INTERORGANELLAR COMMUNICATION: COMPONENTS
Contents Adaptor Proteins: Inter-Organelle Traffic Controllers SNAREs: Membrane Fusion and Beyond ESCRTing around the Cell The Retromer Complex Vesicle Tethers BAR Domains and BAR Domain Superfamily Proteins
Adaptor Proteins: Inter-Organelle Traffic Controllers K Madhivanan, W-C Hsieh, and RC Aguilar, Purdue University Center for Cancer Research, West Lafayette, IN, USA r 2016 Elsevier Inc. All rights reserved.
Adaptor Proteins Adaptors were originally defined as proteins able to link transmembrane cargo proteins to the membrane coat. This definition was implicitly expanded to also require binding to phospholipids and other elements of the trafficking machinery to incorporate cargo into and to facilitate the assembly of transport carriers. Whereas some adaptors have the ability to bind membrane, cargo, and coat proteins, others indirectly fulfill these requirements by interacting with bridging elements and consolidating the protein network (Table 1).
Heterotetrameric Adaptor Complexes Assembly polypeptides (APs) or adaptins are a family of protein complexes involved in vesicle trafficking with five members (AP1–5) found in most eukaryotes (Bonifacino, 2004; Hirst et al., 2013). APs are heterotetramers composed of two large subunits (B100 kDa, β1/γ, β2/α, β3/δ, β4/ε, β5/ζ in AP1–5 respectively), one medium (B50 kDa, μ1–5), and one small subunit (B20 kDa, s1–5) (Figure 1(a)). In addition, AP1–3 exhibit cell type-specific isoforms. While vertebrates and plants have all the 5 members of this family, only AP1–3 are present in flies, worms, and yeast (Hirst et al., 2013). Surprisingly, while in multicellular organisms several APs are required for normal embryonic development, they are dispensable for survival and clathrin-mediated endocytosis (CME) in yeast (Mitsunari et al., 2005; Huang et al., 1999). The large subunits consist of an N-terminal trunk domain, followed by an unstructured hinge domain and a C-terminal appendage domain (Collins et al., 2002; Figure 1(a)). The N-terminal trunks together with the μ and s subunits form the core of the AP complex. While the trunk domains of α, γ, δ, ε,
450
and ζ subunits are involved in binding to target membranes, the μ and s subunits are involved in cargo recognition. AP recruitment to membranes is facilitated by ADP-ribosylation factor (ARF) family of proteins in their membrane-bound, GTP-loaded state. While, Arf-6 primarily fulfils this function at the plasma membrane, Arf-1 does it at the trans-Golgi network (TGN) (Ren et al., 2013). The hinge domains of the β subunits in AP1–3 have one or more LФXФ[D/E] (L represents Leucine, X represents any amino acid, and Ф represents bulky hydrophobic amino acids, such as leucine, isoleucine, methionine, valine, and phenylalanine) motifs responsible for binding to clathrin (Owen et al., 2004). However, β4 and β5 lack this sequence and thus AP4 and AP5 complexes were suggested to mediate clathrin-independent vesicle trafficking (Hirst et al., 2013). The appendage domains serve as a platform for interacting with other adaptor and accessory proteins (Figure 1(b); Owen et al., 2004). While the N-terminal domain of μ subunits contributes to the core, the C-terminal domain recognizes tyrosine-based sorting signals within cargoes. AP complexes mediate the sorting of various transmembrane proteins, including type I, type II, and multi-span proteins. To recruit cargoes to vesicles, APs recognize short sorting signal sequences that reside in the cytosolic region, usually at 4–10 residues from the transmembrane domain. There are two well-characterized AP-targeting sorting signal sequences: tyrosine-based and leucine-based signals (Nakatsu and Ohno, 2003; Traub, 2009). The tyrosine signal consists of four amino acids fitting the YXXФ consensus, where Y stand for tyrosine. The medium μ subunits of AP complexes have been identified to play a central role in YXXФ sequence recognition. The leucine-based signals recognized by APs fit into the [D/E]XXXL[L/Ф] consensus, where the -4 position from the first leucine is either aspartic acid or glutamic acid, then followed by three nonconserved amino acids. This sequence motif precedes two
Encyclopedia of Cell Biology, Volume 2
doi:10.1016/B978-0-12-394447-4.20044-8
Interorganellar Communication: Components: Adaptor Proteins: Inter-Organelle Traffic Controllers
Table 1
Adaptor properties
Adaptor name
Clathrin (CBM)
AP2 (ABM)
Lipid – Adaptor binding domain
Example of cargo (signal) – Adaptor Other domains – binding interacting domain/subunit partners
Classical heterotetrameric adaptor proteins recognizing YxxФ and dill signals AP1 þ PI4P – γ,β1 CI-M6PR (YXXФ)-μ1, (diLL)-[γ/s1] TfR (YXXФ)-μ2, CD4 (diLL)-[α/s2] AP2 þ PI(4,5)P2, PI(3,4,5)P3 – α,m2 AP3 þ ? Lamp-1(YXXФ)-μ3, Tyrosinase (diLL)-[δ/s3] AP4 – ? APP (YKFFE)-m4 AP5 – ? ?
451
Localization
TGN Plasma membrane Endosome TGN Late endosome
Adaptors for cargo carrying acidic DXXLL peptide GGA1,2,3 þ PI4P – GAT
M6PR(DXXLL)-VHS
GAT – Ub
TGN
Adaptors for ubiquitinated-cargo Epsin þ þ
PI(4,5)P2 – ENTH
VEGFR2 – UIM
NPF – Eps15
PI(3,5)P2 – EH
EGFR – UIM
ESFGDGFADFSTLS – AP1
Plasma membrane Plasma membrane
PI4P – ENTH
c-Met receptor – coiled coil Vtib-?
DFxD[F/W] – AP1
Eps15
–
Eps15R
þ
þ
Adaptors for cargo carrying NPxY peptide Dab1 – þ PI(4,5)P2 – PTB
ApoER2 (NPxY) – PTB
Dab2
þ
þ
PI(4,5)P2 – PTB
LDLR (NPxY) – PTB
ARH
þ
þ
PI(4,5)P2 – PTB
LDLR (NPxY) – PTB
Numb
þ
þ
PI(4,5)P2 – PTB
APP (NPxY) – PTB
NPF – Eps15
NPF – Eps15
TGN Plasma membrane Plasma membrane Plasma membrane Plasma membrane
Adaptors for GPCRs (Ser and Thr clusters) β-Arr þ þ PI(4,5)P2
GPCRs – Arrestin fold
Plasma membrane
Adaptors for VAMPs AP180 þ
Plasma membrane Plasma membrane
CALM
þ
þ
PI(4,5)P2 – ANTH
Synaptobrevin2 – ANTH
þ
PI(4,5)P2 – ANTH
Synaptobrevin2 – ANTH
NPF – Eps15
Notes: þ , Motif present; , Motif absent; ?, Not known; Ф, Hydrophobic amino acid; x, Any amino acid, single letter abbreviations of amino acids are used in the table above. Abbreviations: ANTH, AP180 N-terminal homology domain; ApoER2, Apolipoprotein E receptor 2; APP, Amyloid b precursor protein; CI-M6PR, Cation-independent mannose phosphate receptor; EGFR, Epidermal growth factor receptor; EH, Eps15 Homology; ENTH, Epsin N-terminal homology domain; GAT, GGA and Tom; GPCR, G-Protein coupled receptor; LDLR, Low-density lipoprotein 2; PIP, Phosphotidyl inositol phosphate; PTB, Phosphotyrosine binding domain; TfR, Transferrin receptor; TGN, Trans-Golgi network; Ub, ubiquitin; VEGFR, Vascular endothelial growth factor; VHS − Vps27, Hrs and STAM.
leucines that lead to its ‘dileucine signal’ denomination. The first leucine cannot be replaced by any other amino acid, while the second can be substituted with a hydrophobic residue. In some proteins, the acidic residue at -4 position in the consensus can be replaced by a serine that upon phosphorylation acquires a negative charge and becomes functionally equivalent to the D/E residues. The dileucine signal is recognized by γ-s1, α-s2, and δ-s3 hemicomplexes of AP1–3 adaptors (Mattera et al., 2011).
