Sorting nexin 16 regulates EGF receptor trafficking by phosphatidylinositol-3-phosphate interaction with the Phox domain

Jang Hyun Choi1, Won-Pyo Hong1, Myong Jong Kim1, Jae Ho Kim2, Sung Ho Ryu1 and Pann-Ghill Suh1,*

1 Division of Molecular and Life Science, Pohang University of Science and Technology, San 31, Hojadong, Pohang, Kyungbuk 790-784, Republic of Korea
2 Department of Physiology, College of Medicine, Pusan National University, Pusan 602-739, Republic of Korea

* Author for correspondence (e-mail: pgs{at}postech.ac.kr)

Accepted 23 March 2004


    Summary
 Top
 Summary
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Sorting nexins (SNXs) containing the Phox (PX) domain are implicated in the regulation of membrane trafficking and sorting processes of epithelial growth factor receptor (EGFR). In this study, we investigated whether SNX16 regulates EGF-induced cell signaling by regulating EGFR trafficking. SNX16 is localized in early and recycling endosomes via its PX domain. Mutation of the PX domain disrupted the association between SNX16 and phosphatidylinositol 3-phosphate [PtdIns(3)P]. Treatment with wortmannin, a PtdIns 3-kinase inhibitor, abolished the endosomal localization of SNX16, suggesting that the intracellular localization of SNX16 is regulated by PtdIns 3-kinase activity. SNX16 was found to associate with EGFR after stimulation with EGF in COS-7 cells. Moreover, overexpression of SNX16 increased the rate of EGF-induced EGFR degradation and inhibited the EGF-induced up-regulation of ERK and serum response element (SRE). In addition, mutation in the PX domain significantly blocked the inhibitory effect of SNX16 on EGF-induced activation of ERK and SRE. From these results, we suggest that SNX16 directs the sorting of EGFR to the endosomal compartment and thus regulates EGF-induced cell signaling.

Key words: Sorting nexin (SNX), Epidermal growth factor (EGF), EGF receptor (EGFR), Trafficking, Phosphatidylinositol 3-phosphate (PtdIns(3)P)


    Introduction
 Top
 Summary
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Occupation of the receptors of hormones, growth factors and other cellular modulators triggers a complex array of signal transduction events (Clague et al., 2001). It was discovered that the majority of receptor tyrosine kinases (RTKs) and G-protein-coupled receptors (GPCRs) are rapidly endocytosed after ligand-induced activation and that they move to a series of endosomal compartments (Wiley and Burke, 2001Go; Miller and Lefkowitz, 2001Go; Tsao et al., 2001Go). After being internalized by clathrin-coated vesicles, cargo proteins such as Rab5 and Hrs are delivered to peripheral early endosomes by incoming vesicles from the cell surface that fuse with the endosomes (Clague et al., 2001). Proteins in the early endosome are sorted to one of three endosomal compartments: the surface for quick recycling; lysosomes via the late endosomes, or to perinuclear recycling endosomes (Sorkin and Von Zastrow, 2002Go; Pillay et al., 2002Go; Worby and Dixon, 2002Go).

Activated epidermal growth factor (EGF) receptors are rapidly internalized by coated pits, sorted to early endosomes and ultimately degraded in lysosomes by a process known as receptor down-regulation (Sorkin, 2001Go; Stoscheck and Carpenter, 1984Go; Wiley et al., 1991Go). This down-regulation is important for terminating signal transduction cascades that lead to cellular growth and proliferation (Wells et al., 1990Go). Although internalized receptors are ultimately degraded, the rate of receptor degradation is much slower than the rate of internalization (Wiley et al., 1985Go). Accumulated evidence suggests that internalized EGF-EGF receptor (EGFR) complexes may possess the ability to generate cell signaling from endosomes (Wang et al., 2002aGo; Burke et al., 2001Go). Various signaling molecules that regulate Ras activity, including Grb2, SHC and mSOS are co-internalized with EGFR into endosomes and remain associated with the receptor (Rizzo et al., 2000Go; Di Guglielmo et al., 1994Go; Sorkin et al., 2000Go), suggesting that internalized EGFR may play a role in endosomal signaling. Moreover, it has been reported that the inhibition of EGFR endocytosis in the presence of dominant-negative mutant dynamin decreases ERK activation (Kranenburg et al., 1999Go; McPherson et al., 2001Go). Together, these data suggest that the endosomal trafficking of activated EGF receptor is essential for its downstream signaling. Although the major events in the endocytosis of EGFR are fairly well understood, the molecular mechanisms underlying endosomal trafficking of EGFR remain poorly characterized.

The sorting nexins (SNXs) are a family of cytoplasmic and membrane-associated proteins that are believed to function in the intracellular trafficking of plasma membrane receptors such as EGFR. Recent studies have revealed that SNXs exert their function through Phox (PX) domain-mediated interaction with phosphatidylinositol (PtdIns) (Cozier et al., 2002Go; Xu et al.,2001Go). Moreover, it has been reported that SNX1 may play a critical role in the down-regulation of EGFR by trafficking from early endosomes to late endosomes/lysosomes (Kurten et al., 1996Go; Haft et al., 1998Go; Zhong et al., 2002Go). Consistent with these results, the PX domain of SNX1 is conserved in several yeast proteins, which participate in membrane trafficking such as Mvp1, Vps5/Grd2 and Grd19. Mvp1 is involved in sorting proteins in the late Golgi for delivery to the vacuole and Vps5/Grd2 and Grd19 are involved in the retrieval of proteins from the late endosome for return to the Golgi apparatus (Ekena and Stevens, 1995Go; Horazdovsky et al., 1997Go; Northwehr and Hindes, 1997Go; Voos and Stevens, 1998Go). Given this function of the yeast SNX1 homolog, SNX1 was proposed to target EGFR for lysosomal degradation through the endocytic pathway (Haft et al., 1998Go).

