Abstract
Inherited activating mutations in RET predispose humans to Multiple Endocrine Neoplasia type‐2 (MEN2). The MEN2A‐specific mutation RETC634R, RET2A, has been shown to simultaneously downregulate the CDKIs p18 and p27, and upregulate cyclin D1. Importantly, the loss of p18 is necessary and sufficient for RET2A‐mediated hyperproliferation. The loss of N‐Myc in mice results in embryonic lethality due to a lack of neuronal progenitor cells that fail to proliferate, correlate with accumulation of p18 and p27. Therefore, N‐Myc may regulate expression of both CDKIs. Also, N‐Myc is expressed predominantly in neuroendocrine cells that give rise to the primary cell types affected in MEN2A. Together these studies suggest that N‐Myc is a downstream target of RET2A signaling that prevents accumulation of p18 and/or p27. We report that MAPK activation by RET2A leads to a transient induction of N‐Myc mRNA and protein levels, and that N‐Myc induction is required to maintain low p18 and p27 levels. Induced N‐Myc levels correlate with increased binding of N‐Myc to an initiator consensus binding site in the p18 promoter, and this binding is essential for RET2A‐mediated transcriptional regulation of p18. Finally, loss of N‐Myc induction prevents RET2A‐mediated hyperproliferation. Our results demonstrate for the first time that N‐Myc is a downstream target of RET2A signaling, and propose that induction of N‐Myc by RET2A is a key step leading to lower p18 levels during MEN2A tumorigenesis.
Keywords: Cell cycle, p18<sup>Ink4c</sup>, N-Myc, RET, Transcription, <i>Initiator</i> element
Abbreviations
- MEN2
multiple endocrine neoplasia type-2
- CDK
cyclin dependent kinase
- CDKI
CDK inhibitor
- PTC
papillary thyroid carcinoma
- MTC
medullary thyroid carcinoma
- PC
pheochromocytoma
- TF
transcription factor
- inr
initiator
- NPC
neuronal progenitor cells
- GNP
granule neuron progenitor
- qPCR
quantitative polymerase chain reaction
1. Introduction
The mammalian mitotic cell cycle is divided into four distinct phases, G1, S, G2 and M (Sherr, 1994). Progression through the cell cycle is regulated by the activation and subsequent inactivation of a family of serine/threonine kinases known as Cyclin Dependent Kinases (CDKs) (Sherr and Roberts, 1999). In the G1 phase, positive and negative growth signals regulate CDK activity through numerous mechanisms such as changes in the expression patterns of cyclins and CDK Inhibitors (CDKIs) (Sherr, 1993). Association of a CDK with a positively regulating Cyclin D or E subunit leads to its activation, whereas binding with CDKIs allows for negative CDK regulation (Sherr and Roberts, 1999). Based on binding properties, the CDKIs are divided into two families, the Inhibitor of Kinase 4 (Ink4: p16Ink4a, p15Ink4b, p18Ink4c and p19Ink4d) and CDK/Kinase Inhibitory Proteins (Cip/Kip: p21Cip1, p27Kip1 and p57).
Intuitively, overexpression of positively regulating cyclins or loss of negatively regulating CDKIs should lead to uncontrolled growth and tumorigenesis (Sherr, 1996). Indeed, in mice, loss of both p18Ink4c and p27Kip1 (hereafter referred to as p18 and p27 respectively) leads to a tumor phenotype of primarily endocrine organs including the pituitary, pancreas, thyroid, parathyroid and adrenal glands (Franklin et al., 1998, 2000). This multiple tumor phenotype mimics the tumor spectrum seen in human Multiple Endocrine Neoplasia type‐1 and type‐2 combined (MEN1 and MEN2). This suggests that p18 and p27 are regulated by the protein products of MEN1 and RET, mutations in which, are linked to MEN1 and MEN2 syndromes respectively.
RET is a proto‐oncogene encoding a transmembrane receptor tyrosine kinase that is activated by binding to its ligands and co‐receptors (Kodama et al., 2005). Constitutively activating mutations in RET, e.g. the RET/PTC translocation found in the Papillary Thyroid Carcinoma (PTC) patient‐derived RET/PTC cell line, results in constitutive oncogenic activation of RET and leads to a MAPK pathway‐mediated decrease of p27 (Vitagliano et al., 2004). We have previously shown that expression of another constitutively active mutant, RETC634R (RET2A), leads to increased MAPK activation and decreased p18 and p27 levels which correlate with increased proliferation (Joshi et al., 2007). We also showed that low p18 levels are critical for RET2A‐mediated hyperproliferation which underscores the importance of p18 regulation in RET2A‐mediated oncogenesis. In patient‐derived medullary thyroid carcinoma (MTC) and pheochromocytoma (PC) samples, only 13.8% (4 of 29 MTC and PC combined) tumors harbor somatic inactivating mutations in p18 (van Veelen et al., 2009). Furthermore, in all 4 tumors, the inactivating p18 mutations are in a heterozygous state and coincide with a germline RET2A mutation, indicating that the remaining wildtype p18 allele in the tumors may be epigenetically downregulated by oncogenic RET.
The mechanism of p18 regulation is complex and remains largely unexplored. Phelps et al. (1998) have reported that p18 is differentially regulated at both transcriptional and translational levels in C2C12 murine myoblasts. It is transcribed as two distinct mRNA species, a short 1.2 kb p18S and a long 2.4 kb p18L transcript. Although it is known that only one transcript predominates, relevant promoters and regulatory elements remain ill defined. Translationally, only p18S encodes p18 protein (Phelps et al., 1998). Using various human breast cancer cell lines, Blais et al. (2002) showed that E2F1 and sp1 cooperatively enhance p18L transcription however, they were unable to detect the p18S transcript in their model system. In luminal A breast tumors, Pei et al. (2009) show an inverse correlation in GATA3 and p18 expression and increased GATA3 binding to consensus binding sites in the p18 promoter region. However, direct transcriptional response in terms of p18S or p18L transcript levels was not assessed. We have previously reported that expression of RET2A leads to decreased levels of the p18S transcript but not p18L (Joshi et al., 2007). Therefore in order to understand RET2A‐mediated effects on p18S transcription, in this study, we carried out detailed analysis of the putative p18S promoter. Specifically, we searched for a bona fide target transcription factor of RET2A signaling, which could prevent accumulation of p18S mRNA. We report for the first time that the transcription factor N‐Myc is a target of RET2A signaling and that it is induced in a MAPK dependent manner by RET2A. Increased binding of N‐Myc to an initiator (inr) consensus binding site within the p18S promoter in response to RET2A expression leads to maintenance of low p18S mRNA (and protein) levels, correlate with increased proliferation. Lastly, we show that the RET2A‐mediated transient induction of N‐Myc is required for its effects on p18S transcription and hyperproliferation.