AP1 This adaptor is expressed ubiquitously and is essential during mammalian development (Meyer et al., 2000). The AP1
gamma subunit interacts with the Golgi apparatus-enriched lipid phosphatidylinositol-4-phosphate (PI(4)P) and it is involved in cargo transport between the TGN and endosomal/ lysosomal compartments (Wang et al., 2003; Lefkir et al., 2003). The localization of AP1 is also regulated by phosphorylation of its subunits, namely AP1 associates with membranes when the μ1 subunit is phosphorylated, whereas it detaches when β1 is phosphorylated (Ghosh and Kornfeld, 2003). The cargoes of AP1 include furin, mannose-6-phospate receptors (M6PR), and other proteins of the secretory pathway that are processed at the Golgi apparatus (Bonnemaison et al., 2013; Table 1). It has also been suggested that AP1 and Golgilocalized gamma-adaptin ear-containing Arf-binding proteins (GGAs) cooperate in cargo recruitment to clathrin-coated vesicles (CCVs) by recognition of sorting motifs on the
452
Interorganellar Communication: Components: Adaptor Proteins: Inter-Organelle Traffic Controllers
AP1
AP2
AP3
AP4
AP5
1
2
3
4
5
1
2
3
4
5
1
2
3
4
5
(a)
GFP-EPsin2
AP2
Merge
(b)
Figure 1 Heterotetrameric adaptor complexes. (a). Left panel: Cartoon representing the subunit organization of the AP1 complex as example of a heterotetrameric adaptin. Right panel: Subunit composition of known AP complexes. (b) Colocalization of AP2 with the monomeric adaptor Epsin2. HeLa cells expressing GFP-Epsin2 (green) were fixed and the presence of AP2 was revealed by indirect immunofluorescence using an anti-AP2 antibody (red). Scale bar: 20 mm.
cytoplasmic tail of the cargoes (Bonnemaison et al., 2013). In fact, AP1 is believed to be a central player in CCV formation at the TGN affecting a wide variety of transmembrane proteins (Hirst et al., 2012). Additionally, AP1 has also been linked to retrograde transport wherein several receptors were enriched in peripheral compartments upon AP1 depletion (Hirst et al., 2012). The μ1B subunit isoform specifically expresses in epithelial cells forming the AP1B complex which is involved in basolateral sorting of cargo such as the low-density lipoprotein receptor (LDLR) (Gonzalez and Rodriguez-Boulan, 2009). Mutations in AP1 have been linked to X-linked mental retardation (Tarpey et al., 2006), MEDNIK syndrome (Montpetit et al., 2008), and Pettigrew syndrome (Cacciagli et al., 2014).
AP2 This adaptor is expressed ubiquitously and is essential for mammalian development (Mitsunari et al., 2005). AP2 is capable of binding the plasma membrane-enriched lipids PI (4,5)P2 and/or PI(3,4,5)P3 and is a key element for CME. Upon recruitment to the plasma membrane, AP2 undergoes a conformational change from a ‘lock’ state to an ‘open’ state that exposes the clathrin-binding motif on the β2 hinge (Collins et al., 2002). The interaction of AP2 with cargo
stabilizes the open state, and thus facilitating formation and cargo inclusion in clathrin-coated pits (CCPs) (Kelly et al., 2014). The α and β appendage domains interact with other clathrin adaptors and accessory proteins, such as Epsin, ARH, and AP180 to consolidate CCVs. The initial presumption that AP2 was essential for CME was challenged by the observation that although depletion of AP2 reduced CCV formation and specifically blocked transferrin-receptor internalization, epidermal growth factor receptor (EGFR), and LDLR internalization still progressed via CME (Motley et al., 2003).
AP3 Organisms deficient in this adaptor survive to adulthood; however, they have defects in lysosome and lysosomal-related organelle biosynthesis (Yang et al., 2000). AP3 membrane localization is similar to AP1, but with different degree of colocalization with clathrin, suggesting localization/functional differences (Peden et al., 2004). Selective depletion of AP3 subunits resulted in indirect trafficking of lysosomal membrane proteins namely Lamp-1, Lamp-2, and CD63 via a plasma membrane-to-endosome pathway (Dell’Angelica et al., 1999). The β3 and μ3 subunits of the AP3 complex in metazoans have two isoforms: the isoform A is ubiquitously present, while the
Interorganellar Communication: Components: Adaptor Proteins: Inter-Organelle Traffic Controllers
isoform B is specifically expressed in neuronal and neuroendocrine tissues resulting in two AP3 complexes. The ubiquitous AP3A sorts its cargo (Lamp-1, Lamp-2, CD63) to the lysosome, while neuronal AP3B sorts its cargo (Chloride Channel-3, Vesicular glutamate transporter 1, vesicular GABA transporter, and many more synaptic proteins) to synaptic vesicles (Nakatsu and Ohno, 2003). Mutations in the ubiquitous AP3A complex results in Hermansky–Pudlak syndrome type-2, while mutations in the neuronal AP3B complex results in neurological symptoms such as epilepsy and hyperactivity (Badolato and Parolini, 2007; Nakatsu and Ohno, 2003).
VHS
GGA1
GAT
GAE
UIM CBM
Epsin1
ENTH
EpsinR
ENTH
Eps15
E H
Dab2
PTB
ARH
PTB
-Arrestin1
Arr-N
639
NPF
ABM(8)
551
Appendage binding site
E H
453
625 E H
Coiled-coil
12
897
5
1
770
308
AP4 This adaptor is also ubiquitously expressed in all tissues, but is not essential for development as evidenced by the existence of healthy knockout mice (Matsuda et al., 2008). AP4 localizes to the TGN, but does not colocalize with AP1 perhaps due to the lack of CBMs (Hirst et al., 2013). Notably, perturbation of CCVassembly increased AP4 vesicle formation without changing the expression levels of AP4, suggesting a dynamic compensatory mechanism for TGN trafficking (Hirst et al., 2012). Hence, AP4 is speculated to traffic at least some cargo also transported by CCV. Surprisingly, AP4 recognizes various types of sorting motifs on its cargoes, including canonical tyrosine motif on LDLR, YX[F/Y/L][F/L]E motifs on amyloid precursor protein (APP), FI (Phenylalanine-Isoleucine) motif on Furin, and unconventional basolateral sorting signal (EQFPHLAFWQDLG(Bresciani et al., 1997)) on M6PR (Simmen et al., 2002). AP4 complex was required for the basolateral trafficking of these cargoes. Additionally, AP4 is also involved in basolateral trafficking in neurons where its cargoes are LDLR, AMPA (α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid), and δ2 glutamate receptors (Matsuda et al., 2008). AP4-deletion showed aberrant accumulation of APP in TGN rather than in the endosomes implying a role for the AP4 complex in TGN-toendosomes trafficking (Burgos et al., 2010). Deficiency of AP4 in humans has been shown to cause intellectual disability and progressive spasticity that gradually develops to hereditary spastic paraplegia (HSP), suggesting that AP4 participates in human brain development (Hirst et al., 2013).