In this study, we demonstrate some features of the cellular localization and functions of SNX16. First, SNX16 was localized in early endosomes and specifically interacted with phosphatidylinositol 3-phosphate (PtdIns(3)P). Second, SNX16 was associated with EGFR in an EGF-dependent manner. Third, SNX16 accelerated EGF-induced EGFR down-regulation. These data indicate that SNX16 might regulate EGFR trafficking and be a novel regulator of the EGFR-mediated signaling pathway.


    Materials and Methods
 Top
 Summary
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Materials
EGF and Dulbecco's modified Eagle's medium (DMEM) were purchased from Invitrogen (Carlsbad, CA) and horse and bovine calf serums were from HyClone (Logan, UT). Anti-FLAG antibody was obtained from Sigma. Anti-phosphotyrosine antibody, Anti-EGFR antibody and peroxidase-conjugated goat anti-mouse IgG and anti-rabbit IgG were purchased from Upstate Biotechnology (Lake Placid, NY). The enhanced chemiluminescence kit (ECL) and glutathione Sepharose 4B were purchased from Amersham Pharmacia International (Buckinghamshire, UK).

Cell culture
COS-7 cells were maintained in DMEM supplemented with 10% heat-inactivated bovine calf serum in a humid atmosphere of 5% CO2:95% O2 at 37°C. Cells were seeded on 6- or 10-cm tissue culture dishes, which were pre-coated with poly-L-lysine (10 µg/ml) and grown for 1-2 days until they reached 50-80% confluence. Cells were then placed in serum-free DMEM for 18 hours before being treated with EGF (10 ng/ml).

east two-hybrid analysis
A bait plasmid was constructed using the PCR fragment encoding the C-terminal region of EGFR (a.a. 712-1210) cloned into pGBT9 vector (Clontech, CA) downstream of the GAL4 binding domain (referred to as pGBT9/C-EGFR). After transformation of yeast host strain HF7c cells with the pGBT9/C-EGFR plasmid, the yeast cells were sequentially transformed with pACT vector (Clontech, CA), which contained rat brain-derived cDNAs downstream of the GAL4 transcriptional activation domain. Selection was performed on Trp-, Leu-, and His-deleted plates. After cotransfecting the yeast cells, a filter color assay was used to assess the transcriptional activity of lacZ-galactosidase), as an indicator of the physical interaction between EGFR and proteins derived from the rat brain cDNA library. The intensity of the color reaction was scored in a semi-quantitative manner by visual inspection.

Plasmid constructs and mutagenesis
The mammalian expression vector for FLAG-epitope tagged wild type rat SNX16 (GenBank accession no. AF305780) was made by PCR. Amplified products were inserted in-frame with the FLAG-epitope tag of pFLAG-CMV-2 (Sigma, SL). The mutation of Tyr145 to Ala, designated SNX16Y145A and the deletion mutant of PX domain (a.a. 105-218), designated SNX16{Delta}PX were introduced into the FLAG-epitope tagged SNX16 cDNA by PCR-directed mutagenesis.

Cell transfection, immunoprecipitation and immunoblot assay
COS-7 cells were transfected using the liposome-mediated LipofectAMINE transfection (Invitrogen, CA) at a ratio of 1 µg of total plasmid DNA to 6 µl LipofectAMINE in 100 µl of serum free medium. After 4 hours, the medium was changed to DMEM supplemented with 10% heat-inactivated bovine calf serum, and after 36 hours the transfected cells were washed with PBS and treated with lysis buffer A (20 mM Tris-HCl, pH 7.4, 150 mM NaCl, 20 mM NaF, 1% Triton X-100, 1 mM sodium orthovanadate, 1 mM PMSF, 1 µg/ml leupeptin, 5 µg/ml aprotinin). Immunoprecipitation and immunoblotting were performed on lysates scraped from plates with a rubber pipette bulb and the supernatants were obtained after centrifugation at 12,000 g for 15 minutes at 4°C. Cell lysates were then mixed with 5 µg pre-coupled anti-FLAG antibody for 3 hours at 4°C. Immunocomplexes were collected by centrifugation, washed four times with cold lysis buffer A, and solubilized by boiling in 1x Laemmli sample buffer. Proteins were separated in 10% SDS-PAGE and transferred to nitrocellulose membranes. Blocking was performed with TTBS buffer [10 mM Tris-HCl (pH 7.6), 150 mM NaCl, 0.1% Tween 20] containing 5% skimmed milk powder. Membranes were then incubated with anti-FLAG and anti-EGFR antibodies for 4 hours at room temperature. Immunoblots were subsequently washed, incubated with horseradish peroxidase-linked secondary antibody for 1 hour at room temperature, washed four times in TTBS buffer and developed by horseradish peroxidase-dependent chemiluminescence (ECL) (Amersham International, UK).

