Author Response: Nerve Growth Factor Receptor Negates the Tumor Suppressor P53 As a Feedback Regulator
Xiang Zhou,Qian Hao,Peng Liao,Shunjing Luo,Min-Hong Zhang,Guohui Hu,Hongbing Liu,Yiwei Zhang,Bo Cao,Melody Baddoo,Erik K. Flemington,Shelya X. Zeng,Hua Lu
DOI: https://doi.org/10.7554/elife.15099.020
2016-01-01
Abstract:Full text Figures and data Side by side Abstract eLife digest Introduction Results Discussion Materials and methods Data availability References Decision letter Author response Article and author information Metrics Abstract Cancer develops and progresses often by inactivating p53. Here, we unveil nerve growth factor receptor (NGFR, p75NTR or CD271) as a novel p53 inactivator. p53 activates NGFR transcription, whereas NGFR inactivates p53 by promoting its MDM2-mediated ubiquitin-dependent proteolysis and by directly binding to its central DNA binding domain and preventing its DNA-binding activity. Inversely, NGFR ablation activates p53, consequently inducing apoptosis, attenuating survival, and reducing clonogenic capability of cancer cells, as well as sensitizing human cancer cells to chemotherapeutic agents that induce p53 and suppressing mouse xenograft tumor growth. NGFR is highly expressed in human glioblastomas, and its gene is often amplified in breast cancers with wild type p53. Altogether, our results demonstrate that cancers hijack NGFR as an oncogenic inhibitor of p53. https://doi.org/10.7554/eLife.15099.001 eLife digest Cancer often develops as a result of alterations to “tumor suppressor” genes within cells. This results in the cells growing and dividing too much, which causes a tumor to form. One of the most important tumor suppressor genes produces a protein called p53, which is lost or mutated in roughly half of all human cancers. In the other half of cancers p53 itself is normal, but is often disabled by proteins that promote tumor growth. One of the remaining challenges in the field of cancer research is to identify which proteins inhibit p53 directly. Identifying these proteins would help clarify why many human cancers, such as some brain cancers, breast and skin cancers, often maintain a normal form of the p53 tumor suppressor protein. Zhou et al. now provide evidence that shows that a protein called nerve growth factor receptor (NGFR) is one such protein. NGFR was known to be important for the healthy development of the brain and nervous system. Unexpectedly, however, Zhou et al. found that NGFR binds directly to p53 and disables it in several types of human cancer cells. This finding is likely to be important because NGFR is produced in abnormally high amounts in several human cancer types, including skin, breast, bone and some brain cancers. Reducing the levels of NGFR in cancer cells caused the cells to become more sensitive to some anti-cancer drugs. Overall, the results presented by Zhou et al. suggest that developing new drugs that target NGFR could produce new treatments for human cancers that have a normal form of the gene that produces p53. More experiments are also needed to find out whether NGFR has other ways of promoting the development of cancerous tumors. https://doi.org/10.7554/eLife.15099.002 Introduction Tumorigenesis is highly associated with inactivation of the tumor suppressor p53, as it is mutated in ~50% of all types of human cancers, and its functions are impaired through various mechanisms in the rest of human cancers (Levine and Oren, 2009). p53 executes its tumor suppressive function mainly by inducing the expression of a large number of genes involved in cell cycle control, senescence, apoptosis, ferroptosis, autophagy, and metabolism (Jiang et al., 2015; Kruiswijk et al., 2015). Because of the detrimental effects of p53 on normal cells, the cells have developed mechanisms to monitor its activity, which are often hijacked by cancer cells. One key monitor of p53 is MDM2 (HDM2 in human), an oncoprotein encoded by a p53 transcriptional target gene that is amplified or overexpressed in several human tumors (Fakharzadeh et al., 1991; Momand et al., 1992; Oliner et al., 1992, 1993; Wu et al., 1993). MDM2 inhibits p53 activity primarily by binding to and concealing the N-terminal transcriptional activation (TA) domain