AP5 This adaptor is the most recently identified, ubiquitous, heterotetrameric adaptor protein complex. Despite having less than 10% protein sequence identity with other APs, the secondary structure prediction of AP5 subunits is similar to the counterparts in other APs (Hirst et al., 2013). AP5 does not associate with clathrin and was shown to localize on late endosomal compartments. Surprisingly, RNAi knock-down of AP5 resulted in cation-independent M6PR trapping in swollen early endosomes, albeit AP5 localization in late endosomes (Hirst et al., 2013). AP5 endosomal trafficking route and cargoes are yet to be described. Mutations in AP5 ζ subunits, similar to AP4 subunits, lead to HSP. It is possible that the same cargo is transported by AP4 and AP5 at different points along the vesicle trafficking pathway.
AP180
ANTH
CALM
ANTH
Arr-C
1
418 12
1
916 652
Figure 2 Monomeric adaptors. Domain/motif organization of several monomeric adaptor complexes is shown. ABM, AP2-Binding Motif; ANTH, AP180 N-terminal Homology domain; Arr, Arrestin fold; CBM, Clathrin-binding motif; EH, Eps15 homology domain; ENTH, Epsin Nterminal Homology domain; GAE, Gamma-adaptin ear homology domain; GAT, GGA and Tom; NPF, Asparagine (N)-Proline (P)Phenylalanine (F) peptide; PTB, Phosphotyrosine binding domain; UIM: Ubiquitin interacting motif; VHS, Vps27, Hrs, and STAM.
GGAs GGAs were identified in early 2000s, and as their name indicates, they are localized at the Golgi apparatus (TGN), but they can also be found on endosomes (Ghosh and Kornfeld, 2004). GGAs are ubiquitously expressed and evolutionarily conserved, with three GGAs in human and mouse, two in yeast, and one in nematode and fruit fly. The existence of distinct GGA functions of the three GGAs has been demonstrated in mouse models. GGA2 null mice are embryonic or neonatal lethal; double mutant of GGA1 and GGA3 result in neonatal lethal, indicating that GGAs are required for developmental process and not fully redundant (Govero et al., 2012). There are four folding modules in GGAs, from N-terminal to C-terminal: VHS (Vps27, Hrs, and STAM), GAT (GGA and Tom), hinge, and GAE (gamma-adaptin ear homology) domain (Figure 2). The GAT domain has an N-terminal α-helical hook subdomain which interacts with Arf-1 and consequently is recruited to the TGN (Collins et al., 2003; Shiba et al., 2003). Like the hinge region of β subunits in AP1–3, the hinge region of GGAs also contains an LФXФ[D/E] CBM, that allows the GGAs to bind clathrin (Bonifacino, 2004). This indicated that GGAs are involved in CM-trafficking from the TGN. The C-terminus of the GAT domain binds ubiquitin and has been implicated in sorting of ubiquitinated-(Ub)-cargo from TGN to the vacuole in yeast (Prag et al., 2005). The role of GGAs in trafficking Ub-cargo in mammals has not been fully explored. GGAs also recognize cargo via the VHS domain which binds a DXXLL motif located in the cytosolic tail of the transported proteins, called acidic-cluster-dileucine signal. The motif consists of an aspartate residue followed by two leucines
454
Interorganellar Communication: Components: Adaptor Proteins: Inter-Organelle Traffic Controllers
at the third downstream position (Puertollano et al., 2001; Doray et al., 2002). The D and dileucine bind to the sixth to eighth α-helices of VHS where an electropositive pocket and two hydrophobic pockets reside (Misra et al., 2000; Shiba et al., 2002). In addition, acidic-cluster-dileucine signals also have been found in the hinge region of human GGA1 and GGA3, suggesting the formation of intramolecular complexes competing with cargo recognition; i.e., with auto-inhibitory properties (Bonifacino, 2004). The cargoes trafficked by GGAs include M6PRs and Sortilin (Puertollano et al., 2001; Hirst et al., 2012). Notably GGAs and AP1 cooperate for the trafficking of M6PR from the TGN to the lysosome. The GAE domain homologous to the γ-adaptin ear of AP1, also serves as a platform for binding to other endocytic accessory proteins with a DFGXФ motif, namely γ-synergin, p56 , Rabaptin 5 and Clint/EpsinR in mammals (Bonifacino, 2004). In addition, evidence showed the involvement of GGA1 in the retrograde trafficking of β-site APP-cleaving enzyme 1 from endosomes to the TGN while GGA3 plays a role in the targeting of BACE1 to the lysosomes (Tan and Evin, 2012).
The Epsin and Eps15 Families The Epsin Family These endocytic adaptors including Epsin and EpsinR are highly conserved in yeast and most vertebrates (Sen et al., 2012). There are three Epsin paralogs in humans (Rosenthal et al., 1999; Chen et al., 1999; Spradling et al., 2001). While Epsin-1 and 2 are ubiquitously expressed, Epsin3 has a more restricted expression pattern being confined to migratory keratinocytes, stomach cells, and cancer cells (Rosenthal et al., 1999; Chen et al., 1999; Coon et al., 2011; Spradling et al., 2001). The N-terminus of Epsins has a highly conserved Epsin N-terminal homology domain (ENTH) (Figure 2) that binds PI(4,5)P2, a lipid enriched at endocytic and signaling sites (Sen et al., 2012). This interaction triggers a conformational change in the ENTH domain resulting in the formation of an additional α-helix, helix 0 which inserts into the plasma membrane inducing membrane curvature (Ford et al., 2002). This structural transformation of the ENTH domain is believed to constitute the initial step of membrane invagination required for the formation of CCPs (Horvath et al., 2007; LegendreGuillemin et al., 2004). C-terminal to the ENTH domain is an unstructured region containing multiple motifs required for engaging cargo and endocytic machinery. Each Epsin has two or three UIMs (ubiquitin interacting motifs) able to bind ubiquitinated-cargo (Madshus, 2006). Epsins also bind clathrin via 1 (e.g., yeast) or 2 (e.g., mammalian) clathrin-bindng motifs (CBMs) with the consensus LФZФZ, where L is leucine, Ф is any hydrophobic residue, and Z is any polar residue. In higher eukaryotes, the CBMs flank 3–8 DP[W/F] (aspartate, proline, and tryptophan/phenylalanine) repeats that bind to AP2 (Figure 1B). Lastly, the C-terminus houses 2–3 NPFs (asparagine, proline, phenylalanine) which bind to Eps15 homology (EH) domain containing proteins, namely Eps15 and REPS1/2. In fact, Eps15 and Epsin together have been implicated in EGFR internalization (Sigismund et al., 2005).