Cellular fractionations
COS-7 cells were grown in DMEM supplemented with 10% BCS at 37°C and 5% CO2. The medium was removed and the dishes washed with ice cold PBS. The cells were scraped in 0.5 ml of homogenization buffer (100 mM MES, pH 6.5, 0.25% sucrose, 1 mM EGTA, 10 mM okadaic acid and protease inhibitor cocktail). The cell suspension was sonicated three times for 10 seconds each, followed by centrifugation at 650 g for 15 minutes to remove intact cells and nuclei. The post-nuclear supernatant (total fraction) was collected and subjected to centrifugation at 100,000 g in a Beckman TLA 100.3 rotor for 40 minutes. The supernatant was collected (cytosol fraction) and the pellet (membrane fraction) was resuspended in lysis buffer A (20 mM Tris-HCl, pH 7.4, 150 mM NaCl, 20 mM NaF, 1% Triton X-100, 1 mM sodium orthovanadate, 1 mM PMSF, 1 µg/ml leupeptin, 5 µg/ml aprotinin, 10 mM okadaic acid), sonicated and centrifuged at 14,000 g for an additional 15 minutes to remove insoluble membrane proteins. The two supernatant fractions were solubilized by boiling in 1x Laemmli sample buffer.

Protein-lipid overlay assay
The protein-lipid overlay assays were based on the procedure previously described (Dowler et al., 2002Go). Briefly, membrane arrays were generated by spotting the required mass of the indicated phosphoinositides (Cell Signals, Inc) onto a Hybond-C extra membrane. After air drying and blocking with 3% (w/v) fatty acid-free BSA in 10 mM Tris-HCl, pH 8.0, 150 mM NaCl and 0.1% (v/v) Tween-20 (TBS), the membrane was incubated overnight at 4°C in the same solution containing 1 µg/ml of the relevant recombinant protein. The membrane was then extensively washed in TBS and incubated with anti-GST antibody. The membrane was washed as before, prior to being incubated with anti-mouse horseradish peroxidase conjugate. Finally, the membrane was thoroughly washed and the proteins associated with the various phosphoinositides were visualized by enhanced chemiluminescence.

Receptor internalization assay
COS-7 cells were cultured to 60-70% confluence prior to being labeled with 1.5 mg/ml N-hydroxy-succinimide-biotin (Pierce, IL). After labeling with NHS-SS-biotin, the cells were incubated at 37°C for the indicated time in the presence or absence of 10 ng/ml EGF. Endocytosis was then stopped by transferring cells back to 4°C. After treating with reducing solution (15.5 mg/ml glutathione, 75 mM NaCl, 75 mM NaOH, and 10% fetal bovine serum) and 5 mg/ml iodoacetamide (Sigma, SL) in phosphate-buffered saline containing 0.8 mM MgCl2, 1.0 mM CaCl2 plus 1% bovine serum albumin, the cells were lysed in TNE (10 mM Tris, pH 7.5, 150 mM NaCl, 0.5% Nonidet P-40, and 1 mM EDTA). Equal quantities of the cell lysate were used to precipitate the biotinylated proteins with streptavidin beads (Pierce, IL). Biotinylated proteins were eluted from the beads by incubation at 95°C for 5 minutes in Laemmli sample buffer. The amount of receptor bound to the beads was detected by immunoblotting.

Uptake of Rhodamine-conjugated EGF and LysoTracker Red
COS-7 cells expressing GFP-tagged SNX16 were cultured on poly-L-lysine slides. Slides were transferred onto ice, washed once with PBS and incubated at 4°C for 2 hours in a medium containing 10 ng/ml Rhodamine-conjugated EGF or 10 µg/ml LysoTracker Red (Molecular Probes, OR). The cells were then washed with PBS and either processed immediately for fixation or culture medium was added. The slides were incubated for the indicated times at 37°C and then fixed. The cells were then rewashed and blocked for 1 hour with 3% fat-free BSA in PBS. Cells were viewed with a Zeiss LSM 510 confocal microscope equipped with LSM 510 version 2.02 software, an Ar (458 and 488 nm) and a He/Ne (543 and 633 nm) laser.

Reporter gene assay
PC12 cells were grown in poly-L-lysine-coated 24-well plates and transfected with serum responsive element (SRE)-luciferase plasmid using LipofectAMINE reagent (Invitrogen). After 36 hours, the cells were stimulated with EGF (10 ng/ml) for the indicated times. After washing with PBS and lysis, the luciferase activity in 1 µg of lysate was assayed using a luciferase assay kit (Promega, WI) with a luminometer (Labsystems, UK).


    Results
 Top
 Summary
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Identification and the cellular distribution of SNX16
In order to identify proteins interacting with the cytosolic domain of epidermal growth factor receptor (EGFR) (a.a. 712-1210), we used the yeast two-hybrid method. From yeast two-hybrid screening experiments, we found that two overlapping clones had significant homology to SNX16 which was deposited at GenBank (accession no. AF305780). SNX16 contains a central PX domain followed by a coiled-coil region (Fig. 1A). The presence of conserved regions within the SNXs was assessed by multiple sequence alignment using the ClustalW program. The amino acid sequences of the PX domain of SNX16 and of the other SNXs (SNX1, 2, 3 and 4) are presented in Fig. 1B. A comparison of the PX domain sequence of SNX16 with those of the other SNXs showed that all but one of the residues required for PtdIns(3)P-binding to SNX3 were conserved in the PX domain of SNX16 (Fig. 1B). The major residues in the binding site of the PX domain of SNX3 with PtdIns(3)P are one tyrosine and two arginines located at amino acids 69-71 (RRY motif) (Xu et al., 2001Go). It has been reported that the tyrosine residue in SNX3 forms a stacking interaction with the PtdIns(3)P ring, whereas the basic residues interact with the phosphates (Xu et al., 2001Go). As shown in Fig. 1A SNX16 has a conserved `RRY motif' located at 143-145. These results suggest that the PX domain of SNX16 may interact with PtdIns(3)P.