of p53 (Oliner et al., 1993) and by mediating its poly-ubiquitination and proteolysis (Fuchs et al., 1998; Haupt et al., 1997; Kubbutat et al., 1997). Genetically, disruption of the TP53 gene completely rescues the lethal phenotype of Mdm2 knockout mice (Jones et al., 1995; Montes de Oca Luna et al., 1995). A myriad of stresses can orchestrate this MDM2-p53 feedback loop. The ARF tumor suppressor directly associates with MDM2 and inhibits MDM2-mediated p53 ubiquitination and degradation upon oncogenic stress (Palmero et al., 1998; Zhang et al., 1998; Zindy et al., 1998). Also, several ribosomal proteins boost p53 activation by untying the MDM2-p53 loop in response to ribosomal or nucleolar stress (Zhang and Lu, 2009; Zhou et al., 2012, 2015a). But, oncogenic proteins can enhance MDM2 E3 ligase activity towards p53. MDMX (also called MDM4), the MDM2 homologue, can enhance MDM2-mediated p53 proteasomal degradation by binding to MDM2, besides directly interacting with p53 and repressing its activity (Shvarts et al., 1996). High expression of MDM2 and MDMX in several cancers, such as breast cancer and melanoma, is often considered as the reason why these cancers sustain wild type (wt) p53 (Wade et al., 2013), but this could only account for a portion of wt p53-harboring cancers. Thus, it is still unknown if there are other proteins that can also suppress p53 function in the remaining cancers. In this study, we revealed a novel feedback regulation of p53 by nerve growth factor receptor (NGFR, also called p75NTR or CD271). NGFR is a 75 kD single-transmembrane protein without kinase activity and widely expressed in the central and peripheral nervous system (Barker, 2004). Often partnering with other receptors, such as TrkA, it is involved in a multitude of processes during neurogenesis, such as neural cell death, neuronal differentiation, neurite growth, and synaptic plasticity (Barker, 2004). Also, the NGF-NGFR cascade activates NF-κB, leading to inhibition of apoptosis (Carter et al., 1996) and increased survival of schwannoma (Ahmad et al., 2014; Gentry et al., 2000) and breast cancer cells (Descamps et al., 2001). In addition, overexpression of NGFR observed in many metastatic cancers promotes tumor migration and invasion (Boiko et al., 2010; Civenni et al., 2011; Johnston et al., 2007). But, in prostate and bladder cancers, NGFR appears to suppress tumor growth and/or metastasis (Krygier and Djakiew, 2002; Tabassum et al., 2003). It remains largely elusive why and how NGFR plays opposite roles in the context of different cancers. These studies together with our initial findings that p53 binds to the NGFR promoter and induces its expression in cancer cells motivated us to further explore the functional interplay between NGFR and p53, and its role in cancer development. As detailed below, we surprisingly found that NGFR inactivates p53 by directly binding to its central DNA-binding domain and preventing its association with its target promoters and by enhancing its MDM2-mediated ubiquitination and proteolysis. This function is ligand-independent because it occurred in the nucleus and without ligand treatment of cancer cells. Biologically, cancer cells hijack the negative feedback regulation of p53 by NGFR to their growth advantage, as down regulation of NGFR induced p53-dependent apoptosis and cell growth arrest as well as suppressed tumor growth. Furthermore, NGFR was found to be highly expressed in 68.75% (33/48) of human gliomas examined. Consistently, NGFR is amplified in breast cancers that harbor wt TP53 based on the TCGA database (Cerami et al., 2012; Gao et al., 2013). Hence, our discovery of NGFR as another feedback suppressor of p53 could explain why some cancers sustain wt p53 and also suggest NGFR as a potential target for the development of new anti-cancer therapy. Results NGFR is a bona fide transcriptional target of p53 From our previous studies to assess the global effects of Inauhzin (INZ) on p53 pathway in cancer cells (Zhang et al., 2012, 2014; Liao et al., 2012), we identified NGFR as a potential p53-regulated gene. To confirm this result, we treated three types of p53-containing cancer cell lines (HCT116p53+/+, H460 and HepG2) with INZ, Doxorubicin (Dox) and 