2. Results
2.1. Search for a putative transcription factor target of RET2A that mediates p18 transcriptional regulation
To understand how RET2A regulates p18S transcription, we searched for putative transcription factor (TF) targets of RET2A signaling that could bind to the p18 promoter. A detailed search for highly conserved cis‐acting elements across human, mouse and rat genomes in a 5500 basepair region upstream of the p18 translation start site (TSS, set to +1) uncovered the presence of several consensus TF binding sites (method presented in Supplementary methods and data shown in Supplementary Table 1). We used three criteria that would qualify a TF as a likely candidate for RET2A‐mediated p18 transcriptional regulation. First, we predicted that a bona fide downstream target of RET2A signaling capable of regulating p18S transcription would be expressed in adrenal chromaffin and thyroid parafollicular C cells or in endocrine organs. Developmentally, adrenal chromaffin and thyroid parafollicular C cells derive from Neuronal Progenitor Cells (NPCs) and it is known that during murine development, N‐Myc is expressed exclusively in NPCs and other neuroendocrine cells (Downs et al., 1989). Second, N‐Myc is regulated by the MAPK pathway which is required for p18 regulation by RET2A. Treatment of KP‐N‐RT human neuroblastoma cells with IGF‐I results in a MAPK dependent N‐Myc induction (Misawa et al., 2000). Thirdly, in developing cerebella, loss of N‐Myc results in accumulation of p18 and p27, correlate with growth arrest in cerebellar Granule Neuron Progenitors (GNPs) (Zindy et al., 2006). In early neural development, expression of N‐Myc in NPCs is required to maintain low p18 and p27 levels correlate with rapid NPC proliferation (Knoepfler et al., 2002). Loss of both p18 and p27 expression in an N‐Myc null background restores normal proliferation of the GNPs, suggesting that during embryonic development, both p18 and p27 are regulated by N‐Myc to promote proliferation. Taken together, these studies make N‐Myc a likely candidate to study as a downstream intermediate in RET2A/MAPK‐dependent p18 transcriptional repression. N‐Myc has been shown to act as a transcriptional inhibitor by binding to an inr consensus sequence, although the commonly accepted role of N‐Myc is enhancer box (E‐box)–mediated transcriptional activation of target genes (Blackwell et al., 1993; Eberhardy et al., 2000). Our sequence analysis of the p18 promoter region revealed presence of two loosely conserved palindromic inr elements at positions −166 to −161 and −160 to −155 relative to the p18S TSS (Figure 1A). Typically, the sequence of an inr element resembles 5′‐YYAN(A/T)YY‐3′, with considerable variation in conservation as well as identity. The 5′‐GGCAGGCCGACC‐3′ (−167 to −156) sequence of the p18 promoter represents two weak palindromic consensus inr sequences. Both the inr elements, CCGTCC (complimentary strand) and CCGACC, fit the YYAN(A/T)YY consensus sequence, except both are missing the A in the 3rd position. Therefore, we hypothesized that RET2A signaling induces N‐Myc expression, which in turn prevents transcription of p18S.
Figure 1.
RET2A expression correlates with induction of N‐Myc at the protein and mRNA levels. (A) Pictorial representation of 3 kb genomic sequences 5′ to the +1 position of the p18 start codon (ATG) designated as +1. Black boxes denote Exon1 and Exon2 of p18. The N‐Myc binding site is shown in intron1. (B) RET2A expression leads to transient induction of N‐Myc protein and altered cell cycle expression profiles. RET2A cells grown to 90% confluency were treated with Tet (+Tet) or ethanol (−Tet) and grown to 24 h post‐confluency. Protein lysates harvested at indicated timepoints were used for immunoblotting. (C) N‐Myc induction correlates with phosphorylation/activation of ERK and altered cell cycle expression profiles. Cells were treated as described in panel B. (D) Quantitation of western analysis from panel C. (E) Quantitation of mRNA levels from short timecourse. Cells were treated like described in panel C. Relative mRNA levels of indicated genes were determined by QRT‐PCR. ∗ indicates p < 0.05. Results are representative of three independent experiments. All quantitations are average of three independent experiments. Error bars indicate standard deviation.
2.2. RET2A expression leads to a MAPK pathway‐dependent induction of N‐Myc at the mRNA and protein levels
In our previously described tetracycline (Tet) inducible NIH3T3 cell based expression system, in order for p18 and p27 levels to remain low in post‐confluent culture conditions, RET2A must continually be expressed starting in sub‐confluent culture conditions (Joshi et al., 2007). If RET2A expression is initiated after post‐confluent growth arrest is established, p18 and p27 accumulate, remain high, and the levels of neither CDKI can be lowered by RET2A signaling. This suggests two important steps in RET2A‐dependent p18 regulation. First, RET2A signaling prevents induction of p18 rather than actively repressing accumulated p18. Second, the p18 regulatory mechanism leading to high p18 levels in growth arrested cells is active in sub‐confluent conditions, prior to initiation of growth arrest. The latter is supported by similar observations in another type of growth arrest, which is mediated by serum starvation. RET2A expression in serum starvation‐mediated growth arrested cells fails to reduce p18 and p27 levels unless RET2A expression is induced prior to serum starvation.
To determine whether RET2A expression can induce N‐Myc levels, we assayed N‐Myc protein levels starting from sub‐confluent growth conditions in our Tet‐inducible RET2A expression system. This system is particularly suited for such analysis because other available anterior pituitary adenoma cell lines or pheochromocytoma cell lines have impaired p18 expression which makes them ineffective for p18 regulation studies (Acton et al., 2004; Joshi et al., 2007). Total protein lysates from RET2A cells treated with Tet (+Tet), beginning at 80% confluency (Figure 1B 0 h), were harvested at 12 h intervals until the cells reached a 36 h post‐confluency timepoint (48 h). Compared to the −Tet controls, N‐Myc protein levels are increased approximately six‐fold at 12 h and drop to undetectable levels by 24 h (12 h post‐confluency) (Figure 1B). The 12 h timepoint is when cells become 100% confluent and expression of N‐Myc at 12 h is timed perfectly if N‐Myc prevents p18S mRNA accumulation in a RET2A‐dependent manner. Any later would be at a post‐confluency timepoint when RET2A cannot prevent p18 induction. In the absence of RET2A (−Tet), p18 (and p27) protein levels increase at 24, 36 and 48 h, while RET2A expression leads to a three‐fold decrease in both p18 and p27 at the same timepoints. Consistent with previous observations, Cyclin D1/D2 protein levels were approximately three‐fold increased at 12, 24, 36 and 48 h.
Being an immediate early gene, it is possible that N‐Myc is induced prior to 12 h in response to RET2A expression. To test this, we performed a shortened timecourse experiment where we harvested protein lysates at 80% confluency (0 h) and at 3, 6, 12 (100% confluency) and 24 h. We detected a four‐fold induction of N‐Myc protein at 6 h, and a six‐fold induction at 12 h (Figure 1C, quantitated in 1D). Consistent with Figure 1B, N‐Myc was not induced at 24 h. Again expression of N‐Myc prior to 100% confluency is appropriate timing for N‐Myc to prevent the beginning of p18 (and p27) mRNA and/or protein accumulation. Activation of the MAPK pathway in RET2A‐expressing cells, detected by a phospho‐specific ERK antibody, can be seen as early as 3 h and lasts throughout the timecourse. At post‐confluent timepoints, compared to −Tet samples, we detected approximately three‐fold lower levels of p18 and p27, which were comparable to those in sub‐confluent growth conditions. There was a steady increase in Cyc D1/D2 levels at 3, 6, 12 and 24 h consistent with previously published results.
