热卖商品
新闻详情
CD317 Activates EGFR by Regulating Its Association with Lipid...
来自 :
发布时间:2025-01-10
CD317 Activates EGFR by Regulating Its Association with Lipid Rafts Guizhong Zhang, Xin Li, Qian Chen, Junxin Li, Qingguo Ruan, Youhai H. Chen, Xiaolu Yang and Xiaochun WanGuizhong Zhang 1Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People\'s Republic of China.Find this author on Google ScholarFind this author on PubMedSearch for this author on this siteXin Li 1Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People\'s Republic of China.Find this author on Google ScholarFind this author on PubMedSearch for this author on this siteQian Chen 1Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People\'s Republic of China.Find this author on Google ScholarFind this author on PubMedSearch for this author on this siteJunxin Li 1Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People\'s Republic of China.Find this author on Google ScholarFind this author on PubMedSearch for this author on this siteQingguo Ruan 1Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People\'s Republic of China.Find this author on Google ScholarFind this author on PubMedSearch for this author on this siteYouhai H. Chen 2Department of Pathology and Laboratory of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania.Find this author on Google ScholarFind this author on PubMedSearch for this author on this siteXiaolu Yang 3Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.Find this author on Google ScholarFind this author on PubMedSearch for this author on this siteFor correspondence: xyang@pennmedicine.upenn.eduxc.wan@siat.ac.cnXiaochun Wan 1Shenzhen Laboratory of Fully Human Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People\'s Republic of China.Find this author on Google ScholarFind this author on PubMedSearch for this author on this siteFor correspondence: xyang@pennmedicine.upenn.eduxc.wan@siat.ac.cn AbstractEGFR regulates various fundamental cellular processes, and its constitutive activation is a common driver for cancer. Anti-EGFR therapies have shown benefit in cancer patients, yet drug resistance almost inevitably develops, emphasizing the need for a better understanding of the mechanisms that govern EGFR activation. Here we report that CD317, a surface molecule with a unique topology, activated EGFR in hepatocellular carcinoma (HCC) cells by regulating its localization on the plasma membrane. CD317 was upregulated in HCC cells, promoting cell-cycle progression and enhancing tumorigenic potential in a manner dependent on EGFR. Mechanistically, CD317 associated with lipid rafts and released EGFR from these ordered membrane domains, facilitating the activation of EGFR and the initiation of downstream signaling pathways, including the Ras–Raf–MEK–ERK and JAK–STAT pathways. Moreover, in HCC mouse models and patient samples, upregulation of CD317 correlated with EGFR activation. These results reveal a previously unrecognized mode of regulation for EGFR and suggest CD317 as an alternative target for treating EGFR-driven malignancies.Significance: Activation of EGFR by CD317 in hepatocellular carcinoma cells suggests CD317 as an alternative target for treating EGFR-dependent tumors.IntroductionLiver cancer is the second leading cause of cancer-related mortality worldwide, resulting in ∼800,000 deaths annually (1). Unlike most other cancers for which the mortality has declined, death rates of liver cancer cases have been rising each year over the last 10 years in the United States and other countries (2, 3). The vast majority (∼90%) of liver cancers are hepatocellular carcinoma (HCC). HCC can be treated at early stages by liver transplantation or surgical resection; however, the overwhelming majority of patients are in advanced stages at the time of diagnosis with few treatment options. The 5-year survival remains at a dismal rate of ∼18% (4). Although the risk factors for HCC are well known—including chronic infection of hepatitis B and C viruses (HBV and HCV) and alcohol abuse, the molecular events driving the pathogenesis are poorly defined (2, 3).The EGFR (also known as ErbB1 or HER1) belongs to the EGFR/ErbB subfamily of receptor tyrosine kinases (RTK), along with ErbB2/HER2, ErbB3/HER3, and ErbB4/HER4 (5). The cognate ligands for EGFR include EGF, TGFα, and amphiregulin (AREG; ref. 6), which induce EGFR homodimerization or its hetero-dimerization, with the closely related RTKs leading to phosphorylation of EGFR at multiple tyrosine residues in its intracellular region (5). This enables the recruitment of various signaling molecules and the initiation of intracellular signaling pathways (e.g., Ras–Raf–MEK–ERK, JAK–STAT, and PI3K–AKT pathways) to modulate proliferation, survival, mobility, metabolism, differentiation, and other fundamental cellular processes (7). Dysregulation of EGFR drives the development and progression of various tumors (8). Therapies that inhibit EGFR, including monoclonal antibodies (e.g., trastuzumab) and small-molecule tyrosine kinase inhibitors (e.g., erlotinib), are among the most successful examples of targeted cancer therapies, benefiting patients with metastatic lung, colorectal, pancreatic, or head and neck cancers (9). However, resistance to these therapies almost invariably develops (9). Moreover, EGFR is overexpressed in 40% to 70% of human HCCs (10). Still, EGFR inhibitors (cetuximab, gefitinib, and erlotinib) did not show significant efficacy in unselected patients with advanced HCC in clinical trials (11, 12). Therefore, a better understanding of the regulation of EGFR signaling is critical for the therapy of cancer in general and HCC in particular.EGFR is associated with lipid rafts (13). These specialized membrane microdomains—which are enriched in cholesterol, sphingolipids, and certain proteins—are involved in intracellular signaling, trafficking, and pathogen–host interactions. In general, lipid rafts promote interactions among signaling molecules, and their disruption (e.g., by cholesterol depletion) impairs receptor activation (14). However, EGFR appears to be an exception, as it is kept in an auto-inhibitory conformation upon localization in lipid rafts (13). Release of EGFR from lipid rafts can lead to ligand-independent EGFR dimerization and auto-activation (15, 16). Given the importance of lipid rafts in EGFR activation, a salient, yet poorly understood question, is how the association of EGFR with lipid rafts is regulated.CD317 (also known as BST2, HM1.24, or tetherin) is a type II transmembrane protein with an unusual structure, comprising a short cytoplasmic N-terminal region, a transmembrane (TM) domain, a coiled-coil extracellular domain, and a C-terminal glycosylphosphatidylinositol (GPI) anchor (Supplementary Fig. S1A; refs. 17, 18). As such, CD317 is double anchored into the membrane through both the TM domain and GPI, a topology that is unique among human proteins (19, 20). CD317 is thought to be inserted both inside (via the GPI anchor) and outside (via the TM domain) lipid rafts (19, 21). Thus, it may influence the high-order structures of these lipid