5-Fluorouracil (5-FU). The expression of NGFR mRNA was drastically elevated by all the three agents (Figure 1A,B and C). Consistently, NGFR protein level increased in response to Dox or 5-FU treatment in p53-intact, but not p53-null (HCT116p53-/-) or mutated (PCL/PRF/5), cancer cells (Figure 1D and E). Consistently, ectopic wt, but not mutant, p53 induced NGFR mRNA expression in p53-deficient H1299 and HCT116p53-/- cells (Figure 1F and G). Conversely, knockdown of p53 markedly reduced NGFR mRNA level (Figure 1H and I). These results demonstrate that anti-cancer drug-induced NGFR expression in the cells is p53-dependent. Figure 1 Download asset Open asset p53 transcriptionally induces NGFR expression in cancer cells. (A,B,C) NGFR mRNA expression is elevated by p53-inducing agents. HCT116 p53+/+ (A) H460 (B) and HepG2 (C) cells were treated with Inauhzin, Doxorubicin or 5-Fluorouracil for 15 h, and NGFR expression was determined by q-PCR (Mean ± SEM, n = 3). Three biological replicates (independent experiments) and a two-tailed t-test were used for P value calculation, p*<0.01. Most q-PCR were performed by three biological replicates, each including three technical replicates. (D) NGFR protein expression is elevated by p53-inducing agents in colon cancer cell lines. HCT116 p53+/+ and HCT116 p53-/- cells were treated with Doxorubicin or 5-Fluorouracil for 15 hr followed by IB using antibodies as indicated. (E) NGFR protein expression is elevated by p53-inducing agents in liver cancer cell lines. HepG2 and PLC/PRF/5 cells were treated with Doxorubicin or 5-Fluorouracil for 15 hr followed by IB using antibodies as indicated. (F,G) NGFR mRNA expression is induced by ectopic wild-type, but not mutant, p53. HCT116 p53-/- (F) and H1299 (G) cells were transfected with wild-type or mutant p53 for 30 hr and NGFR expression was determined by q-PCR (Mean ± SEM, n = 3). Three biological replicates were used for p value, p*<0.01. (H,I) NGFR mRNA expression is inhibited by p53 knockdown. HCT116 p53+/+ (H) and H460 (I) cells were transfected with p53 or control siRNA for 72 hr, and Doxorubicin or 5-Fluorouracil was supplemented 15 hr before the cells were harvested for q-PCR (Mean ± SEM, n = 3). Three biological replicates were used for p value, p*<0.05. (J) A schematic of predicted p53 responsive elements in the NGFR promoter. (K,L) p53 induces luciferase activity through RE1. Luciferase assay was performed using H1299 and U2OS cells as described in Materials and methods (Mean ± SEM, n = 3 biological replicates). (M,N) p53 is associated with the NGFR promoter. HCT116 p53+/+ cells were treated with or without Doxorubicin for 15 hr followed by ChIP assay using anti-p53 or mouse IgG. https://doi.org/10.7554/eLife.15099.003 Next, we identified two potential p53 responsive DNA elements (RE) in the NGFR promoter by bioinformatics analysis, RE1 and RE2, at −4826 bp and –6516 bp upstream from the transcriptional initiation site, respectively (Figure 1J). The RE1 was responsive to p53, as ectopic p53 markedly activated luciferase reporter expression through RE1, but not RE2, in H1299 (Figure 1K) and U2OS (Figure 1L) cells. This result was validated by chromatin-associated immunoprecipitation (ChIP) assays after treating HCT116 p53+/+ cells with or without Dox. Endogenous p53 specifically associated with the RE1-containing NGFR promoter, but not a non-related promoter; this association was enhanced by Dox treatment (Figure 1M and N). Thus, these results demonstrate that NGFR is a bona fide transcriptional target of p53. NGFR is required for cancer cell survival and clonogenicity Previous studies showed that NGFR induces apoptosis in some cancer cells, but promotes cell survival and growth in other cancer cells (Molloy et al., 2011). Thus, we re-determined the role of NGFR in cancer cell growth and proliferation and whether this role is p53-dependent. Surprisingly, knockdown of NGFR elicited significant apoptosis of H460 (Figure 2A and B) and HCT116 p53+/+ (Figure 2C and D) cells as measured by flow cytometric analyses, consequently reducing cell viability during the 4-day culture (Figure 2E and F). To verify if NGFR supports tumor cell growth and proliferation and to see if this role is p53-dependent, we