Newer
Older
grobid-corpus / fulltext / istex / tei / 7B500C33DCC28A2530BB5A6E4083F9AFDE902102.training.fulltext.tei.xml
<?xml version="1.0" ?>
<tei>
	<teiHeader>
		<fileDesc xml:id="0"/>
	</teiHeader>
	<text xml:lang="en">

		<p>Temporal regulation of c-Myc protein accumulation is essential for<lb/> normal cell
			proliferation. Complete loss of c-Myc or N-Myc function<lb/> results in embryonic
			lethality <ref type="biblio">1,2</ref> , and studies using conditional c-Myc<lb/>
			knockout alleles show that cells lacking c-Myc cease to proliferate and<lb/> exit the
			cell cycle <ref type="biblio">3,4</ref> . In contrast, overexpression of c-Myc in
			cultured<lb/> cells or transgenic animals blocks differentiation, induces
			neoplastic<lb/> transformation and can initiate apoptosis <ref type="biblio">5,6</ref> .
			Moreover, a wide variety<lb/> of naturally occurring tumours exhibit both chromosomal
			transloca-<lb/>tions and amplification of the c-myc locus that result in
			constitutive<lb/> overexpression of c-Myc protein <ref type="biblio">7–9</ref>
			.<lb/></p>

		<p>Myc protein is stabilized after activation of Ras, allowing it to accu-<lb/>mulate to
			high levels <ref type="biblio">10</ref> . Ras promotes stability of c-Myc through
			at<lb/> least two effector pathways: the Raf–MEK–ERK kinase cascade, and the<lb/>
			phosphatidylinositol-3-OH kinase (PI(3)K)–Akt pathway that inhibits<lb/> glycogen
			synthase kinase-3β (GSK-3β) <ref type="biblio">11</ref> . The ERK and GSK-3β<lb/>
			kinases phosphorylate two sites near the amino terminus of c-Myc that<lb/> are highly
			conserved in all mammalian c-Myc isoforms <ref type="biblio">11–14</ref> . These<lb/>
			phosphorylation sites, Thr 58 and Ser 62, exert opposing control on c-<lb/>Myc
			degradation through the ubiquitin-proteasome pathway <ref type="biblio">11</ref> .
			Thus,<lb/> after a growth stimulatory signal, c-myc gene transcription is increased<lb/>
			and newly synthesized c-Myc protein is phosphorylated on Ser 62,<lb/> resulting in its
			stabilization. Phosphorylation at Ser 62 is also required<lb/> for the subsequent
			phosphorylation of c-Myc at Thr 58 by GSK-3β,<lb/> which is associated with c-Myc
			degradation <ref type="biblio">11–14</ref> . During early G1<lb/> phase, however, GSK-3β
			activity is regulated by Ras-mediated activa-<lb/>tion of the PI(3)K/Akt pathway (which
			phosphorylates and inhibits<lb/> GSK-3β), facilitating stabilization of c-Myc <ref
				type="biblio">15</ref> . Later in G1 phase, when<lb/> Akt activity declines, GSK-3β
			becomes active and phosphorylates c-<lb/>Myc on Thr 58, which is important for c-Myc
			turnover.<lb/></p>

		<p>Thr 58 phosphorylation contributes to degradation of c-Myc<lb/> through the ubiquitin
			pathway, as mutation of Thr 58 to alanine<lb/> results in a stable and more oncogenic
			c-Myc protein <ref type="biblio">11–13,16,17</ref> .<lb/> Moreover, the c-Myc T58A
			mutant is no longer a substrate for ubiquiti-<lb/>nation in vivo <ref type="biblio"
				>11</ref> . Nevertheless, how Thr 58 phosphorylation facilitates<lb/> c-Myc
			degradation remains unclear. We now show that Ser 62 dephos-<lb/>phorylation is
			important for ubiquitin-mediated degradation of<lb/> c-Myc and that Thr 58
			phosphorylation promotes dephosphorylation<lb/> of Ser 62. In addition, we show that
			PP2A dephosphorylates Ser 62 on<lb/> c-Myc and that the Pin1 prolyl isomerase, which has
			previously been<lb/> shown to enhance PP2A function <ref type="biblio">18,19</ref> ,
			also regulates c-Myc turnover in<lb/> a process that is dependent on phosphorylation of
			Thr 58. We propose<lb/> that a sequence of Ras-initiated signalling events is required
			to regulate<lb/> c-Myc protein stability and demonstrate that a stabilized c-Myc
			protein<lb/> can replace SV40 small T antigen in the transformation of human
			cells.<lb/></p>