EGFR is a receptor tyrosine kinase belonging to the ErbB family which plays a major role in proliferation and differentiation. Interestingly, two different routes have been proposed for Epsin-bound activated EGFR. Classical route entails EGFR ubiquitination and recruitment to CCPs for internalization (Bertelsen et al., 2011; Hawryluk et al., 2006). However, at high EGF concentrations, a clathrin-independent, Eps15- and Epsin-dependent internalization was also found (Sigismund et al., 2005). Interestingly, it has been reported that Epsin binding to ubiquitin negatively regulates clathrin binding (Chen and De Camilli, 2005), supporting the existence of an Epsin-mediated clathrin-independent route. Notably, the fate of EGFR differs based on the internalization route, wherein the clathrin-dependent route and the clathrinindependent route favor recycling and degradation of the EGFR receptor, respectively (Sigismund et al., 2005). While Epsin can contribute to EGFR signaling termination, it is required for activation of the notch signaling pathway (Wang and Struhl, 2004). Elegant studies done in flies to study the juxtacrine notch signaling pathway has shown that Epsindependent Delta endocytosis in the signal-sending cell is required for signaling (Wang and Struhl, 2004). Supporting evidence comes from Epsin-deficient mutants in flies, worms, and mice where defects in notch signaling pathway such as cardiovascular development, germline, and neural tube differentiation were observed (Tian et al., 2004; Chen et al., 2009). Other Epsin-specific cargoes that undergo UIM-mediated internalization are the epithelial sodium channel ENaC (Wang et al., 2006) and the Vascular Endothelial Growth Factor Receptor-2 (Tessneer et al., 2013). Progression from an initiated pit into a vesicle depends on reaching a critical mass of cargo and endocytic components. Epsin-1 contributes to this checkpoint which when unsatisfied will result in the destabilization and abortion of pits (Sen et al., 2012). In addition to serving as an endocytic adaptor protein, Epsins are proposed to function as stabilizers of the endocytic network. Yeast Epsins have also shown to play an analogous role in expanding and sustaining the endocytic network by mediating protein–protein interaction via their NPFs and UIMs (Dores et al., 2010). The Epsin family of adaptor proteins has been recognized as regulators of cancer progression (Tessneer et al., 2013). Epsins are upregulated in skin, breast, prostate, pancreatic, and lung cancers (Tessneer et al., 2013; Sen et al., 2012). The enhanced invasion potential of Epsin-overexpressing cells is attributed to the nonclassical Epsin function in RhoGTPase signaling regulation that leads to enhanced cell migration and invasion (Coon et al., 2010). In addition, Epsin’s role in angiogenesis by regulating VEGFR2 levels has been demonstrated, and considered as a potential treatment of cancer. Knockout of Epsin in Lewis Lung Cancer model effectively reduced tumorigenesis by misregulation of VEGFR2 and angiogenesis (Tessneer et al., 2013).
The EpsinR Family These adaptors bear an ENTH domain similar to Epsin (Figure 2), but differs in lipid specificity as it binds PI(4)P and
Interorganellar Communication: Components: Adaptor Proteins: Inter-Organelle Traffic Controllers
therefore, localizes to the TGN. EpsinR (epsin-related protein) has an AP1-binding motif [D/E]FxD[F/W] and four identified CBM of the consensus DФF. Some of the cargo proteins under control of EpsinR include M6PR and the SNARE protein Vtib (Hirst et al., 2004; Mills et al., 2003). This adaptor is believed to play a role in cargo recycling to the TGN.
Eps15 Family The presence of UIMs in Eps15 (Figure 2) indicates that, similar to Epsin, this protein is capable of ubiquitinated-cargo recognition (although lacks coat-binding determinants). Eps15 contains NPF-binding, EH domains in the N-terminus which also interacts with PI(3,5)P2 (Naslavsky et al., 2007). The Eps15 coiled-coiled region following the EH domain is involved in protein dimerization (van Bergen En Henegouwen, 2009) and along with the UIMs have been shown to define distinct cargo-specificity (c-MET and EGFR) (Parachoniak and Park, 2009; Sigismund et al., 2005; De Melker et al., 2004; Polo et al., 2002). The C-terminus also houses an array of AP2-interacting motifs, and an AP1-interacting motif. Other members of this family include Eps15R which is structurally similar to Eps15 and Eps15b, but lacks the N-terminal EH-domains (van Bergen En Henegouwen, 2009). In addition to its involvement in endocytosis, Eps15 cooperates with AP1 in sorting cargo at the Golgi apparatus. Indeed, mutation of the AP1 binding site on Eps15 impaired the secretion of nascent secretory proteins and TGN-to-endosomal transport of mannose-6-phosphate receptor (van Bergen En Henegouwen, 2009).
AP180 and Clathrin Assembly Lymphoid Myeloid Leukemia The ubiquitous clathrin assembly lymphoid myeloid leukemia (CALM) protein and its neuronal counterpart the assembly protein 180 (AP180) have a characteristic ANTH (AP180 N-terminal homology) domain (structurally related to the ENTH domain) (Figure 2). AP180/CALM binds to PI(4,5)P2, but does not induce membrane deformation (Stahelin et al., 2003). The C-terminus of AP180/CALM has a subtype of CBMs (DL[L/F] – Aspartate, Leucine[Leucine/Phenylalanine]), and several NPF motifs. Except for the yeast homologs, AP180s contain multiple AP2-binding motifs. AP180s are found conserved in yeast (Yap1801, Yap1802), Caenorhabditis elegans (unc-11), and in Drosophila melanogaster (Lap) while CALM is only present in mammalian cells. CALM and AP180 play a conserved role in the endocytosis of SNAREs, specifically vesicle-associated membrane proteins (VAMP)/ R-SNAREs (Moshkanbaryans et al., 2014). Depletion of AP180 and CALM in hippocampal neurons resulted in aberrant retention of synaptobrevin2/VAMP at the neuronal surface, while the abundant synaptic vesicular protein vGLUT1 was unaffected. CALM also plays a role in VAMP3 and VAMP8 internalization in non-neuronal cells. Study of the CALM– SNARE interface by X-Ray crystallography revealed that the N-terminal half of the SNARE domain binds the ANTH
455
domain in a manner similar to the SNARE complex formation (Moshkanbaryans et al., 2014). The involvement of CALM and AP180 in neurodegenerative diseases has gained enormous support in recent years. Genome wide-association study identified alterations in the PICALM gene encoding CALM as a genetic risk factor in the onset of Alzheimer's disease (AD) (Moshkanbaryans et al., 2014). It has also been implicated in affecting cognitive function with aging. A recent study directly correlated the levels of CALM to the levels of pathogenic Aβ complexes in AD, by affecting the endocytosis and localization s-secretase involved in the production of toxic Aβ (Kanatsu et al., 2014). On the contrary, cytotoxicity of Aβ peptide was reduced in yeast, worms, and in mice cortical neurons by overexpressing AP180 family of proteins (Moshkanbaryans et al., 2014). Although these studies are contradictory, they emphasize the role of CALM in APP processing highlighting its therapeutic potential for treatment of AD.