performed colony formation assays by knocking down NGFR in p53-containing H460 and p53-deficient H1299 cancer cells (Figure 2—figure supplement 1A). NGFR ablation more dramatically repressed the colony formation of H460 cells than that of H1299 cells (Figure 2G). This result suggests that NGFR may support cancer cell survival through both p53-dependent and independent mechanisms. Since NGFR was found to be required for nerve growth factor-mediated NF-κB activation (Carter et al., 1996), knockdown of NGFR in H1299 may impair NF-κB activation, thus leading to partially restrained cell viability and reduced colonies. Additionally, immunohistochemical (IHC) staining of human gliomas and their adjacent normal tissues exhibited hyperexpression of NGFR in the tumor tissues (Figure 2H and Figure 2—figure supplement 1B). Consistently, NGFR was more highly expressed in human glioma tissues compared to adjacent tissues as determined by immunoblotting (IB) (Figure 2I). Taken together, these results suggest that NGFR promotes cancer cell growth and proliferation, likely in part by inactivating p53. Figure 2 with 1 supplement see all Download asset Open asset NGFR is required for cancer cell survival and clonogenicity, and highly expressed in glioma. (A) NGFR knockdown induces apoptosis of H460 cells. Cells were transfected with NGFR or control siRNA for 72 to 96 hr, and Doxorubicin was supplemented 15 hr before the cells were harvested for flow cytometry analyses. (B) Quantification of Sub-G1 population in (A) (Mean ± SEM, n = 3). Three biological replicates were used for p value, p*<0.05. (C) NGFR knockdown induces apoptosis of HCT116 p53+/+cells. Cells were transfected with NGFR or control siRNA for 72 to 96 hr, and Doxorubicin was supplemented 15 hr before the cells were harvested for flow cytometry analyses. (D) Quantification of Sub-G1 population in (C) (Mean ± SEM, n = 2). Three biological replicates were used for p value, p*<0.05. (E, F) NGFR knockdown suppresses cell survival. H460 (E) and HCT116 p53+/+ (F) cells were transfected with NGFR or control siRNA and seeded in 96-well plate the next day (Day 1). Cell viability was evaluated every 24 hr (Mean ± SEM, n = 3). Three biological replicates were used for p value, p*<0.05. (G) NGFR knockdown inhibits clonogenicity. H460 and H1299 cells were transfected with NGFR or control siRNA and seeded in 10-cm plates the next day. Colonies were fixed by methonal and stained with crystal violet solution (left panel). Quantification of colonies is shown in the right panel (Mean ± SEM, n = 2). Two biological replicates were used for p value, p*<0.05. (H) IHC analyses of human glioma and the adjacent noncancerous tissues. (I) IB analyses of human glioma and the adjacent noncancerous tissues (left panel). Quantification of NGFR expression (right panel) (Mean ± SEM, n = 48, p*<0.01). https://doi.org/10.7554/eLife.15099.004 NGFR inactivates p53 by enhancing its MDM2-mediated ubiquitination and proteolysis To test if NGFR negates p53 activity, we first checked if NGFR affects p53 protein level in H460 cells that were treated with Dox or 5-FU. Indeed, p53 level induced by Dox or 5-FU was markedly reduced by ectopic NGFR (Figure 3A). Inversely, knockdown of NGFR strikingly elevated the level of endogenous p53 and that of its target genes p21 and PUMA in human p53-containing H460, HepG2, neuroblastoma SK-N-SH, melanoma SK-MEL-103 and SK-MEL-147 and HCT116 p53+/+ cell lines (Figure 3B), whereas did not affect PUMA expression in HCT116 p53-/- cells (Figure 3—figure supplement 1). Consistently, knockdown of NGFR led to the global expression of a group of known p53 target genes, as measured by RNA-seq analysis in H460 cells (Figure 3C). This result was further confirmed by utilizing a second siRNA (siNGFR#2), as NGFR knockdown by this siRNA also induced the protein level of p53 and the mRNA levels of its target genes, including BAX, BTG2, MDM2, p21, and PUMA (Figure 3D). Also, ablation of NGFR prolonged p53’s half-life (Figure 3E), suggesting that NGFR might regulate the stability of p53. Next, we tested if NGFR affects MDM2-induced p53 ubiquitination by conducting a p53 ubiquitination assay in H1299 cells with ectopic proteins. Surprisingly, ectopic NGFR