microdomains. CD317 plays a multifaceted role in regulating host–pathogen relationship. It is an important restriction factor for various enveloped viruses such as HIV, HBV, and HCV, preventing their release from infected cells (22). When physically sequestering retroviral particles, CD317 also potently activates NF-κB, leading to the expression of proinflammatory gene (23–26). Of note, CD317 is implicated in tumorigenesis. It is overexpressed in multiple myeloma and several other cancers (23, 27–30), and neutralizing monoclonal antibodies against CD317 showed tumor toxicity in animal models (31, 32). Nevertheless, it remains unclear whether CD317 affords a proliferative advantage to tumor cells and if so, what the underlying molecular mechanism may be.In this study, we investigate the role of CD317 in HCC. We find that CD317 is upregulated in HCC samples. CD317 strongly promotes HCC proliferation and cell-cycle progression and enhances its tumorigenicity. Interestingly, the effect of CD317 is mediated by EGFR. CD317 promotes the release of EGFR from the lipid rafts, promoting its auto-activation. These results establish a critical role for CD317 in tumorigenesis and reveal a new mode of regulation for EGFR that likely influences the pathogenesis of HCC and other tumors.Materials and MethodsReagentsDMEM medium and fetal bovine serum (FBS) were purchased from HyClone, l-glutamine from Gibco, Annexin V-FITC/PI Apoptosis Detection Kit from TransGen Biotech, cell-cycle analysis kit from KeyGEN BioTECH, methyl-β-cyclodextrin (C4555) and cholesterol (C3045) from Sigma-Aldrich, and Erlotinib-HCl (OSI-744) from Selleckchem.Cell cultureBel7402, HepG2, and Huh7 were purchased from Shanghai Cell Bank of Chinese Academy of Sciences (Shanghai, China) and were authenticated by the vendor using short tandem repeat (STR) profiling. HEK293T was purchased from ATCC and confirmed by STR (GENEWIZ). Cell lines were cultured in DMEM medium (HyClone) supplemented with 10% FBS (HyClone) and 2 mmol/L l-glutamine (Gibco), and periodically authenticated by morphologic inspection and bioMarkers detection (if applicable). All cells were used within 6 months of continuous passage and checked for the absence of Mycoplasma using detection kit (LT27-710; Lonza).Human samples and IHC stainingTwo human hepatocellular carcinoma arrays were used in this study. The first one contained 35 tumor samples and 8 normal liver tissues (Xi\'an Alena Biotech), and the second one contained 75 tumor samples (Shanghai Outdo Biotech). The other five specimens (three HCC and two normal liver tissues) were obtained from Second People\'s Hospital of Shenzhen, which was approved by the Research Committee of Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences.IHC staining was performed as previously described (33) using following antibodies: anti-CD317 (ab134061; Abcam), anti-pY845 EGFR (BS5013; Bioworld) or (GTX133600; GeneTex), and anti-PCNA (10205-2-AP; Proteintech). All slides were independently analyzed by two pathologists in a blinded manner and scored according to staining intensity (no staining = 0, weak staining = 1, moderate staining = 2, strong staining = 3) and the number of stained cells (0% = 0, 1%–25% = 1, 26%–50% = 2, 51%–75% = 3, 76%–100% = 4). Final immunoreactive scores were determined by multiplying the staining intensity by the number of stained cells, with minimum and maximum scores of 0 and 12, respectively (34). The Mann–Whitney U test was used to evaluate the statistical significance of the results.Xenograft tumor modelsMale BALB/c nude mice at 6 to 8 weeks of age were purchased from Guangdong Medical Laboratory Animal Center (Guangzhou, China) and housed in the SIAT facility under pathogen-free conditions.To investigate the effects of CD317 on established tumor growth, we performed both overexpression and knockdown experiments. For overexpression, 5 × 106 CD317-stable expression HepG2 cells or control cells in 100 μL PBS containing 50% Matrigel (BD Bioscience) were injected subcutaneously into flanks of nude mice. Tumor incidence and growth were monitored. Twenty-eight days later, tumor-bearing and control mice were sacrificed, and tumors were dissected for the measurement of tumor weights and volumes using the formula [length × (width)2]/2. For knockdown, 1.5 × 107 HepG2 cells stably expressing CD317 or control shRNA were injected. Tumor growth was monitored, and tumors were harvested at day 23. All animal experiments were approved by the Institutional Animal Care and Use Committee at SIAT.Bioinformatics analysis of CD317 expression in human HCCsCD317 protein expression in HCC tissues and normal tissues was determined from the human protein atlas (www.proteinatlas.org). HCC CD317 gene expression was determined through analysis of Mas Liver and Wurmbach Liver databases, which are available through Oncomine (www.oncomine.org).Plasmids and siRNAsCD317 (the long isoform) was transiently expressed using MigR1- or pCMV-based plasmids, or stably expressed using PLVX-based lentiviral vectors. The full-length human CD317 cDNA was generated from Jurkat cells by RT-PCR, digested with BglII and XhoI, and cloned into MigR1 or PLVX. The extracellular domain of CD317 (ECD, amino acids: 44–159; ref. 35) was generated via PCR reaction and cloned into pCMV-C-His vector. The plasmids encoding CD317 mutants in which the two N-linked glycosylation sites (Asn-65 and Asn-92) were replaced with Asp were generated by PCR-based site-directed mutagenesis. The delCT and delGPI variants of CD317, which lacked the N-terminal 20 amino acids and C-terminal 19 amino acids, respectively, were fused with HA tag in the N or C terminus and cloned into pCMV-C-His or PLVX vector. siRNA-resistant (SR) CD317, delCT, and delGPI constructs, each tagged with HA, were generated via PCR by making three synonymous mutations in the siRNA recognition site of human CD317, and they are called HA-CD317-SR, HA-delCT-SR, and HA-delGPI-SR, respectively. Specific siRNA for human CD317 and nonspecific negative control were described previously (36). For stable transfection, two shRNAs targeting human CD317 (sh317) and control shRNA (shCtrl) were cloned into pLVTHM vectors. Forward oligonucleotide sequences for shRNAs and siRNAs were provided in Supplementary Table S1.Transfection and lentiviral infectionTransfection of tumor cells with plasmids or siRNAs was performed using Lipofectamine 3000 according to the manufacturer\'s protocol (Invitrogen). For lentivirus production, HEK293T cells were transfected with each lentiviral vector together with helper plasmids Gag, Rev, and VSVG. Forty-eight to 72 hours after transfection, virus-containing media was collected by centrifugation at 100 × g for 5 minutes and concentrated by centrifugation at 50,000 × g for 2 hours. Cells were transduced using lentiviruses with 6 μg/mL polybrene and selected with FACS.Cell viability and colony formation assayTwenty-four hours following transfection, Bel7402, Huh7, or HepG2 cells were seeded in triplicates in 96-well plates at 5,000 cells/well and maintained in medium containing 10% FBS. Cells were strained