Using Quantitative Reverse Transcriptase‐Polymerase Chain Reaction (QRT‐PCR) analysis, we next examined whether RET2A‐mediated induction of N‐Myc protein was due to an increase in N‐Myc mRNA expression. Indeed, there is a two‐ and three‐fold increase in N‐Myc mRNA levels at 6 and 12 h respectively (Figure 1E). At 24 h N‐Myc mRNA and protein levels return to basal levels. We also observed that the three‐fold increase in p27 and p18 mRNA levels at 24 h is prevented by RET2A. These data establish a sequential timeline of events leading to regulation of p18 and p27 in response to RET2A expression. Within 3 h of RET2A expression, there is activation of the MAPK pathway that results in induction of N‐Myc at 6 h, lasting through 12 h and dropping back to basal levels by 24 h. Low levels of p18 and p27, comparable to those in sub‐confluent cells, persist as cells continue into post‐confluency despite the later absence of N‐Myc expression. It is possible that N‐Myc acts as a “molecular switch” that prevents inr‐mediated p18 transcription in sub‐confluent cells which would otherwise allow p18 accumulation and growth arrest due to confluency. Once this point is past, inr‐mediated p18 transcription cannot occur under post‐confluent conditions, and the requirement for N‐Myc expression lessens.
We have previously shown that, RET2A‐mediated maintenance of low p18S transcription in post‐confluent conditions requires a functional MAPK pathway therefore; we tested if the transient induction of N‐Myc is MAPK dependent. We repeated the short timecourse in the presence of a chemical inhibitor of the MAPK pathway, PD98059. Cells treated with PD98059 for 1 h to inhibit MAPK signaling were then treated with Tet to express RET2A. PD98059 efficiently inhibited phosphorylation of ERK throughout the course of the experiment (Figure 2A). N‐Myc induction, previously observed at 6 and 12 h, was not seen (Figure 2A), suggesting that N‐Myc induction is MAPK dependent. Both p18 and p27 accumulate at 24 h in PD98059‐treated cells. These results show that activation of the MAPK pathway by RET2A is required for induction of N‐Myc expression and maintenance of low CDKI levels.
Figure 2.
N‐Myc induction by RET2A is ERK activation dependent and is required for maintenance of low p18 and p27, but not for D‐type cyclin induction. (A) RET2A‐mediated induction of N‐Myc protein is MAPK dependent. RET2A cells grown to ∼90% confluency were treated with PD98059 and/or tetracycline (+Tet) and indicated. Protein lysates isolated at indicated timepoints were used for immunoblotting with antisera against indicated proteins. (B) Induction of N‐Myc is required for RET2A to maintain low p18 and p27, but not to induce D‐type cyclins. RET2A cells transfected with negative control or N‐Myc siRNA were treated with PD98059 or Tet. Western immunoblotting was performed for indicated proteins. (C) Quantitation of western analysis from panel B. (D) Quantitation of mRNA levels from short timecourse. Cells were treated like described in panel B. Relative mRNA levels of indicated genes were determined by QRT‐PCR. ∗ indicates p < 0.05. Results are representative of three independent experiments. All quantitations are average of three independent experiments. Error bars indicate standard deviation.
2.3. N‐Myc induction is required for RET2A‐mediated p18 regulation
We next assessed whether N‐Myc induction is required to maintain low p18 and p27 levels as cells become growth arrested. We used siRNA directed against N‐Myc to inhibit its expression. In negative control (non‐specific siRNA) cells treated with Tet, we detected an approximately six‐fold increase in N‐Myc expression at 12 h. We also detected a four‐fold induction of D‐type cyclins (at 12 and 48 h) and four‐ and 2.5‐fold decrease in levels of p18 and p27 respectively (at 48 h, Figure 2B, quantitated in C). On the other hand, in N‐Myc siRNA transfected cells, expression of N‐Myc was abolished, but activation of the MAPK pathway was still seen at 12 and 48 h. Most importantly, p18 and p27 were no longer maintained at low levels at 48 h. In other words, loss of N‐Myc induction at 12 h prevents RET2A‐mediated maintenance of low p18 and p27 levels under growth arrest conditions at 48 h. A third set of samples treated with PD98059 were included in this analysis, because such treatment inhibits MAPK activation and N‐Myc induction. Similar results were seen in PD98059‐treated samples, which indicates that N‐Myc is downstream of the RET2A‐MAPK signaling pathway and that MAPK dependent N‐Myc induction is essential for RET2A‐mediated prevention in accumulation of p18 and p27. Consistent with our previous report where MAPK activity is not involved in RET2A‐mediated induction of Cyclin D1/D2, their levels remained unaffected in all of the +Tet samples, irrespective of the N‐Myc status.
We also confirmed that protein levels correlate with mRNA expression; total RNA samples from parallel experiments were analyzed by QRT‐PCR for indicated genes. Consistent with protein expression results there was a three‐fold increase in N‐Myc mRNA levels in +Tet and control siRNA samples at 12 h, as compared to −Tet cells (Figure 2D) which was not observed in N‐Myc siRNA or PD98059‐treated cells. Although p18 and p27 mRNAs were unaffected at 12 h, the three‐fold increase at 48 h in p18 and p27 mRNA levels in −Tet samples was absent in +Tet or control siRNA samples, suggesting that they were maintained from sub‐confluent growth conditions. At 48 h, p18 and p27 mRNA levels were reciprocally higher in cells that lack N‐Myc (N‐Myc siRNA and PD98059‐treated cells). Additionally, RET2A‐mediated induction of Cyclin D1/D2 mRNA remained unaffected in the absence of N‐Myc (data not shown). Together, these results suggest that RET2A‐mediated MAPK pathway activation and N‐Myc induction are required to maintain low levels of p18 and p27, but not for the induction of Cyclin D1/D2.
2.4. Promoter sequence containing the N‐Myc consensus binding site is required for RET2A‐mediated effects on p18 transcription
We first determined if the N‐Myc binding site (inr element) resides within the minimal RET2A responsive region of the p18 promoter. We subcloned various fragments of the p18 promoter into the pGL2‐basic firefly luciferase reporter gene vector. In response to RET2A expression, measured luciferase activity of a full‐length (−1 to −3496) and that of a 5′ truncated (−1 to −992, LucA) construct was found to be identical (data not shown), suggesting that there are no RET2A responsive elements in the −992 to −3496 region of the p18 promoter. Therefore, this region was excluded from subsequent promoter analyses. We next determined if the inr element is required for RETA‐mediated prevention of p18 accumulation. LucA and LucB contain 992 bp (−1 to −992) and 365 bp (−1 to −365) immediately 5′ to the p18 translation start site (designated as +1, ATG) respectively, while LucC is 499 bp (−367 to −868) and lacks 367 bp immediately 5′ to the p18 translation start site (i.e. lacks LucB sequence) (Figure 3A). LucA and LucB contain the N‐Myc binding site (−156 to −167, inr element), whereas LucC does not. RET2A cells transfected with LucA, B or C, and negative control or N‐Myc siRNA at 60–80% confluency became ∼90% confluent (0 h timepoint) at 24 h post‐transfection when they were treated with Tet and harvested for protein lysates. The luciferase activities driven by the LucA, B and C fragments at 0 h were ∼3000, ∼4500 and ∼12,500 Relative Luciferase Units (RLU), respectively (Figure 3B). At this timepoint, as compared to −Tet samples, neither Tet treatment nor transfection of control or N‐Myc siRNA had any effect on luciferase activity of any of the three constructs. A parallel set of treated cells were harvested after 48 h of Tet treatment (36 h post‐confluent). At this post‐confluent timepoint, p18 transcription is maintained at low levels by RET2A therefore, we predict lower luciferase activity in +Tet samples. The overall luciferase activity in –Tet cells decreased for all three constructs. Regardless, expression of RET2A leads to a further, statistically significant, three‐fold decrease in luciferase activity of LucA and LucB. This suggests that LucA and LucB contain RET2A responsive transcriptional regulatory elements that could cause lower transcription of p18S. Similar results were obtained in control siRNA transfected RET2A‐expressing cells however, loss of N‐Myc by siRNA leads to restoration of luciferase activity driven by both, LucA and LucB (Figure 3B). This indicates that N‐Myc is required for RET2A‐mediated repression of luciferase activity. The activity of LucC remained unaffected at an average of ∼1900 RLU under all four conditions (−Tet, +Tet, +Tet + control siRNA or +Tet + N‐Myc siRNA). These data suggest that the −1 to −365 region of the p18 locus, containing the inr N‐Myc binding site, is required for RET2A‐mediated repression of luciferase reporter gene activity and for the restoration of luciferase activity in response to N‐Myc siRNA treatment.