enhanced MDM2-mediated p53 ubiquitination in a dose-dependent manner (Figure 3F), although this protein was found to mainly reside in the cytoplasmic membrane (Barker, 2004). Consistently, NGFR also enhanced MDM2-mediated p53 proteasomal degradation, which was abolished by the proteasome inhibitor MG132 (Figure 3G). Without MDM2, NGFR was unable to alter p53 protein level in cells (Figure 4). Altogether, these results indicate that NGFR reduces p53 stability by enhancing its MDM2-mediated ubiquitination and degradation. Figure 3 with 1 supplement see all Download asset Open asset NGFR suppresses p53 activity by enhancing MDM2-mediated ubiquitination and proteasomal degradation. (A) NGFR inhibits p53 activation by Doxorubicin or 5-Fluorouracil. H460 cells were transfected with NGFR for 30 hr and treated with Doxorubicin or 5-Fluorouracil for 15 hr before harvested for IB using antibodies as indicated. (B) NGFR knockdown induces p53 expression and activity. A panel of cancer cell lines were transfected with NGFR or control siRNA followed by IB using antibodies as indicated. The values in the rightmost panel indicate the p53/β-actin ratios. (C) NGFR knockdown induces p53 target gene expression. H460 cells were transfected with NGFR or control siRNA followed by RNA-sequencing analyses. Genes with over 1.5-fold increase in expression were shown (Three biological replicates were used for p value, p<0.05. (D) Two siRNAs targeting different sequences were used for knocking down NGFR in H460 cells. The expression of p53 target genes was assessed by q-PCR (Mean ± SEM, n = 2 biological replicates), while p53 expression was detected by IB. Validation of NGFR knockdown by the two siRNAs is shown in the right corner. (E) NGFR knockdown prolongs p53’s half-life. H460 cells transfected with NGFR or control siRNA for 72 hr were treated with 100 µg/ml of CHX and harvested at the time points as indicated. IB was performed using antibodies as indicated (upper panel) and quantification of p53/β-actin ratio is shown in the lower panel. (F) NGFR promotes MDM2-induced p53 ubiquitination. H1299 cells were transfected with combinations of plasmids encoding p53, HA-MDM2, Myc-NGFR or His-Ub and treated with MG132 6 hr before harvested for in vivo ubiquitination assay. Bound proteins and inputs were detected by IB using antibodies as indicated. (G) NGFR enhancesMDM2-mediated p53 proteasomal degradation. H1299 cells were transfected with combinations of plasmids encoding p53, HA-MDM2 or RFP-NGFR followed by IB using antibodies as indicated. MG132 was supplemented to the medium for 6 hr. https://doi.org/10.7554/eLife.15099.006 Figure 4 with 1 supplement see all Download asset Open asset NGFR inactivates p53 independently of MDM2. (A) NGFR interacts with p53 in the absence of MDM2. MEF p53-/-;Mdm2-/- cells were transfected with plasmids encoding Myc-NGFR or p53 followed by co-IP-IB assays using antibodies as indicated. (B) NGFR does not affect p53 protein expression in the absence of MDM2. MEF p53-/-;Mdm2-/- cells were transfected with combinations of plasmids encoding Myc-NGFR, HA-MDM2 or p53 followed by IB using antibodies as indicated. (C) NGFR represses p53-induced luciferase activity independently of MDM2. MEF p53-/-;Mdm2-/- cells were transfected with plasmids as indicated in the figure and luciferase assay was performed as described in Materials and methods (Mean ± SEM, n = 3). Three biological replicates were used for p value, p*<0.05. (D) NGFR inhibits p53-induced target gene expression independently of MDM2. MEF p53-/-;Mdm2-/- cells were transfected with plasmids as indicated in the figure followed by q-PCR analysis (Mean ± SD, n = 3). Three technical replicates were used for p value, p*<0.05. (E) NGFR impedes p53 association with the p21 promoter in H1299 cells. Cells were transfected with plasmids as indicated and treated with MG132 for 6 hr before harvested for ChIP-PCR analyses. (F) NGFR impedes p53 association with the p21 and Bax promoters independently of MDM2. MEF p53-/-;Mdm2-/- cells were transfected with plasmids as indicated followed by ChIP-PCR analyses. https://doi.org/10.7554/eLife.15099.008 NGFR regulates p53 stability by binding to MDM2 Next, we tested if NGFR interacts with MDM2 