		<head>RESULTS<lb/></head>

		<head>Ubiquitinated c-Myc is phosphorylated on Thr 58, but not on<lb/> Ser 62<lb/></head>

		<p>Newly synthesized c-Myc is phosphorylated on Ser 62, allowing the<lb/> subsequent
			phosphorylation of Thr 58 that is necessary to promote<lb/> ubiquitin-mediated
			degradation of c-Myc <ref type="biblio">11–14</ref> . However, treatment of<lb/> cells
			with a proteasome inhibitor results in the accumulation of species<lb/> of c-Myc with a
			larger relative molecular mass higher (M r ) that is rec-<lb/>ognized by an antibody
			specific for Thr 58-phosphorylated c-Myc,<lb/> but not by an antibody specific for Ser
			62-phosphorylated c-Myc <ref type="biblio">11</ref> .<lb/> This observation suggested
			that ubiquitinated forms of c-Myc might<lb/> be phosphorylated selectively on Thr 58. To
			address this possibility,<lb/> we examined the phosphorylation status of ubiquitinated
			c-Myc.<lb/></p>

		<p>A recombinant adenovirus containing the c-myc gene under the<lb/> control of the
			cytomegalovirus (CMV) enhancer (Ad-Myc) was used<lb/> to produce c-Myc protein in
			otherwise quiescent cells, an approach<lb/> we have employed previously to control the
			conditions in which<lb/> c-Myc accumulates <ref type="biblio">10,11</ref> . We have
			shown that c-Myc is phosphorylated<lb/> at Ser 62 as well as Thr 58 under these
			conditions, and that serum<lb/> stimulation or mutation of Thr 58 increases Ser 62
			phosphoryla-<lb/>tion <ref type="biblio">11</ref> . Here, Ad-Myc-infected cells were
			treated with the proteasome<lb/> inhibitor lactacystin for 6 h and ubiquitinated
			proteins were<lb/> immunoprecipitated from cell lysates with an anti-ubiquitin
			anti-<lb/>body. The immunoprecipitates, along with 10% of the original lysate<lb/> and
			control immunoprecipitates without the ubiquitin antibody,<lb/> were analysed in
			triplicate by SDS–PAGE and immunoblotting with<lb/> either a pan-Myc antibody, a
			phospho-Thr 58 antibody or a phos-<lb/>pho-Ser 62 antibody. A substantial amount of
			ubiquitinated c-Myc<lb/> accumulates after 6 h of proteasome inhibition <ref
				type="figure">(Fig. 1, lane 4)</ref>.<lb/> Ubiquitinated c-Myc runs with a slightly
			higher relative molecular<lb/> mass than c-Myc produced in the absence of proteasome inhibition<lb/>
			<ref type="figure">(Fig. 1, compare lanes 2 and 4)</ref>. In addition, several
			higher-molecu-<lb/>lar-weight species are detected with the pan-Myc antibody that
			we<lb/> assume represent alternate or additional ubiquitinated species of<lb/> c-Myc.
			Ubiquitinated c-Myc, which accumulates in quiescent cells<lb/> after proteasome
			inhibition, is primarily (if not exclusively) phospho-<lb/>rylated on Thr 58 <ref
				type="figure">(Fig. 1, lane 7)</ref>, as there was little or no Ser
			62-<lb/>phosphorylated c-Myc immunoprecipitated by the ubiquitin<lb/> antibody <ref
				type="figure">(Fig. 1, lane 10)</ref>.<lb/></p>