Figure 3.
p18 promoter sequences containing the N‐Myc binding site, and N‐Myc expression is required for RET2A‐mediated repression of luciferase activity. (A) Representation of different pGL2‐Luciferase reporter constructs used for luciferase assay. The sequence relative to the +1 ATG start codon of p18 coding is shown. Exons 1 and 2 are shown as black boxes. (B) RET2A cells were transfected with siRNA or luciferase reporter constructs as indicated. Renilla and firefly luciferase activities from cell extracts taken at indicated times were used to determine Relative Luciferase Units (RLU). ∗ indicates p < 0.05. Results are average of three independent experiments. Error bars indicate standard deviation.
Several other TFs known to be regulated by MAPK, and whose binding sites were present within the −1 to −365 region of the p18 promoter, were unaffected at the mRNA level by RET2A. One exception was the TF c‐Fos, which was induced two‐ and three‐fold at the mRNA level at 3 and 6 h respectively (see Supplementary Figure 1). The timing of c‐Fos induction at 3 and 6 h precedes that of N‐Myc which occurs at 6 and 12 h. Therefore, it is possible that c‐Fos may act as an intermediate TF that induces N‐Myc transcription in response to RET2A expression and MAPK activation. While we have not pursued involvement of c‐Fos in this study, and cannot completely eliminate involvement of other TFs whose putative binding sites are lost due to the DNA sequences deleted in the LucC construct, the requirement of N‐Myc expression for RET2A‐mediated effects on luciferase activity (and p18 transcription, Figure 2B–D) led us to conclude that the inr element within this 365 basepair region appears to be critical for RET2A and N‐Myc dependent transcriptional regulation of p18.
2.5. RET2A expression leads to increased binding of N‐Myc to its target inr site within the p18S promoter
Next, we sought a more direct approach to determine direct binding of N‐Myc to the inr element in the p18 promoter. We used a combination of Chromatin Immuno‐Precipitation (ChIP) followed by quantitative‐PCR (qPCR) analysis of precipitated DNA fragments to determine N‐Myc binding to its consensus inr binding site within the p18 promoter in a RET2A‐dependent manner. RET2A cells transfected with negative control or N‐Myc siRNA or treated with PD98059 were induced with Tet. Nuclear lysates were harvested at 12 h post‐induction, when N‐Myc expression is maximal. Crosslinked and fragmented chromatin was immunoprecipitated with an N‐Myc antibody and the DNA bound to N‐Myc was then used as a template in qPCR reactions to amplify non‐specific or N‐Myc binding site specific amplicons (Figure 4A).
Figure 4.
N‐Myc induction results in elevated binding of N‐Myc to its consensus binding site in the p18 promoter. (A) Primers used for ChIP analysis. Froward and reverse primers, positioned in relation to the p18 start codon (ATG, +1) and designed to amplify an N‐Myc binding site specific amplicon and a non‐specific control amplicon are shown by arrows. The N‐Myc consensus binding site is shown in intron1. (B) ChIP assay showing binding of N‐Myc to p18 promoter sequence containing N‐Myc binding site. Crosslinked chromatin was used from RET2A cells grown to ∼60% confluency, transfected with control or N‐Myc siRNA, and treated with PD98059 or Tet, as indicated. Chromatin immunoprecipitation was performed with N‐Myc Antisera followed by qPCR analysis using primers specific for either the N‐Myc binding site containing amplicon or the non‐specific amplicon. Values from the N‐Myc amplicon normalized to the non‐specific amplicon were used to calculate fold induction in N‐Myc bound fragments. All quantitations are average of three independent experiments. Error bars indicate standard deviation. ∗ indicates p < 0.05. (C) MAPK dependent expression of N‐Myc in the ChIP samples was confirmed by using 1/10th volume of the nuclear extracts for Western blot analysis. Antisera against indicated proteins were used to determine protein levels. Results are representative of three independent experiments.
Expression of RET2A (+Tet) results in a six‐ to seven‐fold enrichment in the N‐Myc consensus binding site specific amplicon relative to the non‐specific amplicon suggesting that there is increased binding of N‐Myc to inr element (Figure 4B). In the presence of N‐Myc siRNA (or PD98059 MAPK inhibitor), we did not detect any discernible N‐Myc expression (Figure 4C, consistent with Figure 2A and B). Hence, it is not surprising that we do not detect any binding of N‐Myc to the N‐Myc binding site under such conditions. The enrichment remained unaffected in negative control siRNA samples. N‐Myc protein expression and phosphorylation of ERK was confirmed by western analysis using 10% of pre‐cleared nuclear extracts (Figure 4C). ERK phosphorylation was inhibited efficiently in PD98059‐treated cells and N‐Myc was induced only in +Tet and + Tet + control siRNA samples. These results demonstrate that N‐Myc binds to its genomic target site within the p18 promoter in response to RET2A expression. Taken together, the luciferase and ChIP assays suggest that the N‐Myc inr binding site within the p18S promoter is critical for RET2A‐mediated regulation of p18 expression and that N‐Myc binds to its consensus binding site in response to RET2A expression in a MAPK‐dependent manner
2.6. N‐Myc induction is required for the RET2A‐mediated increase in proliferation
Continued RET2A expression starting from sub‐confluent through post‐confluent growth arrest conditions confers a proliferative advantage. In other words, RET2A‐expressing cells continue to proliferate instead of arresting under post‐confluent growth conditions. Cells seeded at 60% confluency and RET2A expression induced at 90% confluency (0 h timepoint) were fixed, propidium iodide (PI) stained and analyzed for DNA content at 48 h (36 h post‐confluent). The percentage of cells in various phases of the cell cycle (G0/G1, S and G2/M) was determined. Uninduced 90% sub‐confluent cells (−Tet 0 h) are cycling, as indicated by a proliferative index (S + G2/M phases) of 28.1 + 2.4% (Figure 5A). Cells treated with Tet at 0 h exhibit a similar proliferative index of 28.6 + 2.3%. This is expected since RET2A has no effect on proliferation under sub‐confluent growth conditions (Joshi et al., 2007). As uninduced cells become post‐confluent (−Tet 48 h) they become growth arrested due to contact inhibition, reflected by a reduced proliferative index of 6.11 + 1.52%. RET2A expression (+Tet 48 h) allows post‐confluent cells to continue proliferation (S + G2 M = 17.8 + 4.2%) at a three‐fold higher rate relative to −Tet samples (48 h). Therefore, ethanol‐treated cells growth arrest due to contact inhibition by 48 h while, RET2A‐expressing cells continue to proliferate at a higher rate.
Figure 5.