by conducting a set of reciprocal co-immunoprecipitation (co-IP) assays. Indeed, ectopic NGFR bound to ectopic MDM2, and vice versa (Figure 5A). We then mapped their binding domains by performing a set of co-IP and GST-pull down assays. NGFR was specifically pulled down with the C-terminal aa 284–491 fragment, but not N-terminal fragments, of MDM2, which encompasses the zinc-finger, acidic, and ring-finger domains, in H1299 cells (Figure 5C). This result was verified by in vitro GST-pull down assays (Figure 5D). Using the same approach, we also mapped the MDM2-binding domain of NGFR. Unexpectedly, MDM2 interacted with the N-terminal extracellular and transmembrane domain, but not the C-terminal cytoplasmic domain, of NGFR (Figure 5E). Also, we validated the interaction between endogenous NGFR and MDM2 in human neuroblastoma SK-N-SH cells that sustains high level of NGFR (Figure 5H). To determine if this interaction takes place in the nucleus, we used H460 cells stably expressing NGFR, HCT116 p53+/+ and SK-MEL-147 cells, fractionated them, and conducted IB assays. Indeed, full-length NGFR was present in both cytoplasm and nucleus (Figure 5—figure supplement 1A–C). Consistently, confocal microscopy analysis revealed that NGFR is present in all of the cellular compartments of SK-MEL-147 cells, albeit with less intensity in the nucleus than that in the cytoplasm (Figure 5—figure supplement 1D). Of note, the nuclear and cytoplasmic levels of NGFR increased upon Dox treatment likely due to p53 activation (Figure 5—figure supplement 1D). Also, NGFR was co-immunoprecipitated with MDM2 from the nuclear extracts of NGFR-stably expressed H460 (Figure 5I) and SK-MEL-147 cells in a reciprocal pattern (Figure 5J). Hence, these results demonstrate that the N-terminus of NGFR binds to the C-terminus of MDM2, and this interaction occurs in the nucleus. Figure 5 with 1 supplement see all Download asset Open asset NGFR interacts with MDM2 in the nucleus. (A,B) NGFR interacts with MDM2. H1299 cells were transfected with plasmids encoding Myc-NGFR, Flag-MDM2 or HA-MDM2 followed by co-IP-IB assays using antibodies as indicated. (C) Mapping the NGFR binding domain on MDM2 by co-IP-IB assays. H1299 cells were transfected with the plasmid encoding V5-tagged MDM2 fragment, aa 1–150, aa 1–301 or aa 284–491, along with the Myc-NGFR-encoded plasmid. Co-IP-IB assays were performed using antibodies as indicated. (D) Mapping the NGFR binding domain on MDM2 by GST-pull down assay. The prokaryotic expressed GST-tagged MDM2 fragment, aa 1–150, aa 1–301 or aa 284–491, or GST protein alone was incubated with cell lysates overexpressing Myc-NGFR. Bound proteins were detected by IB using anti-NGFR or coomassie staining. (E) Mapping the MDM2 binding domain on NGFR. H1299 cells were transfected with the plasmid encoding Myc-tagged NGFR fragment, aa 1–272 or aa 273–427, along with the Flag-MDM2-encoded plasmid. Co-IP-IB assays were performed using antibodies as indicated. (F) A schematic of NGFR binding region on MDM2. (G) A schematic of MDM2 binding region on NGFR. (H) Endogenous interaction of NGFR and MDM2 in SK-N-SH cells. Cells treated with Doxorubicin for 12 hr and MG132 for 6 hr were harvested for co-IP-IB assays using antibodies as indicated. (I,J,K) NGFR interacts with both MDM2 and p53 in the nucleus. Nuclear fractions from NGFR-stably expressed H460 (I) and SK-MEL-147 cells (J,K) were subjected to co-IP-IB assays using antibodies as indicated. https://doi.org/10.7554/eLife.15099.010 NGFR interacts with p53 While testing the endogenous NGFR-MDM2 binding, we also detected p53 in the NGFR-MDM2 complex (Figure 5H–K). Thus, we tested if p53 binds to NGFR directly or indirectly through MDM2. First, we conducted co-IP-IB and GST-Pull down assays. Indeed, NGFR and p53 co-immunoprecipitated with each other in reciprocal co-IP-IB assays (Figure 6A–B). Further confirming this interaction was the mapping of the NGFR-binding domain to the central DNA-binding domain of p53 using a similar co-IP-IB assay by transfecting plasmids encoding individual p53 fragments together with NGFR plasmid into H1299 cells (Figure 6C). This mapping was validated by a GST-pull down assay using purified GST-p53 fusion proteins with different