View larger version (42K):
[in this window]
[in a new window]
 
Fig. 1. Identification and cellular distribution of SNX16. (A) Domain structure of SNX16. (B) Multiple sequence alignment of sorting nexins (SNXs) 1-4 (GenBank accession nos. U53225, NP0030901, NP003786 and NP003785, respectively) and SNX16 (GenBank accession no. AF305780) was performed using the ClustalW program, and the aligned sequences are shown. (C) SNX16 and its mutant forms are depicted schematically with black boxes representing the PX domain. Amino acid positions within each construct are indicated by the numbers. (D) COS-7 cells were transiently transfected with FLAG-tagged SNX16 and its mutants followed 24-30 hours later by preparation of total cell lysates (Total), cytosolic fractions (cytosol), and membrane particulates (membrane), as described in the Materials and Methods. The distribution of FLAG-SNX16 and its mutants were determined in each fraction by immunoblotting with anti-FLAG antibody. (E) GFP-tagged SNX16 and its point mutant form in PX domain (green) were expressed in COS-7 cells. Cells were stained for endogenous EEA1 and transferrin receptor (TfR) with anti-EEA1 and anti-TfR primary antibody and visualized with Rhodamine Red-conjugated anti-rabbit IgG secondary antibody (red).

 

To determine whether the PX domain is necessary for the intracellular localization of SNX16, we prepared FLAG-tagged wild type SNX16 (SNX16WT), a PX domain-deleted mutant of SNX16 (SNX16{Delta}PX) and a point mutant of SNX16 in the PX domain by replacing tyrosine-145 with alanine (SNX16Y145A) (Fig. 1C). Post-nuclear supernatants were divided into cytosol and a membrane particulate fraction from SNX16-transfected COS-7 cells. As assessed by the western blotting of the cell fractions, SNX16WT partitioned into both the membrane particulate and cytosolic fraction, but SNX16{Delta}PX and SNX16Y145A were present only in the cytosol fraction (Fig. 1D). To investigate the cellular distribution of SNX16 further, GFP-tagged SNX16WT and SNX16Y145A were expressed in COS-7 cells and their intracellular localizations were analyzed by confocal microscopy. SNX16WT was localized in intracellular vesicles and overlapped with EEA1-positive early endosomes and transferrin receptor (TfR)-positive recycling endosomes. However, SNX16Y145A was diffusely expressed in the cytoplasm, indicating that an intact PX domain structure is essential for the proper targeting of SNX16 to cellular vesicles (Fig. 1E). These results indicate that SNX16 is enriched in early and recycling endosomes and that the PX domain is essential for the cellular localization of SNX16 in endosomal compartments.

PX domain-mediated direct interaction of SNX16 with PtdIns(3)P
To examine whether SNX16 is capable of interacting with phosphoinositides, we conducted a protein-lipid overlay analysis with recombinant SNX16WT and SNX16Y145A proteins. As shown in Fig. 2A, SNX16WT specifically associated with PtdIns(3)P. No significant binding was observed with any other phosphoinositides. However, SNX16Y145A did not associate with PtdIns(3)P, suggesting that the conserved residue in the PX domain of SNX16 is essential for this interaction. Owing to the observed specific binding of the PX domains to PtdIns(3)P, we speculate that PtdIns 3-kinase activity may be involved in the subcellular localization of SNX16. To assess this hypothesis, we examined the effect of wortmannin, a specific inhibitor of PtdIns 3-kinase, on the subcellular localization of SNX16. Treatment with wortmannin abolished the endosomal localization of SNX16, suggesting that the intracellular localization of SNX16 is regulated by PtdIns 3-kinase activity (Fig. 2B). In addition, the association between SNX16 and TfR markedly diminished after wortmannin treatment (Fig. 2C). These results suggest that PtdIns 3-kinase may participate in the association between SNX16 and endosomes.



View larger version (46K):
[in this window]
[in a new window]
 
Fig. 2. PX domain-dependent direct interaction between SNX16 and PtdIns(3)P. (A) The ability of GST-fused SNX16WT and SNX16Y145A to bind a variety of phosphoinositides (PI) was analyzed by protein-lipid overlay assay. 100 pmols of the relevant PI was spotted on to a nitrocellulose membrane, which was then incubated with the purified proteins. The membranes were washed and proteins bound to the membrane by lipid interaction were detected with anti-GST antibody. Results representative of at least three separate experiments are shown. (B) COS-7 cells were transfected with GFP-tagged SNX16 and fixed. Cells were processed to detect SNX16 (green) and TfR (red) simultaneously by confocal microscopy. Like TfR, SNX16 was redistributed from endosomal membranes to the cytosol after treatment with wortmannin. Bars, 10 µm. (C) COS-7 cells were transfected with FLAG-tagged SNX16WT and SNX16Y145A, cultured in the absence of serum for 3 hours and then treated with or without wortmannin (100 nM) for 15 min. Cells were lysed and lysates were immunoreacted with anti-TfR antibody. Complexes associated with TfR were analyzed with anti-FLAG and anti-TfR antibodies. Total cell lysates (40 µg) were immunoblotted with anti-FLAG antibody.