with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Promega) at the indicated time points, and relative viable cells were determined by measuring the OD at an absorbance wavelength of 490 nm.For colony formation assays, transfected Bel7402 or HepG2 cells were plated in triplicates in six-well plate at 1,500 cells/well and maintained in medium containing 10% FBS for 7 to 10 days. Colonies were fixed with methanol and stained with crystal violet staining solution (Beyotime).Cell-cycle and apoptosis analysisCell cycle and apoptosis were assessed by flow cytometry (FACS Canto II; BD Bioscience). For cell-cycle analysis, 48 hours following transfection, Bel7402, HepG2, or Huh7 cells were seeded in triplicates in 12-well plates at 1 × 105 cells/well, synchronized by serum starvation, and allowed to grow for 18 to 24 hours. Cells were collected, washed with PBS, and fixed in cool ethanol at −20°C overnight before being stained with propidium iodide (PI; KeyGEN BioTECH) at room temperature for 30 minutes. For apoptosis analysis, cells were collected, washed with PBS, double-stained with Annexin V-FITC and PI using an Apoptosis Detection Kit (TransGen Biotech) at room temperature for 15 minutes in the dark.RNA isolation and qRT-PCRTotal RNA was extracted with TRIzol reagent (Invitrogen) and used to generate cDNA. Specific primers used for quantitative real-time PCR assays were synthesized by Invitrogen Corporation. Their sequences were shown in Supplementary Table S2.Protein extraction and immunoblottingWhole-cell lysates were prepared by suspending cells in the RIPA buffer (Beyotime) supplemented with 1 × complete protease inhibitors mixture and 1 × phosphatase inhibitor (Roche). Protein concentration was determined by BCA assay (Pierce). Equal quantities of proteins were separated by SDS-PAGE, transferred to a PVDF membrane, and blotted with specific antibodies. Proteins in the membrane were visualized by an enhanced Chemiluminescense Detection Kit (Millipore). Rabbit antibodies against the following proteins or modifications were used with the catalogue numbers and sources indicated: CD317 (ab134061), TGFα (ab208156; Abcam); caspase3 (9662; CST); pY1068 EGFR (BS5010), pY845 EGFR (BS5013), pY705 STAT3 (BS4181), STAT3 (AP0365), ERK1/2 (BS1112), pT202/Y204 ERK1/2 (BS5016), cyclin D1 (BS6352), and p16 INK4a (BS6431), caveolin-1 (BS9878M; Bioworld Technology), and AREG (16036-1-AP; Proteintech). Mouse antibodies against the following proteins/epitopes were used: HA.11 (MMS-101P; Covance); Ki67 (P6834; Sigma); transferrin receptor (13-6800; Invitrogen); GAPDH (MB001; Bioworld Technology); β-actin (sc-47778) and EGFR (sc-373746; Santa Cruz Biotech). HRP-conjugated mouse anti-His (M20020) was purchased from Abmart, mouse HRP-conjugated goat anti-mouse IgG (074-1806) from KPL, and HRP-conjugated goat anti-rabbit IgG (E030120-02) from EARTHOX.Lipid raft and non-raft proteins isolationLipid raft and non-raft proteins were isolated using the Focus Global Fractionation Kit (G Biosciences) according to the manufacturer\'s instruction.CoimmunoprecipitationHepG2 cells were lysed in immunoprecipitation (IP) buffer (150 mmol/L NaCl, 50 mmol/L Tris-HCl, pH 7.4, 50 mmol/L EDTA, 1.0% Nonidet P-40, 1 mmol/L PMSF) supplemented with 1 × complete protease inhibitors mixture (Roche). Extracts were assayed for protein content, using the BCA Protein Assay Kit (Pierce) after clarification by high-speed centrifugation at 4 °C. IP was performed using protein-A/G Dynabeads (Pierce, 88803). In brief, 400 μg protein-A/G Dynabeads was coated with 1 μg specific antibody (Rabbit anti-CD317, sc-99191, or mouse anti-EGFR, sc-373746; Santa Cruz) or Ig control for 1 hour at room temperature with rotation. After removing unbound antibody, the bead–antibody complex was incubated with 500 μL cell lysate for 6 hours at 4 °C with rotation. The captured immunoprecipitates were washed five times with IP buffer and boiled in 2× loading buffer. The eluted proteins were fractionated by SDS-PAGE and detected by Western blot analysis.ELISASupernatant was collected from HepG2 or Bel7402 cells at 48 hours after transfection and kept at −80 °C. The concentration of TGFα and AREG was measured using ELISA kits purchased from ThermoFisher Scientific and BOSTER respectively, according to the manufacturer\'s instruction.Immunofluorescence and microscopyImmunofluorescent staining was performed as previous description (36) using the following antibodies: mouse anti-EGFR (Santa Cruz; sc-120), rabbit anti-pY1068 EGFR (Bioworld Technology; BS5010) and rabbit anti-pY845EGFR (Bioworld Technology; BS5013), rhodamine-conjugated goat anti-rabbit IgG and goat anti-mouse IgG (Molecular Probes). 4, 6-Diamino-2-phenylindole (DAPI; Roche) and Alexa Fluor 488 conjugated Cholera Toxin Subunit B (C-34775; Life Technology) were used to label nuclei and lipid raft, respectively. Ten to fifteen high-powered fields were evaluated on optical microscopy (Olympus IX71), and fixed fluorescence images were analyzed by ImagePro Plus software (Media Cybernetics).Statistical analysisAll analyses were performed using GraphPad Prism Software. Data were expressed as mean ± SEM. Student t test or Mann–Whitney U test was used to compare continuous data for two groups. Pearson correlation co-efficiency was used to analyze the relationship between the phosphor-Y845 EGFR and the CD317 staining levels in tissue sections. Tumor incidences of CD317-overexpression and control group were analyzed using the log-rank test. P values 0.05 were considered to be significant.ResultsCD317 is upregulated in HCCs and enhances tumorigenicityTo investigate the role of CD317 in HCC, we examined its expression in 38 HCC tumor samples and 10 normal liver samples using IHC. As shown in Fig. 1A, the level of CD317 was significantly increased in the majority of the HCC samples compared with the normal liver samples. A survey of a public database (Human Protein Atlas, www.proteinatlas.org) also showed that expression of the CD317 protein was significantly increased in HCCs (Supplementary Fig. S1B). In addition, CD317 mRNA levels were elevated in HCCs compared with normal liver tissues (www.oncomine.org; Supplementary Fig. S1C).Download figureOpen in new tabDownload powerpointFigure 1. Upregulation of CD317 expression correlates with tumorigenesis. A, CD317 upregulation in hepatocellular carcinoma. CD317 protein expression in tumors and normal tissues from patients diagnosed with hepatocellular carcinoma was analyzed by IHC. CD317-positive cells are shown in brown. Quantitation of the CD317 expression in tumors (n = 38) and normal tissues (n = 10) was performed as described in Materials and Methods. Each data point represents a CD317 expression score of patients. Horizontal bars, medians. Scale bar, 100 μm. B, Levels of CD317 protein in the human cell lines. C, Immunoblot analysis of HCC cells with or without CD317 overexpression or knockdown. D, Proliferation of HepG2, Bel7402, and Huh7 cells transiently transfected with control (MigR1) or CD317 plasmid, or with control (siCtrl) or CD317 siRNA (siCD317). *, P 0.05; ***, P 0.001 for