PTB Domain Containing Family (DAB, ARH, and Numb) This family of adaptor proteins is comprised of neuronal disabled-1 (Dab1), ubiquitous Dab2, autosomal recessive hypercholesterolemia (ARH), and numb which share a common phosphotyrosine binding domain (PTB) (Figure 2). This fold is commonly found in signaling molecules and binds to phosphorylated-tyrosines. However, the PTB domain of this adaptor family specifically recognizes the tyrosine-based signal (FX)NPXY on the cytoplasmic tail of receptor proteins (Mishra et al., 2002). The PTB domain structure has been solved and shows simultaneous binding of cargo and PI(4,5)P2 (Dvir et al., 2012; Stolt et al., 2003) and hence regulates vesicle budding from the plasma membrane. These adaptors have been linked to CME via their interaction with AP2 and/or clathrin directly. Dab2 and ARH interact with AP2 and clathrin, while numb and Dab1 only interact with AP2. Additionally, Dab2 has multiple NPF motifs capable of binding to EH domain containing proteins such as Eps15 (Figure 2). Dab2 and ARH have been reported to be functionally redundant for CME of LDLR (Keyel et al., 2006; Maurer and Cooper, 2006). However, while ARH-mediated LDLR internalization required AP2, Dab2-mediated LDLR internalization does not (Keyel et al., 2006; Maurer and Cooper, 2006). The AP2-binding mechanism is very distinct for ARH as it engages AP2 by binding the β2 appendage while the others bind to the α-appendage of AP2 (Mishra et al., 2005). Dab2 was also required for albumin internalization (Maurer and Cooper, 2005) by recognizing the (ФX)NPXY motif on the cytoplasmic tail of the albumin receptor, megalin (Gallagher et al., 2004). PTB domain containing adaptor proteins bind to the amyloid precursor protein family as a (YX)NPXY motif is conserved among members (Howell et al., 1999; Homayouni et al., 1999). Both numb and Dab2 have been implicated in the endocytosis of integrins which also contains NPXY motifs in their cytoplasmic tails (Bridgewater et al., 2012). Mutations in ARH lead to defective endocytosis of LDLR and in consequence to hypercholesterolemia. Dab2 is unable to compensate for the loss of ARH, due to the low levels of
456
Interorganellar Communication: Components: Adaptor Proteins: Inter-Organelle Traffic Controllers
expression of Dab2 in hepatocytes. The role of Dab and Numb in APP processing and production of Aβ has been exploited and depletion of these adaptor proteins was shown to reduce the levels of Aβ production (Xie et al., 2012).
Arrestin Family The arrestin family of adaptor proteins shares a common arrestin-fold domain and consists of α-arrestins, a group of vacuolar protein sorting (Vps) 26 related proteins, visual and nonvisual arrestins or β-arrestins. The members of this family show evolutionary conservation from humans, flies, and worms to yeasts (Aubry et al., 2009). Arrestin family in humans has 14 members: 6 α-arrestins, 4 visual and β-arrestins, and 4 Vps26 genes. Phylogenetic analysis suggests that βarrestins, the best-studied members of this clan, branched from the α-arrestins and coevolved with G-protein coupled receptors (GPCRs) to mediate their internalization (Alvarez, 2008). The Arrestins were named for their scaffolding function in binding and terminating G-protein coupled signaling. However, many more functions of this protein family in assisting the internalization of cargoes including GPCRs have been described since. In addition to functioning in signaling regulation of GPCRs by binding to their cytoplasmic tails, β-arrestins’ ability to act as adaptor proteins became clear as they can bind clathrin and AP2 via motifs in their C-termini (Owen et al., 2004; Figure 2). Also, arrestin interaction with phosphoinosities at the plasma membrane is required for recruitment of the cargo into endocytic vesicles (Gaidarov et al., 1999). Structural studies have shed light on the mechanism of action of β-arrestins. In an inactive phosphorylated state, intramolecular interactions between the N- and C-domains of arrestins lead to the shielding of a core of polar residues involved in the recognition of cargo and binding other elements of the endocytic machinery. Arrestin binding to an activated phosphorylated GPCR and subsequent arrestin dephosphorylation (by an unknown phosphatase) disrupts the intramolecular interactions in arrestins and allows binding to the endocytic machinery (Kang et al., 2014). Arrestins internalize cargoes from the plasma membrane and traffic them to the early endosomes by binding phosphorylated Ser/Thr clusters in their C-termini (Kang et al., 2014). The rates of receptor recycling and degradation depend on the strength of receptor–arrestin interaction. For example, Class-A receptors (β2 Adrenergic receptor, vasopressin 1a) with weak interaction are recycled back quickly, whereas Class-B receptors (angiotensin II type 1A receptor, vasopressin 2 receptor) with strong interaction are recycled slowly (Kang et al., 2014). Arrestins get ubiquitinated by the E3 ubiquitin-ligase Mdm2 and their ability to recruit deubiqutinases (DUBs) and undergo deubiquitination also leads to differential consequences. While Class-A receptor–arrestin complex recruits DUBs leading to receptor recycling, Class-B receptors do not have a conformation that supports DUB recruitment and hence leads to stable ubiqutination and degradation (Kang et al., 2014). Subsequently, arrestin–GPCR interaction strength also affects signaling. Specifically Class-B receptors result in the formation of stable signalosomes due to a strong interaction.
Opsin/rhodopsin family of GPCRs serve as cargoes for the visual arrestins. In addition to classic GPCRs, β-arrestins also interact with activated/phosphorylated unconventional GPCRs (Disheveled and Smoothened), Receptor Tyrosine Kinases (IGF-1R, TGFβIIIR), and ion channels (NHE1/5, Ca(v)1, TRPV4, Nicotinic cholinergic receptor) to mediate their internalization. The vps26-related proteins are structurally more similar to β-arrestins than to other members of the family. Vps26A/B, 29, and 35 are four mammalian members of the Vps family part of the so-called retromer complex. The retromer is involved in endosomes-to-TGN cargo transport. However, Vps26 is not directly involved in cargo recognition and therefore, cannot be considered an adaptor. α-Arrestins lack the helix 1 that forms the polar core of inactive β-arrestins. Another major difference is that the C-terminus of α-arrestins contains PPXY motifs instead of the clathrin- and AP2-binding sites found in β-arrestins. Members of the α-arrestin family include the 10 yeast ARTS (Arrestin related trafficking adaptors) and the 6 α-arrestin (ARRDC 1–5; Arrestin domain containing proteins and TXNIP Thioredoxininteracting protein) mammalian proteins. ARTS in yeast have been shown to function as cargo adaptors for the HECT ubiquitin-ligase Rsp5, i.e., facilitating cargo ubiquitination and internalization (Lin et al., 2008). Similar to ARTS in yeast, the role for ARRDCs in mammalian cells as E3 ligase adaptors has been well-established linking GPCR to E3 HECT ubiquitin ligases Nedd4, AIP4, WWP1, and WWP2. In addition, ARTS have been shown to function as secondary adaptors to recruit cargo, β-arrestin-E3 ligase complex on early endosomes (Nabhan et al., 2010). Further, they interact with components of the ESCRT complex and hence are implicated in multivesicular body/vacuolar sorting. GPCRs represent the largest family of receptors (B1000 members) that localize on the cell surface and serve as target for about 40% of the drugs available. Therefore, arrestins are considered valuable targets to control GPCR signaling regulation. Further, arrestin involvement in pathological conditions has been substantially documented. Mutation of visual arrestins is associated with blindness following photoreceptor death due to impaired receptor desensitization. An alternative mechanism leading to blindness by stable association of visual arrestin to the photoreceptor rhodopsin has been suggested (Chen et al., 2006). Overexpression of β-arrestins in AD contributes to the formation of extracellular neuritic plaques containing the amyloid-β (Aβ) peptide. This key pathogenic component of AD was reduced by disrupting the interaction between β-arrestin and β2-adrenergic receptor which in turn lowers Aβ production (Jiang et al., 2013). β-Arrestins participate in signaling pathways mediated by Smoothened and Frizzled are required for the maintenance and onset of chronic myeloid leukemia (CML). Recently, targeting β-arrestin by RNA-aptamers was effective in reducing tumorigenic growth in CML models and patient samples (Kotula et al., 2014).