 

Association of SNX16 with epidermal growth factor (EGF) receptor
Previous reports have suggested that other sorting nexins interact with RTKs and play a key role in the RTK-mediated signaling pathway (Haft et al., 1998Go; Zhong et al., 2002Go). In this study, our yeast two-hybrid data suggested that SNX16 interacts with EGFR. To confirm the ability of SNX16 to associate with EGFR, COS-7 cells were transiently transfected with SNX16WT. SNX16 was found to associate with EGFR in a time-dependent manner and this reached a maximum 15 minutes after EGF treatment (Fig. 3A). Next, we examined the colocalization of SNX16 with rhodamine-labeled EGF by confocal microscopy. COS-7 cells expressing GFP-SNXWT were incubated with Rhodamine-labeled EGF at 4°C to allow binding between SNX and EGFR. Cells were then incubated at 37°C and the uptake of bound EGF was examined. Receptor-bound EGF did not enter the cells at 4°C (Fig. 3B), however, incubation of the cells at 37°C allowed endocytosis. Moreover, rhodamine-labeled EGFEGFR complexes were colocalized with SNX16 (Fig. 3B). These results suggest that EGFR travels to endosomal compartments containing SNX16 during endocytosis.



View larger version (55K):
[in this window]
[in a new window]
 
Fig. 3. SNX16 associates with EGFR. (A) COS-7 cells were transfected with FLAG-tagged SNX16 and treated with EGF for the indicated times. Cell lysates were immunoprecipitated with anti-FLAG antibody and then immunoblotted with anti-EGFR antibody. Total cell lysates (40 µg) were then immunoblotted with anti-EGFR and anti-FLAG antibody. (B) COS-7 cells were transfected with GFP-tagged SNX16 and treated with EGF for the indicated times. Fixed cells were processed to simultaneously detect SNX16 (green) and EGFR (red) by confocal microscopy. Bars, 10 µm.

 

Effect of SNX16 on the down-regulation of EGFR
Given the association between SNX16 and EGFR, we examined the effect of SNX16 on the trafficking of EGFR in the endosomal compartment. SNX16 potentiated EGF-induced EGFR degradation (Fig. 4A), suggesting that SNX16 may participate in the lysosomal targeting and/or degradation of EGFR. Because EGFR trafficking to endosomes is important in the regulation of receptor-mediated signaling, we examined whether the overexpression of SNX16WT influences EGF-dependent mitogenic-activated protein kinase (MAPK) activation. COS-7 cells transfected with SNX16WT showed a progressive decrease in ERK1/2 phosphorylation after EGF treatment. However, the PX domain-defective mutant showed no difference compared to empty vector-transfected cells.



View larger version (59K):
[in this window]
[in a new window]
 
Fig. 4. Effect of SNX16 on the EGF-induced degradation of EGFR. (A) COS-7 cells expressing FLAG-tagged SNX16WT and SNX16Y145A were treated with EGF for the indicated times and then immunoblotted with antibodies against EGFR, FLAG or actin. The activation of MAPK (phospho-ERK1/2) was also assessed by immunoblotting with anti-phospho ERK1/2 antibody. Anti-ERK1/2 antibody was used to detect levels of ERK1/2 as a loading control. (B) COS-7 cells were transfected with GFP-tagged SNX16 and incubated with Rhodamine-conjugated LysoTracker. Cells were treated with EGF for the indicated times. Fixed cells were processed to detect SNX16 (green) and LysoTracker (red) simultaneously by confocal microscopy. Bars, 10 µm. (C) COS-7 cells were treated with EGF at the indicated times. Cell surface proteins were biotinylated as described in Materials and Methods. Biotinylated proteins were recovered using Streptavidin and the amount of EGFR recovered was assessed by western blotting using anti-EGFR antibody.

 

To determine whether SNX16 is sequestered in lysosomal compartments of COS-7 cells, we examined the colocalization of SNX16 using Lysotracker Red, a lysosome-directed marker (Fig. 4B). In the absence of EGF, colocalized SNX16-LysoTracker Red signals were weak. However, colocalized signals were much stronger after EGF treatment. These results suggest that SNX16 may be involved in lysosomal targeting. In addition, a comparison of the rates of internalization of EGFR with and without SNX16 overexpression revealed no significant differences (Fig. 4C). Taken together, these results suggest that SNX16 may regulate EGFR targeting to lysosomes, but not ligand-induced receptor internalization.

Effect of SNX16 on SRE-dependent transcription activity
As ERK activation stimulates serum response element (SRE)-dependent transcription (Whitmarsh et al., 1995Go), we examined the influence of SNX16 on SRE-dependent transcription using an SRE-luciferase reporter gene assay. In vector-transfected cells, EGF-induced SRE activation elicited a threefold increase in luciferase activity and the overexpression of SNX16Y145A also caused a threefold increased response (Fig. 5). However, the overexpression SNX16WT failed to increase SRE-dependent transcriptional activity. These results suggest that SNX16 may negatively regulate EGFR-mediated signaling by potentiating the degradation of EGFR.



View larger version (19K):
[in this window]
[in a new window]
 
Fig. 5. Role of SNX16 in the regulation of EGF-induced gene transcription. COS-7 cells were transfected with the reporter genes SRE-Luc (10 ng) and lacZ (20 ng) and with FLAG-tagged SNX16WT and SNX16Y145A (10 ng). After EGF treatment for the indicated times, EGF-induced increase in luciferase activity was quantified and expressed as mean±s.d.

 

The PX domain is required for the homo-oligomerization of SNX16
The formation of homo- and hetero-oligomers is a common feature of various sorting nexins. Furthermore, the oligomerization of SNXs is critical for their cellular functions (Haft et al., 1998Go; Kurten et al., 2001Go). To determine whether SNX16 can associate with itself, we transfected HA-tagged or Flag-tagged SNX16 in COS-7 cells (Fig. 6). SNX16WT formed homo-oligomers, but the deletion of the PX domain disrupted the oligomerization of SNX16. Furthermore, SNX16Y145A that abrogates vesicular localization (Fig. 1E) and PtdIns(3)P binding (Fig. 2A) did not associate with itself (Fig. 6). These results suggest that the PX domain is a critical determinant of the self-association of SNX16.