CD317 vs. MigR1 group. ##, P 0.005; ###, P 0.001 for CD317 siRNA vs. control siRNA group. E, Colony formation of HepG2 and Bel7402 cells transiently transfected with MigR1, CD317, siCD317, or control siRNA. **, P 0.005; ***, P 0.001. F–I, HepG2 cells transfected with control (PLVX) or CD317 plasmid were injected subcutaneously into nude mice. Shown are tumor incidences (F) and tumor growth (G) overtime and typical whole-body fluorescence images (H, top), tumor appearance (H, bottom), and weight (I; mean ± SEM) at 28 days postinjection. **, P 0.005. J, Total lysates from PLVX and CD317 tumors were analyzed by Western blotting. K–M, HepG2 cells transfected with control (shCtrl), CD317 shRNA-1 (sh317–1), or CD317 shRNA-2 (sh317-2) were injected s.c. into nude mice. Shown are tumor growth (K) overtime and typical whole-body images (L, top), tumor appearance (L, bottom), and weight (M; mean ± SEM) at 23 days postinjection. N, Total lysates from shCtrl, sh317-1, and sh317-2 tumors were analyzed by Western blotting.To test the function of CD317 in HCC cells, we used three cell lines—HepG2, Bel7402, and Huh7, which express low to high levels of endogenous CD317 mRNA and protein (Fig. 1B; Supplementary Fig. S1D). We knocked down CD317 in HepG2 and Bel7402 cells using siRNA (Fig. 1C). This led to a strong reduction in cell proliferation (Fig. 1D and E; Supplementary Fig. S1E). Conversely, we overexpressed exogenous CD317 in HepG2, Bel7402, and Huh7 cells (Fig. 1C), and observed a marked increase in proliferation (Fig. 1D and E; Supplementary Fig. S1E). Neither knockdown nor overexpression of CD317 affected apoptosis (Supplementary Fig. S1F and S1G), consistent with our previous observations (36).To investigate the role of CD317 in tumor formation in vivo, we implanted control and CD317-overexpressing HepG2 cells (Supplementary Fig. S2A) into immunodeficient nude mice. Compared with control cells, HepG2 cells with CD317 overexpression generated tumors with an earlier onset and a faster rate (Fig. 1F and G). By day 28 following xenograft, CD317-overexpressing cells grew into tumors that were three times as large as those generated by control cells (P 0.05, n = 5–6; Fig. 1H and I; Supplementary Fig. S2B). The increased tumorigenic potential of CD317-overexpressing cells was likely due to enhanced proliferation instead of reduced apoptosis, as tumors generated from these cells showed increased Ki-67 levels, but unchanged caspase-3 activation, compared with tumors generated from control cells (Fig. 1J). Conversely, CD317 knockdown markedly suppressed tumor growth (Fig. 1K–M; Supplementary Fig. S2C) and reduced proliferation, characterizing by low PCNA expression (Fig. 1N). These results indicate that CD317 is required for optimal proliferation and tumor formation of HCC cells.CD317 promotes cell-cycle progression in HCC cellsGiven that CD317 promotes cell proliferation, we analyzed how it affects cell-cycle progression. Compared with their corresponding control cells, CD317-overexpressing HepG2, Bel7402, and Huh7 cells displayed accelerated cell-cycle progression, as evident by a noticeable increase in S-phase cells and a concomitant decrease in G0–G1-phase cells (Fig. 2A; Supplementary Fig. S3A). Conversely, CD317-depleted HepG2 and Bel7402 cells showed a significant reduction in cell-cycle progression, with fewer S-phase cells and more G0–G1-phase cells (Fig. 2B; Supplementary Fig. S3B).Download figureOpen in new tabDownload powerpointFigure 2. CD317 accelerates cell-cycle transition in vitro. A and B, HepG2, Bel7402, and Huh7 cells were transfected with the control vector (MigR1) or either CD317 (A), or HepG2 and Bel7402 cells were transfected with control or CD317 siRNA (B). Percentage of cells in G0–G1, S, and G2–M phase was quantified (means ± SEM). **, P 0.005; ***, P 0.001. C and D, Expression of cell cycle-related proteins in HepG2, Bel7402, and Huh7 cells transfected with MigR1 or CD317 (C), or in HepG2, Bel7402 cells transfected with control or CD317 siRNA (D). Blots in C and D were exposed for different times to better show the effects of CD317 overexpression or knockdown. E and G, Expression of cell-cycle regulatory proteins in HepG2 cells transfected with MigR1 vector, wild-type CD317, or the indicated CD317 mutants (E), or with the control vector pCMV and CD317-ECD-His (G). In G, the endogenous CD317 and CD317-ECD-His were detected by anti-CD317 antibody, whereas CD317-ECD-His was also detected by anti-His antibody. F and H, Cell-cycle progression of HepG2 cells transfected with MigR1 vector, wild-type CD317, or the indicated CD317 mutants (F), or with the control vector pCMV and CD317-ECD-His (H). Values represent means ± SEM. **, P 0.005; ***, P 0.001. The experiments were repeated at least three times with similar results.Forced expression of CD317 in HepG2, Bel7402, and Huh7 cells elevated the levels of the CDK4/6 activator cyclin D1 and the DNA polymerase cofactor PCNA, while reducing the levels of the CDK inhibitor p16 (Fig. 2C). Conversely, knockdown of CD317 in HepG2 and Bel7402 cells markedly decreased the expression of cyclin D1 and PCNA, while enhancing the expression of p16 (Fig. 2D).Glycosylation and membrane localization are essential for CD317 functionCD317 is a type II transmembrane protein, with its ectodomain containing two N-linked glycosylation sites, Asn65 and Asn92 (Supplementary Fig. S1A). Previous studies suggested that N-linked glycosylation at these residues may be required for the correct folding, but not antiviral activity, of CD317 (37). However, the role of this modification in tumor cells has not been addressed. We mutated Asn65 and Asn92 to Asp individually (N65D and N92D, respectively) or in combination (N65D/N92D). Consistent with a previous report (37), the N65D and N92D mutations reduced the apparent molecular weight of CD317 from 30 to 36 kDa to ∼28 kDa, whereas the N65D/N92D mutation further reduced the apparent molecular weight to ∼21 kDa (Fig. 2E). Unlike wild-type CD317, none of the glycosylation-defective mutants affected the levels of PCNA, cyclin D1, or p16 (Fig. 2E). Consistently, in contrast to wild-type CD317, cells expressing these mutations showed no difference in cell-cycle progression compared with the parental cells (Fig. 2F; Supplementary Fig. S3C). Additionally, we generated CD317-ECD, a truncated mutant that contained only the ectodomain and hence was not anchored in the plasma membrane. CD317-ECD did not affect the progression of cell cycle or the levels of cell-cycle regulators either (Fig. 2G and H; Supplementary Fig. S3D). Thus, both glycosylation and membrane localization are required for the function of CD317 in cell-cycle progression.CD317 activates the EGFR–STAT/ERK pathwayNext, we investigated the mechanism through which CD317 promotes cell proliferation. By analyzing various mitogenic pathways, we observed that CD317 strongly influenced both the RAS–Raf–MEK–ERK and JAK–STAT signaling pathways. Specifically, overexpression of CD317 increased, while knocking down CD317 decreased, the activation of ERK1/2 and STAT3 in HepG2, Bel7402, and Huh7 cells (Fig. 3A). While evaluating the signaling events upstream of both pathways, we noticed that CD317 is a potent activator for EGFR. Specifically, forced expression of CD317 led to a strong increase in the auto-phosphorylation of EGFR at Tyr1068, as well as Src-mediated phosphorylation of EGFR at Tyr845 (Fig. 3B). Conversely, depletion of CD317 by siRNA resulted in a noticeable reduction in these phosphorylation events (Fig. 3B). In contrast, CD317-N65D/N92D, the glycosylation mutant that failed to accelerate cell cycle, did not affect EGFR activation (Fig. 3B).Download figureOpen in new tabDownload powerpointFigure 3. CD317 function is dependent on EGFR–STAT3/ERK axis. A, STAT3 and ERK1/2 phosphorylation levels in HepG2, Bel7402, and Huh7 cells transiently transfected with MigR1 or CD317 (left), or with control siRNA or CD317 siRNA (right). The levels of CD317, pSTAT3, total STAT3, pERK1/2, and total ERK1/2 were determined by Western blot analysis. B, HepG2 cells were transfected with control MigR1 vector, CD317, or CD317 N65/92D (left). Huh7 cells were transfected with control MigR1 vector or CD317 (middle), and Bel7402 cells were transfected with control or CD317 siRNA (right). The levels of CD317, pY845 EGFR, pY1068 EGFR, and total EGFR were analyzed by Western blot analysis. C, Representative FACS graphs (left) and statistical analysis (right) of EGFR phosphorylation levels in HepG2 cells transiently transfected with control MigR1 vector or CD317. Mean fluorescence intensity (MFI) values of GFP-negative and -positive cells was analyzed as indicated. *, P 0.05; **, P 0.005; ***, P 0.001. D, HepG2 cells transfected with MigR1 or CD317 were treated with or without erlotinib (50 μmol/L). Cells were analyzed for the activation of EGFR, STAT3, and ERK1/2 (top), and expression of cell cycle-related proteins (bottom). E and F, HepG2 cells stably infected with control (PLVX) or CD317-expressing lentiviruses and Huh7 cells transfected with control (MigR1) or CD317 vectors were treated with or without erlotinib (50 μmol/L). Cell-cycle distribution (E) and cell proliferation (F) were analyzed. **, P 0.005; ***, P 0.001 for CD317 vs. control group. G, Huh7 cells transfected with control (MigR1) or CD317 vectors were treated with control or EGFR siRNA (siEGFR). Activation of STAT3 and ERK1/2 was analyzed. H–J, HepG2 cells stably infected with PLVX or CD317 lentiviruses were treated with control or EGFR siRNA. Activation of EGFR, STAT3, and ERK1/2 (H, left), expression of cell cycle-related proteins (H, right), cell-cycle distribution (I), and cell proliferation (J) were analyzed. ***, P 0.001 for CD317 vs. PLVX group.To confirm these results, we analyzed the levels of phosphorylated EGFR using immunofluorescence staining and FACS analysis. We transfected HepG2 cells with a CD317 plasmid that coexpressed GFP, as well as the control plasmid expressing only GFP. By staining cells with anti-phospho-Tyr845 (anti-pY845) and anti-pY1068 antibodies, we observed that phosphorylation of endogenous EGFR was significantly enhanced in cells with CD317 overexpression (CD317 group, GFP-positive), but not in cells without CD317 overexpression (CD317 group, GFP-negative; and control group, GFP-positive and -negative; Supplementary Fig. S4A). A FACS analysis further confirmed that forced expression of CD317 enhanced the levels of pY845 and pY1068 EGFR in HepG2 cells (Fig. 3C). Additionally, CD317 knockdown sensitized tumor cells to erlotinib treatment, as shown by a decreased IC50 in CD317-depleted tumor cells (Supplementary Fig. S4B). Moreover, tumors formed by the CD317-overexpressing HepG2 cells showed noticeably higher EGFR activation compared with tumors formed by control HepG2 cells (Fig. 1J), whereas CD317-knockdwon tumors showed substantially lower EGFR activation compared with control tumors (Fig. 1N).To determine whether CD317 activates RAS–Raf–MEK–MAPK and JAK–STAT pathways via EGFR, we treated HepG2 cells with the EGFR inhibitor erlotinib at a dose that almost completely blocked EGFR activation (Fig. 3D). Under this condition, CD317 was no longer able to stimulate the activation of STAT3 and ERK (Fig. 3D, top) or alter the expression of cyclin D1, PCNA, and p16 (Fig. 3D, bottom). Moreover, erlotinib effectively negated CD317-mediated acceleration in cell-cycle progression (Fig. 3E and Supplementary Fig. S4C) and proliferation (Fig. 3F). We also used siRNAs to knock down EGFR in HepG2 and Huh7 cells. This markedly impaired CD317-induced activation of STAT3 and ERK (Fig. 3G and Fig. 3H, left), and effectively abrogated the effect of CD317 on cyclin D1, PCNA, and p16 (Fig. 3H, right). Knocking down EGFR also rendered CD317 incapable of stimulating cell cycle progression (Fig. 3I; Supplementary Fig. S4D) or proliferation (Fig. 3J). Collectively, these data show that CD317 promotes mitogenic signaling, as well as cell-cycle progression and proliferation, via the activation of EGFR.CD317 regulates EGFR in a lipid raft-dependent mannerCD317 did not influence the expression of EGFR (Fig. 4A and B). Moreover, CD317 did not appear to stably associate with EGFR, as shown by a coimmunoprecipitation assay (Fig. 4C). EGFR is activated by binding to its cognate ligands, including EGF, TGFα (5), and AREG (6). However, the expression of EGF was minimal in HepG2 and Bel7402 cells (Fig. 4A and B). Moreover, neither CD317 overexpression (in HepG2 cells) nor knockdown (in Bel7402 cells) significantly altered the protein and mRNA levels of TGFα or AREG (Fig. 4A and B). Moreover, knockdown of both TGFα and AREG did not influence CD317-mediated EGFR activation (Supplementary Fig. S4E), suggesting that CD137-induced EGFR activation is not mediated by these EGFR ligands.Download figureOpen in new tabDownload powerpointFigure 4. CD317 regulates EGFR in a lipid raft-dependent manner. A and B, HepG2 cells were transfected with MigR1 or CD317 plasmid (A), and Bel7402 cells were transfected with control or CD317 siRNA (B). Forty-eight hours later, mRNA levels of CD317, EGFR, EGF, TGFα, and AREG were determined by real-time PCR (left), and the protein levels of TGFα and AREG in culture supernatants were detected by ELISA (right). Values represent means ± SEM. **, P 0.005. The experiments were repeated at least three times with similar results. C, Lysates of HepG2 cells were immunoprecipitated with anti-CD317 or anti-EGFR antibodies as indicated. Cell lysates (input) and immunoprecipitates were analyzed by Western blotting. D, HepG2 cells transfected with control or CD317 siRNA were immunostained for EGFR (red), lipid rafts (green), and DNA (DAPI, blue). Shown are representative fluorescence images and EGFR-lipid rafts colocalization plot (left), and Pearson\'s correlation coefficient (R) (right; mean ± SEM for at least 30 cells). ***, P 0.001. Scale bar, 20 μm. E, HepG2 cells were transfected with control or CD317 siRNA, along with control (pLVX) or siRNA-resistant (SR) CD317, delCT, and delGPI plasmids. EGFR in nonlipid raft (N) and lipid raft fractions were analyzed by Western blot analysis. Transferrin receptor (TfR) and caveolin-1 served as loading controls