The Adaptor Protein Network As described above, adaptors bear multiple-binding motifs that mediate several types of protein–protein interactions. This
Interorganellar Communication: Components: Adaptor Proteins: Inter-Organelle Traffic Controllers
promiscuous binding behavior has two general consequences: (1) it allows low-affinity (micromolar range) protein–protein interactions to sustain stable, but dynamic, multi-protein structures, and (2) it confers these massive complexes with classical network properties. For example, it is clear that AP2 at the plasma membrane (or AP1 at the TGN/endosomal level) along with clathrin constitute the ‘hubs’ (maximal connectivity centers) of the adaptor network. This observation explains why these key components are regulatory targets allowing the fast assembly or disassembly of the adaptor network.
See also: Interorganellar Communication: Interplay and Processes: Clathrin and Clathrin-Dependent Endocytosis
References Alvarez, C.E., 2008. On the origins of arrestin and rhodopsin. BMC Evolutionary Biology 8, 222. Aubry, L., Guetta, D., Klein, G., 2009. The arrestin fold: Variations on a theme. Current Genomics 10, 133–142. Badolato, R., Parolini, S., 2007. Novel insights from adaptor protein 3 complex deficiency. Journal of Allergy and Clinical Immunology 120, 735–741. quiz 742−743. van Bergen En Henegouwen, P.M., 2009. Eps15: A multifunctional adaptor protein regulating intracellular trafficking. Cell Communication and Signaling 7, 24. Bertelsen, V., Sak, M.M., Breen, K., et al., 2011. A chimeric pre-ubiquitinated EGF receptor is constitutively endocytosed in a clathrin-dependent, but kinaseindependent manner. Traffic 12, 507–520. Bonifacino, J.S., 2004. The GGA proteins: Adaptors on the move. Nature Reviews Molecular Cell Biology 5, 23–32. Bonnemaison, M.L., Eipper, B.A., Mains, R.E., 2013. Role of adaptor proteins in secretory granule biogenesis and maturation. Front Endocrinol (Lausanne) 4, 101. Bresciani, R., Denzer, K., Pohlmann, R., Von Figura, K., 1997. The 46 kDa mannose-6phosphate receptor contains a signal for basolateral sorting within the 19 juxtamembrane cytosolic residues. Biochemical Journal 327 (Pt 3), 811–818. Bridgewater, R.E., Norman, J.C., Caswell, P.T., 2012. Integrin trafficking at a glance. Journal of Cell Science 125, 3695–3701. Burgos, P.V., Mardones, G.A., Rojas, A.L., et al., 2010. Sorting of the Alzheimer's disease amyloid precursor protein mediated by the AP-4 complex. Developmental Cell 18, 425–436. Cacciagli, P., Desvignes, J.P., Girard, N., et al., 2014. AP1S2 is mutated in X-linked Dandy-Walker malformation with intellectual disability, basal ganglia disease and seizures (Pettigrew syndrome). European Journal of Human Genetics 22, 363–368. Chen, H., De Camilli, P., 2005. The association of epsin with ubiquitinated cargo along the endocytic pathway is negatively regulated by its interaction with clathrin. Proceedings of the National Academy of Sciences of the United States of America 102, 2766–2771. Chen, H., Ko, G., Zatti, A., et al., 2009. Embryonic arrest at midgestation and disruption of Notch signaling produced by the absence of both Epsin 1 and Epsin 2 in mice. Proceedings of the National Academy of Sciences of the United States of America 106, 13838–13843. Chen, H., Slepnev, V.I., Di Fiore, P.P., De Camilli, P., 1999. The Interaction of Epsin and Eps15 with the clathrin adaptor AP-2 is inhibited by mitotic phosphorylation and enhanced by stimulation-dependent dephosphorylation in nerve terminals. Journal of Biological Chemistry 274, 3257–3260. Chen, J., Shi, G., Concepcion, F.A., Xie, G., Oprian, D., 2006. Stable rhodopsin/ arrestin complex leads to retinal degeneration in a transgenic mouse model of autosomal dominant retinitis pigmentosa. Journal of Neuroscience 26, 11929–11937. Collins, B.M., McCoy, A.J., Kent, H.M., Evans, P.R., Owen, D.J., 2002. Molecular architecture and functional model of the endocytic AP2 complex. Cell 109, 523–535. Collins, B.M., Watson, P.J., Owen, D.J., 2003. The structure of the GGA1-GAT domain reveals the molecular basis for ARF binding and membrane association of GGAs. Developmental Cell 4, 321–332.