View larger version (41K):
[in this window]
[in a new window]
 
Fig. 6. The PX domain is required for the homo-oligomerization of SNX16. Various HA or FLAG-tagged SNX mutants were coexpressed in COS-7 cells. FLAG-tagged proteins were immunoprecipitated and analyzed by immunoblotting using anti-HA antibody. Total cell lysates (40 µg) were then immunoblotted with anti-HA and anti-FLAG antibody.

 


    Discussion
 Top
 Summary
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Sorting nexins (SNXs) were identified recently as important components of the molecular machinery responsible for sorting membrane proteins in the trans-Golgi network and in endocytic systems (Worby and Dixon, 2002Go). SNX1, the sorting nexin first identified, was found to associate with EGFR and function in the lysosomal trafficking of EGFR (Kurten et al., 1996Go). In addition, the microinjection of antibodies into the Grd19p homologue of SNX3 slowed the transport of transferrin receptor from early to recycling endosomes (Xu et al., 2001Go). These results suggest that SNXs are involved in various aspects of endocytosis and in the trafficking of receptor tyrosine kinases. In this study, we demonstrate that SNX16 is preferentially associated with intermediate endosomes involved in the transport of internalized EGFR from early endosomes to lysosomes, which suggests that SNX16 can regulate EGFR trafficking and that it is a novel regulator of the EGFR-mediated signaling pathway.

Knowledge of the intracellular location of SNXs is critical for defining their function. Various PX domain-containing proteins have been localized in early endosomes. Furthermore, previous studies have demonstrated that SNX1 and SNX3 are partly localized in perinuclear vesicles or early endosome, suggesting that SNX1 or SNX3 may be important for vesicular trafficking (Xu et al., 2001Go; Zhong et al., 2002Go). In the present study, we found that SNX16 preferentially localized into early and recycling endosomes (Fig. 1E). Moreover, mutation of the PX domain disrupted the vesicular localization of SNX16, suggesting that the PX domain of SNX16 is essentially required for this process.

Various phosphoinositides are known to regulate diverse cellular signal transduction pathways and/or membrane transport events (Toker, 2002Go). The cellular level of PtdIns(3)P is regulated by the action of PtdIns 3-kinase, or by the actions of the corresponding lipid phosphatases or lipases (Cantley, 2002Go). It has been reported that Vps34p, a Saccharamyces cerevisiae homologue of PtdIns 3-kinase, plays an essential role in membrane transport of PtdIns(3)P from the Golgi apparatus to vacuoles via endosomes (Burda et al., 2002Go). In the present study, several lines of evidence suggest that the endosomal localization of SNX16 is dependent on the ability of its PX domain to bind PtdIns(3)P. First, point mutated SNX16 (SNX16Y145A) was incapable of binding to PtdIns(3)P, and did not localize to the endosomal compartments (Fig. 1E and Fig. 2A). Second, treatment with wortmannin, a specific inhibitor of PtdIns 3-kinase, abolished the endosomal localization of SNX16, suggesting that the intracellular localization of SNX16 is regulated by PtdIns 3-kinase activity (Fig. 2B,C). Taken together, these results strongly suggest that the PX domain of SNX16 participates in the association with PtdIns(3)P required for targeting to the endosomal compartments.

SNX1 was cloned by using a yeast two-hybrid screen with a portion of cytoplasmic domain of EGFR as bait (Kurten et al., 1996Go). In addition, other SNXs such as SNX2 and SNX4 have also been associated with EGFR and PDGFR (Haft et al., 1998Go). However, these reports showed that the SNX-receptor interactions were not affected by the activation of the receptors by ligand. Interestingly, we observed that SNX16 specifically associates with EGFR in an EGF-dependent manner (Fig. 3). Furthermore, SNX16 colocalized with the EGF-EGFR complexes in intracellular vesicles. This may be due to EGF-induced redistribution of EGFR to intracellular compartments containing SNX16. Our findings strongly suggest that SNX16 participates in the endosomal trafficking of EGFR by the specific interaction.

SNXs have been shown to regulate the endocytosis of a variety of receptors such as EGFR, protease-activated receptor-1 and low-density lipoprotein receptors (Kurten et al., 1996Go; Wang et al., 2002bGo; Stockinger et al., 2002Go). Furthermore, many homologues of SNXs found in yeast have been found to play an essential role in receptor recycling between early endosomes and late endosomes/lysosomes (Horazdovsky et al., 1997Go). In mammalian cells, the overexpression of SNX1 was noted to accelerate EGFR degradation, implying a role for this protein in endosomal transport (Kurten et al., 1996Go). Consistent with this report, we demonstrate the SNX16 has a function in the degradation of EGFR. Three pieces of evidence support the physiological significance of the function of SNX16 on the regulation of the endosome-to-lysosome trafficking of EGFR. First, SNX16 associates with EGFR in an EGF-induced manner (Fig. 3), and colocalizes with lysosomes after EGF treatment (Fig. 4B). These findings suggest that SNX16 participates in the endosomal trafficking of EGFR. Second, the overexpression of SNX16 accelerates the degradation of EGFR, but point mutation of SNX16 (SNX16Y145A) inhibits EGFR degradation (Fig. 4A), indicating that the PX domain of SNX16 may be critical for the function of SNX16 on EGFR trafficking. Furthermore, the overexpression of SNX16 was found to have no effect on receptor internalization (Fig. 4C), suggesting that SNX16 may be involved in the regulation of the endosome-to-lysosome trafficking of EGFR. Third, the overexpression of SNX16 was found to inhibit serum responsive element (SRE)-dependent transcriptional activity (Fig. 5). Taken together, these results suggest that SNX16 may negatively regulate EGFR-mediated signaling by potentiating the degradation of EGFR.