for non-raft proteins and lipid raft proteins, respectively. F, EGFR activation in CD317-knockdown HepG2 cells expressing the indicated siRNA-resistant CD317 plasmids or the PLVX control vector. G and H, HepG2 cells expressing MigR1, CD317, HA-delCT-CD317 (delCT), and delGPI-CD317-HA (delGPI) (H, right) were analyzed for cell-cycle progression (G) and EGFR activation (H, left). **, P 0.005; ***, P 0.001 for CD317 or delCT vs. PLVX group. I, HepG2 and Huh7 cells were transfected with MigR1 or CD317 plasmid, and Bel7402 cells were transfected with Ctrl or CD317 siRNA and treated with or without 10 mmol/L methyl-β-cyclodextrin (MβCD) for 30 minutes. Thereafter, cholesterol was reloaded for 90 minutes using cholesterol-saturated MβCD (2:10 chol-MβCD, 2 mmol/L cholesterol, and 10 mmol/L MβCD) in Huh7 cells. The levels of CD317, pY845-EGFR, pY1068-EGFR, and total EGFR were determined by Western blot analysis.The activation of EGFR is regulated by its localization to lipid rafts (13, 15, 16). Considering that CD317 is associated with lipid rafts (38), we investigated whether CD317 affects the raft versus non-raft distribution of EGFR. Using a fluorescently-tagged cholera toxin B subunit (CTB) that specifically labels lipid rafts (39), we observed that CD317 knockdown cells exhibited high levels of colocalization of EGFR with lipid rafts, characterized by an increased coefficient value between EGFR and lipid raft staining (Fig. 4D). We also detected EGFR by Western blot analysis and found that knocking down CD317 led to a dramatic re-distribution of EGFR to the lipid raft fraction (Fig. 4E). Upon the expression of an siRNA-resistant form of CD317 in the CD317-knockdown cells, the localization of EGFR was essentially restored to that in control cells (Fig. 4E), confirming the specificity of the CD317 siRNA.The extracellular C-terminal GPI anchor of CD317 enables its association with the lipid rafts, and the cytoplasmic N-terminal region of CD317 links to the actin cytoskeleton (17, 40). We generated siRNA-resistant CD317 variants, delGPI and delCT, in which the GPI modification signal and the cytoplasmic tail were deleted, respectively (Fig. 4E). delCT, like the siRNA-resistant full-length CD317, was able to reverse the association of EGFR with lipid rafts in CD317 knockdown HepG2 cells, whereas delGPI showed no such an activity (Fig. 4E). Also, delCT elicited a similar effect as the full-length CD317 in promoting EGFR activation in HepG2 cells devoid of CD317, but delGPI failed to do so (Fig. 4F). Consistently, delCT, but not delGPI, promoted cell-cycle progression (Fig. 4G; Supplementary Fig. S4F) and EGFR activation (Fig. 4H) as effectively as the full-length CD317.Furthermore, we disrupted lipid rafts by depleting cholesterol with methyl-β-cyclodextrin (MβCD). In accordance with previous reports (41), this treatment resulted in EGFR activation in HepG2, Bel7402, and Huh7 cells (Fig. 4I). Importantly, under this condition, neither CD317 overexpression nor CD317 depletion influenced the activation of EGFR in these cells (Fig. 4I). However, this effect of MβCD on CD317-mediated EGFR activation can be reverted by replenishment with cholesterol (Fig. 4I), further supporting the notion that lipid raft is required for effect of CD317 on EGFR. Collectively, these data indicate that CD317 regulates EGFR activation by promoting the release of EGFR from lipid rafts.Activation of EGFR correlates with CD317 expression in human HCCsTo investigate the role of CD317 in EGFR activation in human tumors, we analyzed CD317 expression, EGFR activation, and hepatocyte proliferation (using nuclear PCNA as a marker) in 110 HCC samples. Interestingly, the activated form of EGFR (pY845 EGFR) positively correlated with the CD317 expression (rs = 0.4370, P 0.0001; Fig. 5A and B). Patients with low CD317 expression (expression score 6, N = 56) also showed reduced nuclear PCNA than patients with high CD317 expression (expression score ≥6, N = 54; Fig. 5B, bottom). These results suggest that CD317 promotes HCC development through EGFR-mediated mitogenic signals.Download figureOpen in new tabDownload powerpointFigure 5. Upregulation of CD317 correlates with the activation of EGFR in HCC. A, Immunohistological staining of CD317, pY845-EGFR, and PCNA in HCC sections. Scale bars in black and white indicate 50 and 20 μm, respectively. B, Statistical results of data shown in A. Shown are correlation co-efficiency between the phosphor-Y845 EGFR and the CD317 staining levels in tissue sections (n = 110; top), and nuclear PCNA expression in CD317high (score ≥ 6; n = 56) and CD317low (score n = 54) patients (bottom). Each data point represents a patient. Horizontal bars, medians. C, Proposed role of CD317 in HCC development. CD317 expression is markedly upregulated in HCC, leading to overactivation of EGFR and the downstream pathways, such as STAT3 and ERK1/2 pathways, by modulating lipid raft dynamic. STAT3 collaborates with ERK1/2 signaling to activate the expression of cyclin D1 and PCNA, and to inhibit p16 expression, which consequently accelerates G1–S phase transition and cell proliferation. Therefore, CD317 promotes HCC development.DiscussionEGFR was among the very first RTKs that were identified and is frequently dysregulated in tumors (5, 42). As such, EGFR has been extensively studied as a prototype for RTK-mediated signaling and a target for cancer therapy (5, 9, 42). Here we reveal that the activation of EGFR is controlled via CD317-mediated release from the lipid rafts (Fig. 5C). This finding has important implications in the development of more effective treatments for EGFR-driven tumors.The activation mechanism of EGFR (and the other EGFR/ErbB family members) is unique among RTKs in several important aspects. The ligands for EGFR are monomers, rather than dimers, and they promote receptor dimerization by inducing a dramatic conformational change that exposes a dimerization region (43). The subsequently EGFR activation involves the formation of an asymmetric dimer of the intracellular kinase domain, instead of auto-phosphorylation in the activation-loop as seen in the other RTKs (43). Importantly, EGFR is associated from lipid raft, and its activation requires its release from these highly organized membrane microdomains (15). Although the molecular basis for this requirement remains unclear, the dramatic conformational changes required for the activation of EGFR might be constrained in lipid rafts.The organizational principle of lipid rafts, as well as the localization of proteins in and out of these ordered lipid domains, is poorly defined. Nevertheless, the ability of CD317 to release EGFR from lipid rafts is likely related to its unique topology. CD317 is thought to be associated with lipid rafts via the GPI anchor, but its TM domain is likely anchored outside lipid rafts (19, 21). Moreover, the rigid CD317 dimers formed by the coiled-coil extracellular domain can further dimerize through the anti-parallel association of the N-terminal region of the dimer (17). As such, CD317 can form a tetrameric complex that may bring micro-lipid