p53 fragments. NGFR specifically bound to the full-length p53 and its central DNA-binding domain (aa 101–300) (Figure 6D). This finding explained why NGFR, MDM2 and p53 could form a ternary complex (Figure 5H–k). We also mapped the p53-binding domain of NGFR by conducting a set of co-IP-IB assays. Like MDM2, p53 also bound to the N-terminal region of NGFR (Figure 6E). Together with the result showing that NGFR binds to p53 in the nucleus (Figure 5I–K), these results demonstrate that the N-terminus of NGFR binds to the central DNA-binding domain of p53, and this interaction also occurs in the nucleus. Figure 6 Download asset Open asset NGFR interacts with p53 in the nucleus. (A, B) NGFR interacts with p53. H1299 cells were transfected with plasmids encoding Myc-NGFR, Flag-p53 or p53 followed by co-IP-IB assays using antibodies as indicated. (C) Mapping the NGFR binding domain on p53 by co-IP-IB assays. H1299 cells were transfected with the plasmid encoding flag-tagged p53 fragment, aa 1–300, aa 101–300, aa 101–393 or aa 301–393, along with the Myc-NGFR-encoded plasmid. Co-IP-IB assays were performed using antibodies as indicated. (D) Mapping the NGFR binding domain on p53 by GST-pull down assay. The prokaryotic expressed GST-tagged full-length p53 or p53 fragment, aa 1–73, aa 100–290 or aa 291–393, or GST protein alone was incubated with cell lysates overexpressing Myc-NGFR. Bound proteins were detected by IB using anti-NGFR or coomassie staining. (E) Mapping the p53-binding domain on NGFR. H1299 cells were transfected with the plasmid encoding Myc-tagged NGFR fragment, aa 1–272 or aa 273–427, along with the V5-p53-encoded plasmid. Co-IP-IB assays were performed using antibodies as indicated. (F) A schematic of NGFR binding region on p53. (G) A schematic of p53 binding region on NGFR. https://doi.org/10.7554/eLife.15099.012 NGFR attenuates p53 activity independently of MDM2 The finding that NGFR binds to the central DNA-binding domain of p53 suggests that this protein might directly regulate p53 transcriptional activity independently of MDM2. To test this idea, we first determined if NGFR can bind to p53 in the absence of MDM2 by introducing their expression plasmids alone or together into MEFMdm2-/-;p53-/- cells followed by co-IP-IB assays. Indeed, NGFR was co-immunoprecipitated with p53 in these cells (Figure 4A). Interestingly and as also mentioned above, ectopic NGFR at different doses failed to decrease the level of ectopic p53 in MEFMdm2-/-;p53-/- cells (Figure 4B), suggesting that NGFR cannot directly degrade p53 without MDM2. Then, we tested if NGFR could regulate p53 transcriptional activity in the absence of MDM2 by performing luciferase reporter assays. Interestingly, ectopic NGFR inhibited p53-induced luciferase activity driven by the miR-34a promoter in MEFMdm2-/-;p53-/- cells in a dose dependent fashion (Figure 4C). Also, NGFR repressed ectopic p53-induced expression of p21 and Puma in MEFMdm2-/-;p53-/- cells (Figure 4D). Consistently, ectopic NGFR markedly reduced the association of p53 with the endogenous p21 and Bax promoters in these cells as analyzed by ChIP assays in H1299 or MEFMdm2-/-;p53-/- cells (Figure 4E). Since MDMX, the partner of MDM2, also bound to NGFR (Figure 4—figure supplement 1A), MEFMdm2-/-; Mdmx-/-; p53-/- cells were used to determine if MDMX is required for p53 inactivation by NGFR. NGFR still inhibited p53 activity as measured by p21 and Puma expression (Figure 4—figure supplement 1B). Collectively, these results demonstrate that NGFR also inhibits p53 by directly binding to its central DNA-binding domain and preventing its association with its target promoter. Depletion of NGFR sensitizes tumor cells to cisplatin and doxorubicin The discovery that NGFR is an inhibitor of p53 in cancer cells suggested the possibility of that depletion of this protein could sensitize cancer cells to chemotherapeutic agents. We tested this idea by treating H460 and SK-N-SH cells with different doses of Dox and Cisplatin after introducing control or NGFR siRNAs into the cells. Indeed, both of the chemotherapeutic drugs induced p53 and its target PUMA or p21 more apparently when the cells were transfected with NGFR siRNA compared to those with control siRNA in