In yeast, the oligomerization of SNXs is essential for organizing the sorting and recycling of functional complexes (Horazdovsky et al., 1997Go). Furthermore, in mammalian cells, it has been reported that the homo- and heterodimerization of SNXs is critical for receptor sorting and degradation (Haft et al., 2000Go; Kurten et al., 2001Go). Moreover, the PX domain of SNX1 was shown to mediate oligomerization with itself and with SNX2 (Haft et al., 1998Go). Furthermore, SNX6 and SNX15 also exhibit self-association mediated by the PX domain (Parks et al., 2001Go; Phillips et al., 2001Go). We observed that the PX domain of SNX16 is essential for its oligomerization (Fig. 6), suggesting that SNX16, which forms the sorting complexes, specifically regulates the function of EGFR and facilitates the degradation of EGFR in an EGF-dependent manner.

In conclusion, this study demonstrates for the first time that SNX16 functions as a sorting cargo in the endosome pathway to lysosomes. Moreover, it establishes a correlation between the ability of SNX16 to bind PtdIns(3)P, the association between SNX16 and the endosomal membrane compartment and its regulation of endosomal function. The endosomal association of SNX16 and the functional regulation of endosomes by SNX16 overexpression depend absolutely on its PtdIns(3)P-binding activity, thus linking the subcellular localization and biochemical properties of SNX16 to its regulation of endosomal function. These findings provide a new insight into how SNX16 might function in protein trafficking in mammalian cells. The challenge now is to elucidate the mechanism by which SNX16 regulates the sorting of EGFR from early endosomes to lysosomes, an event critical for the termination of receptor signaling.


    Acknowledgments
 
This work was supported by a grant from the 21C Frontier Functional Human Genome Project from the Ministery of Science and Technology of Korea (FG00-0304-001) and by the National Research and Development Program for Fusion Strategy of Advanced Technologies of MOST.


    References
 Top
 Summary
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 

Burda, P., Padilla, S. M., Sarkar, S. and Emr, S. D. (2002). Retromer function in endosome-to-Golgi retrograde transport is regulated by the yeast Vps34 PtdIns 3-kinase. J. Cell Sci. 115, 3889-3900.[Abstract/Free Full Text]

Burke, P., Schooler, K. and Wiley, H. S. (2001). Regulation of epidermal growth factor receptor signaling by endocytosis and intracellular trafficking. Mol. Biol. Cell 12, 1897-1910.[Abstract/Free Full Text]

Cantley, L. C. (2002). The phosphoinositide 3-kinase pathway. Science 296, 1655-1657.[Abstract/Free Full Text]

Clague, M. J. and Urbe, S. (2001). The interface of receptor trafficking and signaling. J. Cell Sci. 114, 3075-3081.[Abstract/Free Full Text]

Cozier, G. E., Carlton, J., McGregor, A. H., Gleeson, P. A., Teasdale, R. D., Mellor, H. and Cullen, P. J. (2002). The phox homology (PX) domain-dependent, 3-phosphoinositide-mediated association of sorting nexin-1 with an early sorting endosomal compartment is required for its ability to regulate epidermal growth factor receptor degradation. J. Biol. Chem. 277, 48730-48736.[Abstract/Free Full Text]

Di Guglielmo, G. M., Baass, P. C., Ou, W. J., Posner, B. I. and Bergeron, J. J. (1994). Compartmentalization of SHC, GRB2 and mSOS, and hyperphosphorylation of Raf-1 by EGF but not insulin in liver parenchyma. EMBO J. 13, 4269-4277.[Abstract]

Dowler, S., Kular, G. and Alessi, D. R. (2002). Protein lipid overlay assay. Sci. STKE 129, PL6.

Ekena, K. and Stevens, T. H. (1995). The Saccharomyces cerevisiae MVP1 gene interacts with VPS1 and is required for vacuolar protein sorting. Mol. Cell. Biol. 15, 1671-1678.[Abstract]

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.[Abstract/Free Full Text]

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.[Abstract/Free Full Text]

Horazdovsky, B. F., Davies, B. A., Seaman, M. N. N., McLaughlin, S. A., Yoon, S. H. and Emr, S. D. (1997). A sorting nexin-1 homologue, Vps5p, forms a complex with Vps17p and is required for recycling the vacuolar protein-sorting receptor. Mol. Biol. Cell 8, 1529-1541.[Abstract]

Kranenburg, O., Verlaan, I. and Moolenaar, W. H. (1999). Dynamin is required for the activation of mitogen-activated protein (MAP) kinase by MAP kinase kinase. J. Biol. Chem. 274, 35301-35304.[Abstract/Free Full Text]

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.[Abstract]

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.[Abstract/Free Full Text]

McPherson, P. S., Kay, B. K. and Hussain, N. K. (2001). Signaling on the endocytic pathway. Traffic 2, 375-384.[CrossRef][Medline]