N‐Myc is required for RET2A‐mediated hyperproliferation. (A) RET2A expression correlates with an increased proliferative index under growth arrest conditions. Cells grown to 80% confluency and treated with Tet were harvested at indicated timepoints. Fixed and PI stained cells were sorted by FACS into G0/G1, S and G2/M fractions. The S + G2/M fraction (proliferative index) of samples is shown. (B) Loss of N‐Myc expression negates RET2A‐mediated hyperproliferation. RET2A cells grown to 60% confluency, transfected with no DNA (Mock), control or N‐Myc siRNA and treated with Tet were harvested at indicated times. Fixed and PI stained cells were sorted by FACS into G0/G1, S and G2/M fractions. Relative proliferative index for each sample, calculated by taking the ratio of the proliferative indices of +Tet/−Tet is shown. ∗ indicates p < 0.05. Results are representative of three independent experiments. Error bars indicate standard deviation.
Since RET2A‐dependent induction of N‐Myc is required to maintain low levels of p18, and low levels of p18 are critical for RET2A‐mediated hyperproliferation, it is possible that N‐Myc is required for RET2A‐mediated hyperproliferation. Using FACS analysis we assessed whether RET2A‐expressing cells could hyperproliferate under growth arrest conditions in the absence of N‐Myc. RET2A cells transfected at 60–80% confluency with no DNA (Mock), control siRNA or N‐Myc siRNA were induced 24 h post‐transfection when cells reached 90% confluency (0 h timepoint) with Tet or ethanol for 48 h (approximately 24 h post‐confluent). RET2A‐expressing (+Tet), mock or control siRNA transfected post‐confluent cells continue to proliferate at an approximately two‐fold higher rate, as compared to ethanol‐treated cells (Figure 5B). However, cells lacking N‐Myc fail to gain a growth advantage, reflected by no change in the proliferative index relative to the ethanol‐treated N‐Myc siRNA sample. Together these data suggest that the proliferative advantage conferred by RET2A expression requires expression of N‐Myc.
3. Discussion
RET expression is required primarily for kidney and neuronal development. In adults, activating RET mutations, such as RET2A, cause oncogenic lesions affecting mainly the endocrine organs. While various studies have uncovered details regarding normal RET activation and numerous signaling pathways that it activates [Reviewed in (Kodama et al., 2005)], we still lack detailed knowledge of which pathway is critical for RET2A‐mediated oncogenesis. Our previous study demonstrated that constitutive RET2A signaling leads to a simultaneous decrease in p18 and p27 and a concomitant increase in Cyclin D1/D2 (Joshi et al., 2007). We also showed that maintenance of low p18 mRNA levels at the transition from “proliferating cells” to “confluency mediated growth arrested cells” is a rate limiting step for RET2A‐mediated hyperproliferation. Our present study seeks to elucidate details of transcriptional regulation of p18 at this transition, to uncover the pathway that starts with RET signaling and leads to p18‐mediated hyperproliferative advantage imparted to cells. We show that RET2A expression leads to ERK phosphorylation/activation, required for induction of N‐Myc mRNA and protein expression (1, 2A). Induction in N‐Myc precedes the decrease in p18 and p27 levels (1, 2A). We also demonstrate that the MAPK dependent increase in N‐Myc is required for RET2A‐mediated maintenance of low p18 and p27 levels, but not for induction of Cyclin D1/D2 (Figure 2B–D) under growth arrest conditions. N‐Myc expression and a 365 bp sequence of the p18S promoter containing the N‐Myc inr binding site both are required for RET2A‐mediated transcriptional regulation of p18S (Figure 3). We also show that N‐Myc binds to this inr site in response to RET2A expression (Figure 4). Finally, cells that express RET2A but lack N‐Myc cannot gain the proliferative advantage conferred by RET2A (Figure 5). These results support a critical role for N‐Myc as a mediator of RET2A signals to transcriptionally regulate p18 expression, and perhaps help explain a mechanism by which the RET2A mutation predisposes MEN2A tumorigenesis.
A striking feature of our study is that the timing of N‐Myc induction precedes the effect on p18 mRNA and protein levels. More specifically, the induction of N‐Myc begins as cells are transitioning between sub‐confluent proliferation and post‐confluent growth arrest conditions. At this transition, RET2A expression induces N‐Myc as an immediate early gene perhaps to prevent formation of TF complexes that would induce transcription of p18S mRNA and p18 protein. Even though N‐Myc expression is transient, formation of an activating complex required for p18 induction could be repressed at this transition. This notion is supported by our previous work (Joshi et al., 2007), that the growth advantage imparted by RET2A under arrest conditions requires maintenance of low p18 levels, similar to those seen in cycling sub‐confluent cells. Also, RET2A‐mediated p18 repression is independent of the mechanism of growth arrest, i.e. contact inhibition or serum starvation, thus eliminating the possibility that our findings are experimental artifacts. Moreover, there have been in vivo observations that show the timing of maximal N‐Myc expression during murine embryogenesis at 9.5 dpc (Charron et al., 1992), while embryonic lethality caused by genetic ablation of N‐Myc occurs between 10.5 and 12.5 dpc (Sawai et al., 1991). This suggests that N‐Myc function and cellular response as a consequence of its loss, particularly proliferation, can be temporally separated. We propose that elevated N‐Myc levels prevent the setting in motion of a timed “program” involving p18 induction as a central event in confluency mediated growth arrest.
Mechanistically, competitive binding of N‐Myc causes displacement of the p300 co‐activator from a Miz1:p300 complex bound to an inr element (Peukert et al., 1997; Schneider et al., 1997), leading to Miz1 sequestration. This prevents transcription of target genes which would otherwise be activated by the Miz1:p300 complex. The induction of p300, although not studied in this work, may be transient and occur at the transition of active cycling cells and initiation of growth arrest. Once the opportunity for p300 mediated induction of p18 is missed in RET2A‐expressing cells due to the presence of N‐Myc, p18 may fail to accumulate as cells continue to grow in spite of becoming confluent. Moreover, presence of the inr sequence in the p18S promoter region suggests that only p18S transcription would be affected by N‐Myc. The p18L transcript initiates more than 1.5 kb upstream of the inr sequence, and would not be regulated by this mechanism. This is consistent with our previous report where only p18S is regulated by RET2A signals, not p18L (Joshi et al., 2007).
Cyclin D1 overexpression has been shown to lead to transformation in a variety of cell types (Bianchi et al., 1993; Hunter and Pines, 1991; Loves et al., 1994). In RET2A‐expressing cells, while cyclin D1 induction alone is not sufficient to increase proliferation under growth arrest conditions, concomitant lower p18 levels are required for RET2A‐mediated hyperproliferation (Joshi et al., 2007). This implies that N‐Myc cooperates with an increase in Cyclin D1 to promote proliferation. Lovec et al. (1994) show that E‐Mu driven overexpression of Cyclin D1 cooperates with N‐Myc to induce lymphomagenesis. Perhaps, N‐Myc prevents p18 accumulation in the background of high Cyclin D1 levels leading to increased lymphomagenesis in their study. Aubry et al. have shown that N‐Myc and c‐Myc share cellular functions such as S phase entry in quiescent cells and induction of apoptosis in serum deprived cells (Aubry and Charron, 2000); however, deletion of either N‐Myc or c‐Myc produces different phenotypes (Charron et al., 1992; Davis et al., 1993). This suggests that the differential roles of these proteins are due to mutually exclusive expression patterns. Additionally, N‐Myc expression can induce entry into S phase from quiescence even in the absence of external stimuli (Aubry and Charron, 2000). Therefore, it is possible that constitutive signaling by RET2A in thyroid C cells and/or adrenal chromaffin cells leads to induction of N‐Myc, which in turn maintains low p18 and p27 levels and allows for inappropriate entry into S phase.
