011, doi:10.1042/BJ20111226 c The Authors Journal compilation c 2012 Biochemical Society 214 Lim, K. P. and Hong, W. (2004) Human Nischarin/imidazoline receptor antisera-selected protein is targeted to the endosomes by a combined action of a PX domain and a coiled-coil region. J. Biol. Chem. 279, 54770–54782 215 Alahari, S. K., Lee, J. W. and Juliano, R. L. (2000) Nischarin, a novel protein that interacts with the integrin α5 subunit and inhibits cell migration. J. Cell Biol. 151, 1141–1154 216 Alahari, S. K. and Nasrallah, H. (2004) A membrane proximal region of the integrin α5 subunit is important for its interaction with nischarin. Biochem. J. 377, 449–457 217 Sano, H., Liu, S. C., Lane, W. S., Piletz, J. E. and Lienhard, G. E. (2002) Insulin receptor substrate 4 associates with the protein IRAS. J. Biol. Chem. 277, 19439–19447 218 Alahari, S. K., Reddig, P. J. and Juliano, R. L. (2004) The integrin-binding protein nischarin regulates cell migration by inhibiting PAK. EMBO J. 23, 2777–2788 219 Ding, Y., Milosavljevic, T. and Alahari, S. K. (2008) Nischarin inhibits LIM kinase to regulate cofilin phosphorylation and cell invasion. Mol. Cell. Biol. 28, 3742–3756 220 Reddig, P. J., Xu, D. and Juliano, R. L. (2005) Regulation of p21-activated kinase-independent Rac1 signal transduction by nischarin. J. Biol. Chem. 280, 30994–31002 221 Choi, J. H., Hong, W. P., Kim, M. J., Kim, J. H., Ryu, S. H. and Suh, P. G. (2004) Sorting nexin 16 regulates EGF receptor trafficking by phosphatidylinositol-3-phosphate interaction with the Phox domain. J. Cell Sci. 117, 4209–4218 222 Hanson, B. J. and Hong, W. (2003) Evidence for a role of SNX16 in regulating traffic between the early and later endosomal compartments. J. Biol. Chem. 278, 34617–34630 223 Kienle, N., Kloepper, T. H. and Fasshauer, D. (2009) Phylogeny of the SNARE vesicle fusion machinery yields insights into the conservation of the secretory pathway in fungi. BMC Evol. Biol. 9, 19
© Copyright 2025 Paperzz