rafts close to each other while, at the same time, preventing their coalescing into large assemblies. This may facilitate the release of EGFR into the space between lipid rafts. An alternative, but not mutually exclusive, scenario may be related to the recently-identified palmitoylation of EGFR. Proteins modified by palmitate, like GPI-anchored proteins, are often found in lipid rafts. Interestingly, a recent study showed that EGFR is modified by the palmitoyltransferase DHHC20 in the intracellular tail, which enables the insertion of the disordered region C-terminal to the kinase domain into the plasma membrane (44). Presumably, the C-terminal region of EGFR may insert into lipid rafts, impeding EGFR activation. CD317 might inhibit EGFR palmitoylation or increase its de-palmitoylation, for example, by influencing the interaction of EGFR with acyl-transferases or acyl-protein thioesterases. Regardless of the mechanism, CD317-mediated release of EGFR from lipid rafts likely represents a previously unrecognized, important mode of regulation. Further characterization of this regulation will likely help unravel intricate mechanisms that govern the activation of EGFR and the closely-related RTKs.Constitutive signaling emanating from EGFR and other members of the EGFR/ErbB family contributes to a wide range of malignancies (8). Mechanisms underlying aberrant EGFR signaling include increased expression of EGFR, overproduction of its ligands, and mutations of EGFR especially in the extracellular and kinase domains (5, 45). Here we find that the levels of CD317 proteins are increased in HCC, and a survey of public database show that CD317 transcripts are enhanced in several cancers, including pancreatic and ovarian cancers, suggesting a mechanism for EGFR activation that is distinct from the canonical mode of dysregulation. These cancers with high CD317 expression showed poor prognosis (27, 29, 46, 47), but neutralizing monoclonal antibodies and small molecule tyrosine inhibitors of EGFR offer marginal therapeutic benefit for these tumors. Thus, our findings provide a rationale for targeting CD317 as alternative therapy strategy for EGFR-driven tumors. In this context, it is noted that monoclonal antibodies for CD317 have been generated and tested in animal models for multiple myeloma (31, 32). These antibodies may be applied to treat cancers with CD317 upregulation. Moreover, as shown recently, activation of EGFR via de-palmitoylation renders tumor cells addicted to EGFR, creating a vulnerability (44). Upregulation of CD317 may induce a similar reliance on EGFR signaling, hence sensitizing tumor cells to therapies targeting EGFR. Therefore, for tumors that are initially responsive to therapies targeting EGFR, combination with CD317-targeted therapies may help resolve the problem of resistance in the clinic.Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.Authors\' ContributionsConception and design: G. Zhang, Y.H. Chen, X. Yang, X. WanDevelopment of methodology: G. Zhang, X. LiAcquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): G. Zhang, X. Li, Q. ChenAnalysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): G. Zhang, X. Li, J. Li, X. YangWriting, review, and/or revision of the manuscript: G. Zhang, Y.H. Chen, X. Yang, X. WanAdministrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Q. Chen, Q. RuanStudy supervision: X. Yang, X. WanAcknowledgmentsWe thank Eric Witze for helpful comments on the manuscript. This study was supported by the National Natural Science Foundation of China (NSFC; 81373112 and 81701559) to X. Wan and G. Zhang, respectively; the China Postdoctoral Science Foundation (2016M602541) to G. Zhang; Special Funds for Major Science and Technology of Guangdong Province (2013A022100037); Shenzhen Special Funds for Industry of the Future (Shenzhen Municipal Development and Reform Commission [2015] No. 971); and Shenzhen Basic Science Research Project (JCYJ20170413153158716) to X. Wan; and the U.S. National Institutes of Health (R01CA182675 and R01CA184867) to X. Yang.The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.FootnotesNote: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).Received August 29, 2018.Revision received January 24, 2019.Accepted March 14, 2019.Published first March 19, 2019.©2019 American Association for Cancer Research.References1.↵Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A.Global cancer statistics, 2012.CA Cancer J Clin2015;65:87–108.OpenUrlCrossRefPubMed2.↵Farazi PA, DePinho RA.Hepatocellular carcinoma pathogenesis: from genes to environment.Nat Rev Cancer2006;6:674–87.OpenUrlCrossRefPubMed3.↵Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, et al.Hepatocellular carcinoma.Nat Rev Dis Primers2016;2:16018.OpenUrlPubMed4.↵Mederacke I, Schwabe RF.NAD(+) supplementation as a novel approach to cURIng HCC?Cancer Cell2014;26:777–8.OpenUrl5.↵Yarden Y, Sliwkowski MX.Untangling the ErbB signalling network.Nat Rev Mol Cell Biol2001;2:127–37.OpenUrlCrossRefPubMed6.↵Castillo J, Erroba E, Perugorria MJ, Santamaria M, Lee DC, Prieto J, et al.Amphiregulin contributes to the transformed phenotype of human hepatocellular carcinoma cells.Cancer Res2006;66:6129–38.OpenUrlAbstract/FREE Full Text7.↵Ciardiello F, Tortora G.EGFR antagonists in cancer treatment.N Engl J Med2008;358:1160–74.OpenUrlCrossRefPubMed8.↵Shostak K, Chariot A.EGFR and NF-kappaB: partners in cancer.Trends Mol Med2015;21:385–93.OpenUrlCrossRefPubMed9.↵Chong CR, Janne PA.The quest to overcome resistance to EGFR-targeted therapies in cancer.Nat Med2013;19:1389–400.OpenUrlCrossRefPubMed10.↵Buckley AF, Burgart LJ, Sahai V, Kakar S.Epidermal growth factor receptor expression and gene copy number in conventional hepatocellular carcinoma.Am J Clin Pathol2008;129:245–51.OpenUrlCrossRefPubMed11.↵Whittaker S, Marais R, Zhu AX.The role of signaling pathways in the development and treatment of hepatocellular carcinoma.Oncogene2010;29:4989–5005.OpenUrlCrossRefPubMed12.↵Mallarkey G, Coombes RC.Targeted therapies in medical oncology: successes, failures and next steps.Ther Adv Med Oncol2013;5:5–16.OpenUrlCrossRefPubMed13.↵Mineo C, James GL, Smart EJ, Anderson RG.Localization of epidermal growth factor-stimulated Ras/Raf-1 interaction to caveolae membrane.J Biol Chem1996;271:11930–5.OpenUrlAbstract/FREE Full Text14.↵Lingwood D, Simons K.Lipid rafts as a membrane-organizing principle.Science2010;327:46–50.OpenUrlAbstract/FREE Full Text15.↵Mineo C, Gill GN, Anderson RG.Regulated migration of epidermal growth factor receptor from caveolae.J Biol Chem1999;274:30636–43.OpenUrlAbstract/FREE Full Text16.↵Lambert S, Ameels H, Gniadecki R, Herin M, Poumay Y.Internalization of EGF receptor following lipid rafts disruption in keratinocytes is delayed and dependent on p38 MAPK activation.J Cell Physiol2008;217:834–45.OpenUrlCrossRefPubMed17.↵Schubert HL, Zhai Q, Sandrin V, Eckert DM, Garcia-Maya M, Saul L, et al.Structural and functional studies on the extracellular domain of BST2/tetherin in reduced and oxidized conformations.Proc Natl Acad Sci U S A2010;107:17951–6.OpenUrlAbstract/FREE Full Text18.