Based on our results, we propose that ligand‐induced activation of wildtype RET during development leads to MAPK‐mediated induction of N‐Myc, which prevents accumulation of p18 and p27 (Figure 6A). The sustained low levels of p18 and p27 allow for proliferation of neuronal progenitor cells during development (Knoepfler et al., 2002). N‐Myc null mice die in utero due to lack of neuronal proliferation, caused by growth arrest in neuronal progenitors. This arrest is correlate with an accumulation of p18 and p27, supporting a critical role for N‐Myc in maintaining low p18 and p27 levels in vivo (Zindy et al., 2006). The observation that mice deleted for both p18 and p27 exhibit no neuronal phenotype suggests that proliferation of neuronal progenitors is unaffected by loss of p18 and p27 (Franklin et al., 1998, 2000). Prior to differentiation and neurogenesis, p18 and p27 would be maintained at low levels to allow efficient proliferation of neuronal progenitors. As progenitors initiate differentiation, they undergo growth arrest mediated by accumulation of another CDKI such as p19Ink4c (Zindy et al., 1999). Such a role for p19 is supported by a study which shows that loss of both p19 and p27 leads to postnatal continued proliferation of some neuronal precursors. Therefore, expression of RET2A during development may inappropriately prevent p18 and p27 accumulation. The low levels of p18 and p27 are inconsequential for neurogenesis, since neuronal progenitor cells growth arrest via p19 accumulation. However, expression of RET2A in normally arrested adult adrenal chromaffin cells and/or thyroid C cells could lead to an inappropriate increase in N‐Myc, thereby preventing accumulation of p18 and p27 in these cells (Figure 6B). Deregulated p18 and p27 expression in turn could cause hyperproliferation. This makes N‐Myc a key regulator of cell proliferation in RET2A‐mediated oncogenesis. A critical role for N‐Myc in MEN2A tumorigenesis is particularly exciting in light of a recent study that shows systemic reversible inhibition of c‐Myc in a mouse lung cancer model that provides validity for c‐Myc as a useful target for regression of aggressive lung cancer (Soucek et al., 2008). The side effects of systemic c‐Myc inhibition were shown to be readily and rapidly reversible. Therefore, targeting N‐Myc in MEN2A patients may prove to be an effective therapeutic strategy.
Figure 6.
Proposed model for RET2A‐mediated deregulation of p18. (A) Activation of wildtype RET by its ligands or expression of constitutively active RET2A during development in Neuronal Progenitor Cells (NPCs) leads to induction of N‐Myc which in turn prevents accumulation of p18 and p27. Sustained low levels of p18 and p27 lead to proliferation of NPCs which then are growth arrested prior to differentiation in a p19 dependent manner. (B) Expression of the ligand independent constitutively active mutant RET2A in adrenal chromaffin cells and thyroid C cells leads to inappropriate expression of N‐Myc, which maintains decreased p18 and p27 mRNA and protein levels. Low p18 and p27 levels result in an increase in proliferation and tumorigenesis in RET2A‐expressing cells.
4. Materials and methods
4.1. Plasmids and cell culture
LucA (−1 to −992) and LucB (−1 to −365) fragments were amplified by PCR from wild type genomic mouse tail DNA using LucAF (5′‐ATGGTACCAAGTTACTGGTGAGCTGGCCGGGTGCA‐3′) and LucBF (5′‐ACGGTACCACGTGACAGCTCTGCCTACA‐3′), respectively as forward primers with a common reverse primer LucR (5′‐CGCTCGAGTCTTTAGGGTCCTGGCGATCGG‐3′). The PCR conditions used were 95 °C/5 min–one cycle; 95 °C/1 min, 60 °C/1 min, 72 °C/2 min–40 cycles; 72 °C/10 min–one cycle, 4 °C hold. KpnI and XhoI linkers were added to the forward and reverse primers respectively. LucA and LucB PCR products digested with KpnI/XhoI were ligated with KpnI/XhoI digested pGL2‐Basic firefly luciferase reporter gene vector. The pGL2‐Basic and pGL2‐LucC (designated as pGL2‐Luc17, kindly provided by Dr. Yue Xiong (Phelps et al., 1998). The N‐Myc siRNA was purchased (Santa Cruz Biotechnology, Santa Cruz, CA, USA) and used according to manufacturer's recommendations. The control siRNA was kindly provided by Dr. Elizabeth Taparowsky.
NIH3T3 fibroblasts harboring both pcDNA6‐TR and pcDNA4‐RET2A have been described previously (hereafter referred to as RET2A cells) (Joshi et al., 2007). Cells were grown in Dulbecco's modified Eagle's medium (DMEM) (Invitrogen, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (FBS) (Mediatech, Inc., Herndon, VA, USA) and 500 U/ml penicillin ‐ 500 μg/ml streptomycin (Invitrogen, Carlsbad, CA, USA) and incubated at 37 °C and 5% CO2. Transfection was carried out using Polyfect transfection reagent according to the manufacturer's protocol (Qiagen, Valencia, CA, USA). Expression of RET2A was induced by addition of 2 μg/ml tetracycline (Sigma, St. Louis, MO, USA) (+Tet). As a negative vehicle control, ethanol was added (−Tet) for all experiments. Ethanol or tetracycline was boosted every 12 h. The MEK inhibitor PD98059 (100 μM, final concentration) (Calbiochem, San Diego, CA, USA) was added to culture medium 1 h prior to addition of tetracycline and continued for the course of the experiment.
4.2. Antibodies and immunoblotting
For immunoblotting, the following commercially available antibodies were purchased: rabbit anti‐ERK‐MAPK (C‐16), rabbit anti‐phospho‐ERK‐MAPK antisera (Santa Cruz Biotechnology, Santa Cruz, CA, USA), rabbit anti‐N‐Myc (M‐50) (Santa Cruz Biotechnology, Santa Cruz, CA, USA), mouse anti‐tubulin (Ab‐4, NeoMarkers, Fremont, CA, USA). Polyclonal rabbit anti‐RET (03‐20), rabbit anti‐p27 (03‐22) (Joshi et al., 2007), rabbit anti‐cyclin D1 and rabbit anti‐p18 (Franklin and Xiong, 1996; Phelps and Xiong, 1998) polyclonal antibodies have been described previously. All procedures for immunoblotting have been described previously (Franklin et al., 1998, 2000, 1996, 1995, 1998). Total quantity of proteins in lysates was determined using Bradford assay. Individual bands on the immunoblots were quantified using Licor Odyssey v1.2. and normalized to tubulin (with the exception of Figure 4C). In Figure 4C, total ERK was used as a loading control. Fold change was calculated as the ratio of induced (+Tet) to uninduced (−Tet) values. The average relative fold change in proteins at the 0 h timepoint was set to +1 for proteins that are induced by RET2A or to −1 for proteins that are repressed by RET2A. P‐values were obtained by performing Student's t‐test.