Miller, W. E. and Lefkowitz, R. J. (2001). Expanding roles for beta-arrestins as scaffolds and adapters in GPCR signaling and trafficking. Curr. Opin. Cell. Biol. 13, 139-145.[CrossRef][Medline]

Northwehr, S. F. and Hindes, A. E. (1997). A yeast VPS5/GRD2 gene encodes a sorting nexin-1-like protein required for localizing membrane proteins to the late Golgi. J. Cell Sci. 110, 1063-1072.[Abstract/Free Full Text]

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-beta family of receptor serine-threonine kinases. J. Biol. Chem. 276, 19332-19339.[Abstract/Free Full Text]

Phillips, S. A., Barr, V. A., Haft, D. H., Taylor, S. I. and Renfrew-Haft, C. (2001). Identification and characterization of SNX15, a novel sorting nexin involved in protein trafficking. J. Biol. Chem. 276, 5074-5084.[Abstract/Free Full Text]

Pillay, C. S., Elliott, E. and Dennison, C. (2002). Endolysosomal proteolysis and its regulation. Biochem. J. 363, 417-429.[CrossRef][Medline]

Rizzo, M. A., Shome, K., Watkins, S. C. and Romero, G. (2000). The recruitment of Raf-1 to membranes is mediated by direct interaction with phosphatidic acid and is independent of association with Ras. J. Biol. Chem. 275, 23911-23918.[Abstract/Free Full Text]

Sorkin, A. (2001). Internalization of the epidermal growth factor receptor: role in signaling. Biochem. Soc. Trans. 29, 480-484.[Medline]

Sorkin, A. and von Zastrow, M. (2002). Signal transduction and endocytosis: close encounters of many kinds. Nat. Rev. Mol. Cell. Biol. 3, 600-614.[CrossRef][Medline]

Sorkin, A., McClure, M., Huang, F. and Carter, R. (2000). Interaction of EGF receptor and grb2 in living cells visualized by fluorescence resonance energy transfer (FRET) microscopy. Curr. Biol. 10, 1395-1398.[CrossRef][Medline]

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.[Abstract/Free Full Text]

Stoscheck, C. M. and Carpenter, G. (1984). Down-regulation of epidermal growth factor receptors: direct demonstration of receptor degradation in human fibroblasts. J. Cell Biol. 98, 1048-1053.[Abstract]

Toker, A. (2002). Phosphoinositides and signal transduction. Cell. Mol. Life Sci. 59, 761-779.[CrossRef][Medline]

Tsao, P., Cao, T. and von Zastrow, M. (2001). Role of endocytosis in mediating downregulation of G-protein-coupled receptors. Trends Pharmacol. Sci. 22, 91-96.[CrossRef][Medline]

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, 577-589.[Abstract/Free Full Text]

Wang, Y., Pennock, S., Chen, X. and Wang, Z. (2002a). Internalization of inactive EGF receptor into endosomes and the subsequent activation of endosome-associated EGF receptors. Sci. STKE 161, PL17.

Wang, Y., Zhou, Y., Szabo, K., Haft, C. R. and Trejo, J. (2002b). Down-regulation of protease-activated receptor-1 is regulated by sorting nexin 1. Mol. Biol. Cell 13, 1965-1976.[Abstract/Free Full Text]

Wells, A., Welsh, J. B., Lazar, C. S., Wiley, H. S., Gill, G. N. and Rosenfeld, M. G. (1990). Ligand-induced transformation by a noninternalizing epidermal growth factor receptor. Science 247, 962-964.[Medline]

Whitmarsh, A. J., Shore, P., Sharrocks, A. D. and Davis, R. J. (1995). Integration of MAP kinase signal transduction pathways at the serum response element. Science 269, 403-407.[Medline]

Wiley, H. S. and Burke, P. M. (2001). Regulation of receptor tyrosine kinase signaling by endocytic trafficking. Traffic 2, 12-18.[CrossRef][Medline]

Wiley, H. S., VanNostrand, W., McKinley, D. N. and Cunningham, D. D. (1985). Intracellular processing of epidermal growth factor and its effect on ligand-receptor interactions. J. Biol. Chem. 260, 5290-5295.[Abstract]

Wiley, H. S., Herbst, J. J., Walsh, B. J., Lauffenburger, D. A., Rosenfeld, M. G. and Gill, G. N. (1991). The role of tyrosine kinase activity in endocytosis, compartmentation, and down-regulation of the epidermal growth factor receptor. J. Biol. Chem. 266, 11083-11094.[Abstract/Free Full Text]

Worby, C. A. and Dixon, J. E. (2002). Sorting out the cellular functions of sorting nexins. Nat. Rev. Mol. Cell. Biol. 3, 919-931.[CrossRef][Medline]

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.[CrossRef][Medline]

Zhong, Q., Lazar, C. S., Tronchere, H., Sato, T., Meerloo, T., Yeo, M., Songyang, Z., Emr, S. D. and Gill, G. N. (2002). Endosomal localization and function of sorting nexin 1. Proc. Natl. Acad. Sci. USA 99, 6767-6772.[Abstract/Free Full Text]





This Article
Summary
Figures Only
Full Text (PDF)
All Versions of this Article:
jcs.01233v1
117/18/4209    most recent
Alert me when this article is cited
Alert me if a correction is posted
Services
Email this article to a friend
Similar articles in this journal
Similar articles in PubMed
Alert me to new issues of the journal
Download to citation manager
Google Scholar
Articles by Choi, J. H.
Articles by Suh, P.-G.
Articles citing this Article
PubMed
PubMed Citation
Articles by Choi, J. H.
Articles by Suh, P.-G.