↵Swiecki M, Scheaffer SM, Allaire M, Fremont DH, Colonna M, Brett TJ.Structural and biophysical analysis of BST-2/tetherin ectodomains reveals an evolutionary conserved design to inhibit virus release.J Biol Chem2011;286:2987–97.OpenUrlAbstract/FREE Full Text19.↵Kupzig S, Korolchuk V, Rollason R, Sugden A, Wilde A, Banting G.Bst-2/HM1.24 is a raft-associated apical membrane protein with an unusual topology.Traffic2003;4:694–709.OpenUrlCrossRefPubMed20.↵Swiecki M, Omattage NS, Brett TJ.BST-2/tetherin: structural biology, viral antagonism, and immunobiology of a potent host antiviral factor.Mol Immunol2013;54:132–9.OpenUrlCrossRefPubMed21.↵Billcliff PG, Gorleku OA, Chamberlain LH, Banting G.The cytosolic N-terminus of CD317/tetherin is a membrane microdomain exclusion motif.Biol Open2013;2:1253–63.OpenUrlAbstract/FREE Full Text22.↵Evans DT, Serra-Moreno R, Singh RK, Guatelli JC.BST-2/tetherin: a new component of the innate immune response to enveloped viruses.Trends Microbiol2010;18:388–96.OpenUrlCrossRefPubMed23.↵Galao RP, Le Tortorec A, Pickering S, Kueck T, Neil SJ.Innate sensing of HIV-1 assembly by Tetherin induces NFkappaB-dependent proinflammatory responses.Cell Host Microbe2012;12:633–44.OpenUrlCrossRefPubMed24.↵Galao RP, Pickering S, Curnock R, Neil SJ.Retroviral retention activates a Syk-dependent HemITAM in human tetherin.Cell Host Microbe2014;16:291–303.OpenUrlCrossRefPubMed25.↵Cocka LJ, Bates P.Identification of alternatively translated Tetherin isoforms with differing antiviral and signaling activities.PLoS Pathog2012;8:e1002931.OpenUrlCrossRefPubMed26.↵Tokarev A, Suarez M, Kwan W, Fitzpatrick K, Singh R, Guatelli J.Stimulation of NF-kappaB activity by the HIV restriction factor BST2.J Virol2013;87:2046–57.OpenUrlAbstract/FREE Full Text27.↵Fang KH, Kao HK, Chi LM, Liang Y, Liu SC, Hseuh C, et al.Overexpression of BST2 is associated with nodal metastasis and poorer prognosis in oral cavity cancer.Laryngoscope2014;124:E354–E360.OpenUrl28.↵Chiang SF, Kan CY, Hsiao YC, Tang R, Hsieh LL, Chiang JM, et al.Bone marrow stromal antigen 2 is a novel plasma biomarker and prognosticator for colorectal carcinoma: a secretome-based verification study.Dis Markers2015;2015:874054.OpenUrl29.↵Mukai S, Oue N, Oshima T, Mukai R, Tatsumoto Y, Sakamoto N, et al.Overexpression of transmembrane protein BST2 is associated with poor survival of patients with esophageal, gastric, or colorectal cancer.Ann Surg Oncol2017;24:594–602.OpenUrl30.↵Gong S, Osei ES, Kaplan D, Chen YH, Meyerson H.CD317 is over-expressed in B-cell chronic lymphocytic leukemia, but not B-cell acute lymphoblastic leukemia.Int J Clin Exp Pathol2015;8:1613–21.OpenUrl31.↵Kawai S, Yoshimura Y, Iida SI, Kinoshita Y, Koishihara Y, Ozaki S, et al.Antitumor activity of humanized monoclonal antibody against HM1.24 antigen in human myeloma xenograft models.Oncology Reports2006;15:361–367.OpenUrlPubMed32.↵Harada T, Ozaki S.Targeted Therapy for HM1.24 (CD317) on Multiple Myeloma Cells.Biomed Res Intl2014;2014:965384. doi: 10.1155/2014/965384.OpenUrl33.↵Zhang G, Hao C, Lou Y, Xi W, Wang X, Wang Y, et al.Tissue-specific expression of TIPE2 provides insights into its function.Mol Immunol2010;47:2435–42.OpenUrlCrossRefPubMed34.↵Yue X, Zhang Z, Liang X, Gao L, Zhang X, Zhao D, et al.Zinc fingers and homeoboxes 2 inhibits hepatocellular carcinoma cell proliferation and represses expression of Cyclins A and E.Gastroenterology2012;142:1559–70.e2.OpenUrlCrossRefPubMed35.↵Yoo H, Park SH, Ye SK, Kim M.IFN-gamma-induced BST2 mediates monocyte adhesion to human endothelial cells.Cell Immunol2011;267:23–9.OpenUrlCrossRefPubMed36.↵Li X, Zhang G, Chen Q, Lin Y, Li J, Ruan Q, et al.CD317 Promotes the survival of cancer cells through apoptosis-inducing factor.J Exp Clin Cancer Res2016;35:117. doi: 10.1186/s13046-016-0391-2.OpenUrl37.↵Perez-Caballero D, Zang T, Ebrahimi A, McNatt MW, Gregory DA, Johnson MC, et al.Tetherin inhibits HIV-1 release by directly tethering virions to cells.Cell2009;139:499–511.OpenUrlCrossRefPubMed38.↵Billcliff PG, Rollason R, Prior I, Owen DM, Gaus K, Banting G.CD317/tetherin is an organiser of membrane microdomains.J Cell Sci2013;126(Pt 7):1553–64.OpenUrlAbstract/FREE Full Text39.↵Harder T, Scheiffele P, Verkade P, Simons K.Lipid domain structure of the plasma membrane revealed by patching of membrane components.J Cell Biol1998;141:929–42.OpenUrlAbstract/FREE Full Text40.↵Rollason R, Korolchuk V, Hamilton C, Jepson M, Banting G.A CD317/tetherin-RICH2 complex plays a critical role in the organization of the subapical actin cytoskeleton in polarized epithelial cells.J Cell Biol2009;184:721–36.OpenUrlAbstract/FREE Full Text41.↵Lambert S, Vind-Kezunovic D, Karvinen S, Gniadecki R.Ligand-independent activation of the EGFR by lipid raft disruption.J Invest Dermatol2006;126:954–62.OpenUrlCrossRefPubMed42.↵Mitsudomi T, Yatabe Y.Epidermal growth factor receptor in relation to tumor development: EGFR gene and cancer.FEBS J2010;277:301–8.OpenUrlCrossRefPubMed43.↵Kaplan M, Narasimhan S, de Heus C, Mance D, van Doorn S, Houben K, et al.EGFR Dynamics Change during Activation in Native Membranes as Revealed by NMR.Cell2016;167:1241–1251.e11.OpenUrlCrossRef44.↵Runkle KB, Kharbanda A, Stypulkowski E, Cao XJ, Wang W, Garcia BA, et al.Inhibition of DHHC20-mediated EGFR palmitoylation creates a dependence on EGFR signaling.Mol Cell2016;62:385–396.OpenUrlCrossRefPubMed45.↵Gullick WJ.Prevalence of aberrant expression of the epidermal growth factor receptor in human cancers.Br Med Bull1991;47:87–98.OpenUrlCrossRefPubMed46.↵Cai DQ, Cao J, Li Z, Zheng X, Yao Y, Li WL, et al.Up-regulation of bone marrow stromal protein 2 (BST2) in breast cancer with bone metastasis.BMC Cancer2009;9:102. doi: 10.1186/1471-2407-9-102.OpenUrlCrossRefPubMed47.↵Sayeed A, Luciani-Torres G, Meng ZH, Bennington JL, Moore DH, Dairkee SH.Aberrant Regulation of the BST2 (Tetherin) Promoter Enhances Cell Proliferation and Apoptosis Evasion in High Grade Breast Cancer Cells.PLoS One2013;8:e67191.OpenUrl Thank you for sharing this Cancer Research article.NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses. Message Body (Your Name) thought you would be interested in this article in Cancer Research.CAPTCHAThis question is for testing whether or not you are a human visitor and to prevent automated spam submissions. CD317 Activates EGFR by Regulating Its Association with Lipid Rafts Guizhong Zhang, Xin Li, Qian Chen, Junxin Li, Qingguo Ruan, Youhai H. Chen, Xiaolu Yang and Xiaochun Wan Cancer Res May 1 2019 (79) (9) 2220-2231; DOI: 10.1158/0008-5472.CAN-18-2603 CD317 Activates EGFR by Regulating Its Association with Lipid Rafts Guizhong Zhang, Xin Li, Qian Chen, Junxin Li, Qingguo Ruan, Youhai H. Chen, Xiaolu Yang and Xiaochun Wan Cancer Res May 1 2019 (79) (9) 2220-2231; DOI: 10.1158/0008-5472.CAN-18-2603 Cancer Research Print ISSN: 0008-5472Journal of Cancer Research ISSN: 0099-7013American Journal of Cancer ISSN: 0099-7374
本文链接: http://origene.immuno-online.com/view-306847198.html
发布于 : 2025-01-10
阅读()
最新动态
1970-01-01
1970-01-01
1970-01-01
1970-01-01
1970-01-01
1970-01-01
1970-01-02
1979-09-22
2010-05-18
2012-04-11
品牌分类
抗体
联络我们