4.3. QRT‐PCR
Total RNA isolation, synthesis of cDNA, SYBR green based QRT‐PCR and data analysis has been previously described (Joshi et al., 2007). Each QRT‐PCR reaction was carried out in the ABI prism 7300 sequence detection system (Applied Biosystems, Foster City, CA, USA) Fold induction or repression of target gene transcripts relative to 18S rRNA were obtained and subjected to statistical analysis using Student's t‐test. The Primer Express software provided by Applied Biosystems was used to design primers for each target gene. Sequences for each primer are as follows:
18S rRNA forward primer (5′‐CACGGCCGGTACAGTGAAA‐3′),18S rRNA reverse primer (5′‐AGAGGAGCGAGCGACCAA‐3′);
p18S CDKI forward primer (5′‐ACCATCCCAGTCCTTCTGTCA‐3′),p18S CDKI reverse primer (5′‐GTTGCTTCACTTTTTCCCCTTTC‐3′);
p27 CDKI forward primer (5′‐CTTCCGCCTGCAGAAATCTCT‐3′),p27 CDKI reverse primer (5′‐CGGCAGTGCTTCTCCAAGTC‐3′);
N‐Myc forward primer (5′‐CTGAGCTGGTGAAGAACGAGAA‐3′),N‐Myc reverse primer (5′‐CTCGGTGGCCTTTTTCAAGA‐3′);
4.4. Luciferase assay
RET2A cells seeded in 12‐well plates were grown to 60–80% confluency at the time of transfection. Polyfect (Qiagen, Valencia, CA, USA) transfection reagent was used according to manufacturer's protocol for transfection. For each transfection, 1 μg of the pGL2‐LucA, B or C plasmid along with 0.1 μg of pRL‐null plasmid was used. 1 μg of control or N‐Myc siRNA plasmid was included in the transfection where indicated. After 23 h transfection, PD98059 (Calbiochem, San Diego, CA, USA) was added to a final concentration of 100 μM. At 24 h post‐transfection, tetracycline (2 μg/ml) or 100% ethanol (vehicle) were added. The 0 h samples were harvested at this timepoint. The 48 h samples were further incubated by boosting RET2A expression with tetracycline at 12 h intervals. Cells were washed twice with cold PBS and lysed with 100 μl of passive lysis buffer (Promega, Madison, WI, USA). 10 μl of cell extract was used to determine Firefly and Renilla luciferase activity by using firefly luciferase substrate (Promega, Madison, WI, USA) and Coelenterazine (Sigma, St. Louis, MO, USA) respectively, according to the manufacturer's protocol in the lumat LB9501 luminometer (Berthold Technologies, Oak Ridge, TN, USA). The firefly luciferase activity was normalized to Renilla luciferase activity and represented as relative luciferase units (RLU).
4.5. Chromatin Immunoprecipitation (ChIP) assay
RET2A cells seeded in 100 cm plates were grown to 60–80% confluency for transfections using the Polyfect (Qiagen, Valencia, CA, USA) transfection reagent according to manufacturer's protocol. For each transfection, 10 μg of the control or N‐Myc siRNA plasmid was used. One hour prior to tetracycline addition, PD98059 (Calbiochem, San Diego, CA, USA) was added to a final concentration of 100 μM where indicated. At 24 h post‐transfection, tetracycline (2 μg/ml) or 100% ethanol (vehicle) were added. 12 h after treatment with Tet (or ethanol) cells were treated with 0.25% formaldehyde (Sigma, St. Louis, MO, USA) followed by 1 M glycine (Sigma, St. Louis, MO, USA). Cells were then washed with PBS + protease inhibitor cocktail (100 mM aprotinin, 100 mM Laupeptin, 100 mM trypsin inhibitor and 100 mM benzamidine). Cytoplasmic lysis was accomplished by using the ChIP lysis buffer (10 mM Hepes pH 7.9, 10 mM KCl, 1.5 mM MgCl2, 0.01% 2‐mercaptoethanol, 0.1% NP‐40, 1 mM protease inhibitor cocktail) followed by nuclear lysis using the ChIP dilution buffer (0.1% sodium dodecoyl sulfate, 1% triton X‐100, 16 mM Tris–HCl pH 8.1, 165 mM sodium chloride). Chromatin was fragmented by sonicating three times for 15 s pulses at an interval of 5 min. Fragmented chromatin was then pre‐cleared using protein A sepharose beads (Pierce Biotechnologies, Rockford, IL, USA) with rocking for 1 h. Cleared lysates were then used for overnight immunoprecipitation with 10 μg of an N‐Myc antibody (M − 50) (Santa Cruz Biotechnology, Santa Cruz, CA, USA). Precipitates were reverse‐cross‐linked (250 mM NaCl) and treated with Proteinase K (Promega, Madison, WI, USA). DNA was purified using a PCR purification kit (Qiagen, Valencia, CA, USA) according to manufacturer's protocol. Equal amounts of DNA and SYBR Green PCR Master Mix (Applied Biosystems, Foster City, CA, USA) were used in each reaction of qPCR in the ABI prism 7300 sequence detection system (Applied Biosystems, CA, USA). The qPCR parameters were: 50 °C/2 min–one cycle; 95 °C/10 min–one cycle and 95 °C/15 s, 60 °C/1 min–40 cycles. Data were analyzed, according to the manufacturer's recommendations to obtain fold induction of N‐Myc amplicon relative to a non‐specific amplicon, and subjected to statistical analysis using Student's t‐test. Primers specific for the N‐Myc amplicon, p18ChIP‐NMyc forward primer (5′‐TGGGCTCACTTTTGCTGAATAA‐3′) and p18ChIP‐NMyc reverse primer (5′‐ GTCGGCCTGCCAAAAGC‐3′) or non‐specific amplicon, p18ChIP‐control forward primer (5′‐ CCCGGCGGTTTTGGTT‐3′) and p18ChIP‐control reverse primer (5′‐ CAGGACAGCCAGGGCTATACA‐3′) were used (Figure 4A).
4.6. Fluorescence activated cell sorting (FACS) analysis
Cells were harvested and fixed as described previously (Juan and Darzynkiewicz, 2001). Fixed cells were washed with phosphate buffered saline and treated with 100 mg/ml RNAse for 5 min and stained with 50 mg/ml propidium iodide for 30 min in dark. 675 nm emission from the DNA bound propidium iodide was measured with a Coulter FC‐500 flow cytometer. Approximately 10,000 cells from each sample were analyzed. The computer program ModFit was used to determine the percentages of cells within the G1, S and G2/M phases of the cell cycle. The proliferative index was calculated by obtaining the sum of all S and G2/M‐phase populations in any given sample. Then, relative proliferative index was calculated by taking the ratio of the induced (+Tet) to uninduced (−Tet) samples for a given sample. Student's t‐test was performed to obtain P‐values.
Supporting information
The following are the Supplementary data related to this article:
Supplementary data
Acknowledgements
This work was supported by the American Cancer Society (RSG‐03‐236‐01‐MGO), Purdue University Department of Biological Sciences and the Purdue University Cancer Center. We thank Cheryl Holdman, Kathy Ragheb, Dr. Stephen Konieczny and Dr. Elizabeth Taparowsky for technical assistance and Dr. Michael Hodsdon for critical review of this manuscript.
Supplementary material 1.
Supplementary data related to this article can be found online at doi:10.1016/j.molonc.2010.10.004.
Kulkarni Mandar V., Franklin David S., (2011), N‐Myc is a downstream target of RET signaling and is required for transcriptional regulation of p18Ink4c by the transforming mutant RETC634R , Molecular Oncology, 5, doi: 10.1016/j.molonc.2010.10.004.