457
Coon, B.G., Burgner, J., Camonis, J.H., Aguilar, R.C., 2010. The Epsin family of endocytic adaptors promotes fibrosarcoma migration and invasion. Journal of Biological Chemistry 285, 33073–33081. Coon, B.G., Direnzo, D.M., Konieczny, S.F., Aguilar, R.C., 2011. Epsins' novel role in cancer cell invasion. Communicative & Integrative Biology 4, 95–97. De Melker, A.A., Van Der Horst, G., Borst, J., 2004. c-Cbl directs EGF receptors into an endocytic pathway that involves the ubiquitin-interacting motif of Eps15. Journal of Cell Science 117, 5001–5012. Dell’Angelica, E.C., Shotelersuk, V., Aguilar, R.C., Gahl, W.A., Bonifacino, J.S., 1999. Altered trafficking of lysosomal proteins in Hermansky-Pudlak syndrome due to mutations in the β3A subunit of the AP-3 adaptor. Molecular Cell 3, 11–21. Doray, B., Bruns, K., Ghosh, P., Kornfeld, S., 2002. Interaction of the cationdependent mannose 6-phosphate receptor with GGA proteins. Journal of Biological Chemistry 277, 18477–18482. Dores, M.R., Schnell, J.D., Maldonado-Baez, L., Wendland, B., Hicke, L., 2010. The function of yeast Epsin and Ede1 ubiquitin-binding domains during receptor internalization. Traffic 11, 151–160. Dvir, H., Shah, M., Girardi, E., et al., 2012. Atomic structure of the autosomal recessive hypercholesterolemia phosphotyrosine-binding domain in complex with the LDL-receptor tail. Proceedings of the National Academy of Sciences of the United States of America 109, 6916–6921. Ford, M.G., Mills, I.G., Peter, B.J., et al., 2002. Curvature of clathrin-coated pits driven by Epsin. Nature 419, 361–366. Gaidarov, I., Krupnick, J.G., Falck, J.R., Benovic, J.L., Keen, J.H., 1999. Arrestin function in G protein-coupled receptor endocytosis requires phosphoinositide binding. EMBO Journal 18, 871–881. Gallagher, H., Oleinikov, A.V., Fenske, C., Newman, D.J., 2004. The adaptor disabled-2 binds to the third psi xNPxY sequence on the cytoplasmic tail of megalin. Biochimie 86, 179–182. Ghosh, P., Kornfeld, S., 2003. AP-1 binding to sorting signals and release from clathrin-coated vesicles is regulated by phosphorylation. Journal of Cell Biology 160, 699–708. Ghosh, P., Kornfeld, S., 2004. The GGA proteins: Key players in protein sorting at the trans-Golgi network. European Journal of Cell Biology 83, 257–262. Gonzalez, A., Rodriguez-Boulan, E., 2009. Clathrin and AP1B: Key roles in basolateral trafficking through trans-endosomal routes. FEBS Letters 583, 3784–3795. Govero, J., Doray, B., Bai, H., Kornfeld, S., 2012. Analysis of Gga null mice demonstrates a non-redundant role for mammalian GGA2 during development. PLoS One 7, e30184. Hawryluk, M.J., Keyel, P.A., Mishra, S.K., et al., 2006. Epsin 1 is a polyubiquitinselective clathrin-associated sorting protein. Traffic 7, 262–281. Hirst, J., Borner, G.H., Antrobus, R., et al., 2012. Distinct and overlapping roles for AP-1 and GGAs revealed by the "knocksideways" system. Current Biology 22, 1711–1716. Hirst, J., Irving, C., Borner, G.H., 2013. Adaptor protein complexes AP-4 and AP-5: New players in endosomal trafficking and progressive spastic paraplegia. Traffic 14, 153–164. Hirst, J., Miller, S.E., Taylor, M.J., Von Mollard, G.F., Robinson, M.S., 2004. EpsinR is an adaptor for the SNARE protein Vti1b. Molecular Biology of the Cell 15, 5593–5602. Homayouni, R., Rice, D.S., Sheldon, M., Curran, T., 1999. Disabled-1 binds to the cytoplasmic domain of amyloid precursor-like protein 1. Journal of Neuroscience 19, 7507–7515. Horvath, C.A., Vanden Broeck, D., Boulet, G.A., Bogers, J., De Wolf, M.J., 2007. Epsin: Inducing membrane curvature. International Journal of Biochemistry & Cell Biology 39, 1765–1770. Howell, B.W., Lanier, L.M., Frank, R., Gertler, F.B., Cooper, J.A., 1999. The disabled 1 phosphotyrosine-binding domain binds to the internalization signals of transmembrane glycoproteins and to phospholipids. Molecular and Cellular Biology 19, 5179–5188. Huang, K.M., D'Hondt, K., Riezman, H., Lemmon, S.K., 1999. Clathrin functions in the absence of heterotetrameric adaptors and AP180-related proteins in yeast. EMBO Journal 18, 3897–3908. Jiang, T., Yu, J.T., Tan, M.S., Zhu, X.C., Tan, L., 2013. Beta-Arrestins as potential therapeutic targets for Alzheimer's disease. Molecular Neurobiology 48, 812–818. Kanatsu, K., Morohashi, Y., Suzuki, M., et al., 2014. Decreased CALM expression reduces Abeta42 to total Abeta ratio through clathrin-mediated endocytosis of gamma-secretase. Nature Communications 5, 3386. Kang, D.S., Tian, X.,Benovic, J.L., 2014. Role of beta-arrestins and arrestin domaincontaining proteins in G protein-coupled receptor trafficking. Current Opinions in Cell Biology 27, 63–71.
458
Interorganellar Communication: Components: Adaptor Proteins: Inter-Organelle Traffic Controllers
Kelly, B.T., Graham, S.C., Liska, N., et al., 2014. Clathrin adaptors. AP2 controls clathrin polymerization with a membrane-activated switch. Science 345, 459–463. Keyel, P.A., Mishra, S.K., Roth, R., et al., 2006. A single common portal for clathrin-mediated endocytosis of distinct cargo governed by cargo-selective adaptors. Molecular Biology of the Cell 17, 4300–4317. Kotula, J.W., Sun, J., Li, M., et al., 2014. Targeted disruption of beta-arrestin 2mediated signaling pathways by aptamer chimeras leads to inhibition of leukemic cell growth. PLoS One 9, e93441. Lefkir, Y., De Chassey, B., Dubois, A., et al., 2003. The AP-1 clathrin-adaptor is required for lysosomal enzymes sorting and biogenesis of the contractile vacuole complex in Dictyostelium cells. Molecular Biology of the Cell 14, 1835–1851. Legendre-Guillemin, V., Wasiak, S., Hussain, N.K., Angers, A., McPherson, P.S., 2004. ENTH/ANTH proteins and clathrin-mediated membrane budding. Journal of Cell Science 117, 9–18. Lin, C.H., MacGurn, J.A., Chu, T., Stefan, C.J., Emr, S.D., 2008. Arrestin-related ubiquitin-ligase adaptors regulate endocytosis and protein turnover at the cell surface. Cell 135, 714–725. Madshus, I.H., 2006. Ubiquitin binding in endocytosis – How tight should it be and where does it happen? Traffic 7, 258–261. Matsuda, S., Miura, E., Matsuda, K., et al., 2008. Accumulation of AMPA receptors in autophagosomes in neuronal axons lacking adaptor protein AP-4. Neuron 57, 730–745. Mattera, R., Boehm, M., Chaudhuri, R., Prabhu, Y., Bonifacino, J.S., 2011. Conservation and diversification of dileucine signal recognition by adaptor protein (AP) complex variants. Journal of Biological Chemistry 286, 2022–2030. Maurer, M.E., Cooper, J.A., 2005. Endocytosis of megalin by visceral endoderm cells requires the Dab2 adaptor protein. Journal of Cell Science 118, 5345–5355. Maurer, M.E., Cooper, J.A., 2006. The adaptor protein Dab2 sorts LDL receptors into coated pits independently of AP-2 and ARH. Journal of Cell Science 119, 4235–4246. Meyer, C., Zizioli, D., Lausmann, S., et al., 2000. mu1A-adaptin-deficient mice: Lethality, loss of AP-1 binding and rerouting of mannose 6-phosphate receptors. EMBO Journal 19, 2193–2203. Mills, I.G., Praefcke, G.J., Vallis, Y., et al., 2003. EpsinR: An AP1/clathrin interacting protein involved in vesicle trafficking. Journal of Cell Biology 160, 213–222. Mishra, S.K., Keyel, P.A., Edeling, M.A., et al., 2005. Functional dissection of an AP-2 beta2 appendage-binding sequence within the autosomal recessive hypercholesterolemia protein. Journal of Biological Chemistry 280, 19270–19280. Mishra, S.K., Keyel, P.A., Hawryluk, M.J., et al., 2002. Disabled-2 exhibits the properties of a cargo-selective endocytic clathrin adaptor. EMBO Journal 21, 4915–4926. Misra, S., Beach, B.M., Hurley, J.H., 2000. Structure of the VHS domain of human Tom1 (target of myb 1): Insights into interactions with proteins and membranes. Biochemistry 39, 11282–11290. Mitsunari, T., Nakatsu, F., Shioda, N., et al., 2005. Clathrin adaptor AP-2 is essential for early embryonal development. Molecular and Cellular Biology 25, 9318–9323. Montpetit, A., Cote, S., Brustein, E., et al., 2008. Disruption of AP1S1, causing a novel neurocutaneous syndrome, perturbs development of the skin and spinal cord. PLOS Genetics 4, e1000296. Moshkanbaryans, L., Chan, L.S., Graham, M.E., 2014. The biochemical properties and functions of CALM and AP180 in clathrin mediated endocytosis. Membranes (Basel) 4, 388–413. Motley, A., Bright, N.A., Seaman, M.N., Robinson, M.S., 2003. Clathrinmediated endocytosis in AP-2-depleted cells. Journal of Cell Biology 162, 909–918. Nabhan, J.F., Pan, H., Lu, Q., 2010. Arrestin domain-containing protein 3 recruits the NEDD4 E3 ligase to mediate ubiquitination of the beta2-adrenergic receptor. EMBO Reports 11, 605–611. Nakatsu, F., Ohno, H., 2003. Adaptor protein complexes as the key regulators of protein sorting in the post-Golgi network. Cell Structure and Function 28, 419–429. Naslavsky, N., Rahajeng, J., Chenavas, S., Sorgen, P.L., Caplan, S., 2007. EHD1 and Eps15 interact with phosphatidylinositols via their Eps15 homology domains. Journal of Biological Chemistry 282, 16612–16622. Owen, D.J., Collins, B.M., Evans, P.R., 2004. Adaptors for clathrin coats: Structure and function. Annual Review of Cell and Developmental Biology 20, 153–191. Parachoniak, C.A., Park, M., 2009. Distinct recruitment of Eps15 via Its coiled-coil domain is required for efficient down-regulation of the met receptor tyrosine kinase. Journal of Biological Chemistry 284, 8382–8394.