Contributor Information
Mandar V. Kulkarni, Email: mandar.kulkarni@yale.edu
David S. Franklin, Email: franklin@tulane.edu
References
- Acton, A. , Hoppener, J.W.M. , Lips, C.J.M. , 2004. Involvement of the cell cycle regulator P18/INK4C/CDKN2C in human medullary thyroid carcinoma development. J. Intern. Med. 255, 696–730. [Google Scholar]
- Aubry, S. , Charron, J. , 2000. N-Myc shares cellular functions wiht c-Myc. DNA Cell Biol. 19, 353–364. [DOI] [PubMed] [Google Scholar]
- Bianchi, A.B. , 1993. Overexpression of cyclin D1 in mouse skin carcinogenesis. Oncogene. 8, 1127–1133. [PubMed] [Google Scholar]
- Blackwell, T.K. , 1993. Binding of myc proteins to canonical and noncanonical DNA sequences. Mol. Cell Biol. 13, 5216–5224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blais, A. , 2002. Regulation of the human cyclin-dependent kinase inhibitor p18INK4c by the transcription factors E2F1 and Sp1. J. Biol. Chem. 277, 31679–31693. [DOI] [PubMed] [Google Scholar]
- Charron, J. , 1992. Embryonic lethality in mice homozygous for a targeted disruption of the N-myc gene. Genes Dev. 6, 2248–2257. [DOI] [PubMed] [Google Scholar]
- Davis, A.C. , 1993. A null c-myc mutation causes lethality before 10.5 days of gestation in homozygotes and reduced fertility in heterozygous female mice. Genes Dev. 7, 671–682. [DOI] [PubMed] [Google Scholar]
- Downs, K.M. , 1989. Contrasting patterns of myc and N-myc expression during gastrulation of the mouse embryo. Genes Dev. 3, 860–869. [DOI] [PubMed] [Google Scholar]
- Eberhardy, S.R. , 2000. Direct examination of histone acetylation on Myc target genes using chromatin immunoprecipitation. J. Biol. Chem. 275, 33798–33805. [DOI] [PubMed] [Google Scholar]
- Franklin, D. , 1998. CDK inhibitors p18INK4c and p27KIP1 mediate two separate pathways to collaboratively suppress pituitary tumorigenesis. Genes Dev. 12, 2899–2911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Franklin, D. , 2000. Functional collaboration between different cyclin-dependent kinase inhibitors Suppresses tumor growth with distinct tissue specificity. Mol. Cell. Biol. 20, 6147–6158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Franklin, D. , Xiong, Y. , 1996. Induction of p18INK4c and its predominant association with CDK4 and CDK6 during myogenic differentiation. Mol. Biol. Cell. 7, 1587–1599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hunter, T. , Pines, J. , 1991. Cyclins and cancer. Cell. 66, 1071–1074. [DOI] [PubMed] [Google Scholar]
- Jenkins, C. , Xiong, Y. , 1995. Immunoprecipitation and immunoblotting in cell cycle studies. In Pagano M., Cell Cycle: Material and Methods. Springer-Verlag; New York: 250–263. [Google Scholar]
- Joshi, P.P. , 2007. Simultaneous downregulation of CDK inhibitors p18(Ink4c) and p27(Kip1) is required for MEN2A-RET-mediated mitogenesis. Oncogene. 26, 554–570. [DOI] [PubMed] [Google Scholar]
- Juan, G. , Darzynkiewicz, Z. , 2001. Bivariate analysis of DNA content and expression of cyclin proteins. Curr Protoc. Cytom. Chapter 7, Unit 7.9 [DOI] [PubMed] [Google Scholar]
- Knoepfler, P.S. , 2002. N-myc is essential during neurogenesis for the rapid expansion of progenitor cell populations and the inhibition of neuronal differentiation. Genes Dev. 16, 2699–2712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kodama, Y. , 2005. The RET proto-oncogene: a molecular therapeutic target in thyroid cancer. Cancer Sci. 96, 143–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lovec, H. , 1994. Cyclin D1/bcl-1 cooperates with myc genes in the generation of B-cell lymphoma in transgenic mice. EMBO J. 13, 3487–3495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loves, H. , 1994. Oncogenic activity of cyclin D1 revealed through cooperation with Ha-ras: link between cell cycle control and malignant transformation. Oncogene. 9, 323–326. [PubMed] [Google Scholar]
- Misawa, A. , 2000. N-Myc induction stimulated by insulin-like growth factor I through mitogen-activated protein kinase signaling pathway in human neuroblastoma cells. Cancer Res. 60, 64–69. [PubMed] [Google Scholar]
- Pei, X.H. , 2009. CDK inhibitor p18(INK4c) is a downstream target of GATA3 and restrains mammary luminal progenitor cell proliferation and tumorigenesis. Cancer Cell. 15, 389–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peukert, K. , 1997. An alternative pathway for gene regulation by Myc. EMBO J. 16, 5672–5686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phelps, D.E. , 1998. Coupled transcriptional and translational control of cyclin-dependent kinase inhibitor p18INK4c expression during myogenesis. Mol. Cell. Biol. 18, 2334–2343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phelps, D.E. , Xiong, Y. , 1998. Regulation of CDK4 during adipogenesis involves switching of cyclin D subunits and concurrent binding of p18INK4c and p27KIP1 . Cell Growth Differ. 9, 595–610. [PubMed] [Google Scholar]
- Sawai, S. , 1991. Embryonic lethality resulting from disruption of both N-myc alleles in mouse zygotes. New Biol. 3, 861–869. [PubMed] [Google Scholar]
- Schneider, A. , 1997. Association of Myc with the zinc-finger protein Miz-1 defines a novel pathway for gene regulation by Myc. Curr. Top. Microbiol. Immunol. 224, 137–146. [DOI] [PubMed] [Google Scholar]
- Sherr, C. , 1993. Mammalian G1 cyclins. Cell. 73, 1059–1065. [DOI] [PubMed] [Google Scholar]
- Sherr, C. , Roberts, J. , 1999. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev. 13, 1501–1512. [DOI] [PubMed] [Google Scholar]
- Sherr, C.J. , 1994. G1 phase progression: cycling on cue. Cell. 79, 551–555. [DOI] [PubMed] [Google Scholar]
- Sherr, C.J. , 1996. Cancer cell cycle. Science. 274, 1672–1677. [DOI] [PubMed] [Google Scholar]
- Soucek, L. , 2008. Modelling Myc inhibition as a cancer therapy. Nature. 455, 679–683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Veelen, W. , 2009. P18 is a tumor suppressor gene involved in human medullary thyroid carcinoma and pheochromocytoma development. Int. J. Cancer. 124, 339–345. [DOI] [PubMed] [Google Scholar]
- Vitagliano, D. , 2004. Regulation of p27Kip1 protein levels contributes to mitogenic effects of the RET/PTC kinase in thyroid carcinoma cells. Cancer Res. 64, 3823–3829. [DOI] [PubMed] [Google Scholar]
- Zindy, F. , 1999. Postnatal neuronal proliferation in mice lacking Ink4d and Kip1 inhibitors of cyclin-dependent kinases. Proc. Natl. Acad. Sci. U.S.A. 96, 13462–13467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zindy, F. , 2006. N-Myc and the cyclin-dependent kinase inhibitors p18Ink4c and p27Kip1 coordinately regulate cerebellar development. Proc. Natl. Acad. Sci. U.S.A. 103, 11579–11583. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
The following are the Supplementary data related to this article:
Supplementary data