Peden, A.A., Oorschot, V., Hesser, B.A., et al., 2004. Localization of the AP-3 adaptor complex defines a novel endosomal exit site for lysosomal membrane proteins. Journal of Cell Biology 164, 1065–1076. Polo, S., Sigismund, S., Faretta, M., et al., 2002. A single motif responsible for ubiquitin recognition and monoubiquitination in endocytic proteins. Nature 416, 451–455. Prag, G., Lee, S., Mattera, R., et al., 2005. Structural mechanism for ubiquitinatedcargo recognition by the Golgi-localized, gamma-ear-containing, ADPribosylation-factor-binding proteins. Proceedings of the National Academy of Sciences of the United States of America 102, 2334–2339. Puertollano, R., Aguilar, R.C., Gorshkova, I., Crouch, R.J., Bonifacino, J.S., 2001. Sorting of mannose 6-phosphate receptors mediated by the GGAs. Science 292, 1712–1716. Ren, X., Farias, G.G., Canagarajah, B.J., Bonifacino, J.S., Hurley, J.H., 2013. Structural basis for recruitment and activation of the AP-1 clathrin adaptor complex by Arf1. Cell 152, 755–767. Rosenthal, J.A., Chen, H., Slepnev, V.I., et al., 1999. The epsins define a family of proteins that interact with components of the clathrin coat and contain a new protein module. Journal of Biological Chemistry 274, 33959–33965. Sen, A., Madhivanan, K., Mukherjee, D., Aguilar, R.C., 2012. The epsin protein family: Coordinators of endocytosis and signaling. Biomolecular Concepts 3, 117–126. Shiba, T., Kawasaki, M., Takatsu, H., et al., 2003. Molecular mechanism of membrane recruitment of GGA by ARF in lysosomal protein transport. Nature Structural & Molecular Biology 10, 386–393. Shiba, T., Takatsu, H., Nogi, T., et al., 2002. Structural basis for recognition of acidic-cluster dileucine sequence by GGA1. Nature 415, 937–941. Sigismund, S., Woelk, T., Puri, C., et al., 2005. Clathrin-independent endocytosis of ubiquitinated cargos. Proceedings of the National Academy of Sciences of the United States of America 102, 2760–2765. Simmen, T., Honing, S., Icking, A., Tikkanen, R., Hunziker, W., 2002. AP-4 binds basolateral signals and participates in basolateral sorting in epithelial MDCK cells. Nature Cell Biology 4, 154–159. Spradling, K.D., McDaniel, A.E., Lohi, J., Pilcher, B.K., 2001. Epsin 3 is a novel extracellular matrix-induced transcript specific to wounded epithelia. Journal of Biological Chemistry 276, 29257–29267. Stahelin, R.V., Long, F., Peter, B.J., et al., 2003. Contrasting membrane interaction mechanisms of AP180 N-terminal homology (ANTH) and Epsin N-terminal homology (ENTH) domains. Journal of Biological Chemistry 278, 28993–28999. Stolt, P.C., Jeon, H., Song, H.K., et al., 2003. Origins of peptide selectivity and phosphoinositide binding revealed by structures of disabled-1 PTB domain complexes. Structure 11, 569–579. Tan, J., Evin, G., 2012. Beta-site APP-cleaving enzyme 1 trafficking and Alzheimer's disease pathogenesis. Journal of Neurochemistry 120, 869–880. Tarpey, P.S., Stevens, C., Teague, J., et al., 2006. Mutations in the gene encoding the Sigma 2 subunit of the adaptor protein 1 complex, AP1S2, cause X-linked mental retardation. American Journal of Human Genetics 79, 1119–1124. Tessneer, K.L., Cai, X., Pasula, S., et al., 2013. Epsin family of endocytic adaptor proteins as oncogenic regulators of cancer progression. Journal of Cancer Research Updates 2, 144–150. Tian, X., Hansen, D., Schedl, T., Skeath, J.B., 2004. Epsin potentiates Notch pathway activity in Drosophila and C. elegans. Development 131, 5807–5815. Traub, L.M., 2009. Tickets to ride: Selecting cargo for clathrin-regulated internalization. Nature Reviews. Molecular Cell Biology 10, 583–596. Wang, H., Traub, L.M., Weixel, K.M., et al., 2006. Clathrin-mediated endocytosis of the epithelial sodium channel. Role of Epsin. Journal of Biological Chemistry 281, 14129–14135. Wang, W., Struhl, G., 2004. Drosophila Epsin mediates a select endocytic pathway that DSL ligands must enter to activate Notch. Development 131, 5367–5380. Wang, Y.J., Wang, J., Sun, H.Q., et al., 2003. Phosphatidylinositol 4 phosphate regulates targeting of clathrin adaptor AP-1 complexes to the Golgi. Cell 114, 299–310. Xie, Z., Dong, Y., Maeda, U., Xia, W., Tanzi, R.E., 2012. RNAi-mediated knock-down of Dab and Numb attenuate Abeta levels via gamma-secretase mediated APP processing. Translational Neurodegeneration 1, 8. Yang, W., Li, C., Ward, D.M., Kaplan, J., Mansour, S.L., 2000. Defective organellar membrane protein trafficking in Ap3b1-deficient cells. Journal of Cell Science 113 (Pt 22), 4077–4086.