		<head>A role for PP2A in controlling c-Myc degradation<lb/></head>

		<p>The observations that GSK-3β-mediated phosphorylation at Thr 58<lb/> requires earlier
			phosphorylation of c-Myc at Ser 62 <ref type="biblio">(refs 11–14)</ref> and<lb/> that
			ubiquitinated c-Myc is only phosphorylated at Thr 58 suggest that<lb/> the Ser 62
			phosphate is removed in the process of c-Myc ubiquitina-<lb/>tion. Assays using various
			cell-permeable inhibitors specific for com-<lb/>mon serine/threonine phosphatases
			indicated a possible role for PP2A<lb/> in the control of c-Myc stability. The level of
			c-Myc present in quiescent<lb/> cells was increased by okadaic acid <ref type="figure"
				>(Fig. 2a, compare lanes 1 and 2)</ref>,<lb/> which is a potent inhibitor of PP2A
			with some activity against PP1, but<lb/> was not affected by cyclosporin A <ref
				type="figure">(Fig. 2a, lane 3)</ref>, which specifically<lb/> inhibits PP2B. This
			data suggest that PP2A, or possibly PP1, could be<lb/> involved in controlling c-Myc
			protein accumulation.<lb/></p>

		<p>As a more specific test to examine the role of PP2A in controlling<lb/> accumulation of
			c-Myc, we used a recombinant adenovirus that<lb/> expresses SV40 small T antigen, a
			highly selective inhibitor of PP2A.<lb/> Small T antigen functions by displacing
			specific regulatory B subunits<lb/> that normally incorporate into cellular PP2A
			complexes and thereby<lb/> reduces PP2A activity against certain endogenous substrates
			20 .<lb/> Increasing doses of the Ad-small T antigen virus had a marked effect on<lb/>
			c-Myc protein levels, resulting in a substantial increase in the accumu-<lb/>lation of
			c-Myc in quiescent fibroblasts <ref type="figure">(Fig. 2b)</ref>. The effects of<lb/>
			expressing small T antigen on accumulation of endogenous c-Myc in<lb/> quiescent or
			asynchronous REF52 fibroblasts were also examined.<lb/> Endogenous c-Myc accumulation
			was increased substantially by inhi-<lb/>bition of PP2A with small T antigen in either
			serum-starved or asyn-<lb/>chronously growing cells <ref type="figure">(Fig.
			2c)</ref>.<lb/></p>

		<p>To test whether the increase in c-Myc accumulation observed after<lb/> PP2A inhibition is
			caused by protein stabilization, and to rule out<lb/> effects of viral infection on the
			interpretation of our results, the effect<lb/> of okadaic acid on the half-life of
			endogenous c-Myc protein was<lb/> examined by pulse-chase analysis in quiescent REF52
			fibroblasts. c-<lb/>Myc protein produced in growth-arrested cells was short-lived, with
			a<lb/> half-life of 13 min. Importantly, this was true for endogenous c-Myc in<lb/>
			uninfected cells <ref type="figure">(Fig. 2d, top)</ref>, as well as for c-Myc produced
			by aden-<lb/>ovirus <ref type="figure">(Fig. 2d, bottom)</ref>. In contrast, endogenous
			c-Myc produced in<lb/> uninfected quiescent cells treated with okadaic acid was
			significantly<lb/> more stable, with a half-life of 62 min. A similar stabilization of
			c-Myc<lb/> was observed in cells infected with the Ad-small T antigen virus
			(see<lb/></p>

		<figure>Untreated<lb/> cell lysate<lb/> In p u t<lb/> A n t i-U b IP<lb/> C o n t r o l
			IP<lb/> In p u t<lb/> A n t i-U b IP<lb/> C o n t r o l IP<lb/> In p u t<lb/> A n t i -U
			b IP<lb/> C o n t r o l IP<lb/> 105 -<lb/>-105<lb/> -75<lb/> -50<lb/> 75 -<lb/>50
			-<lb/>M r (K)<lb/> Ubi−Myc<lb/> IgG<lb/> c-Myc -<lb/>1<lb/> 2<lb/> 3<lb/> 4<lb/> 5<lb/>
			6<lb/> 7<lb/> 8<lb/> 9<lb/> 1 0<lb/> 1 1<lb/> c-Myc<lb/> c-Myc<lb/> c-Myc (P-T58)<lb/>
			c-Myc (P-S62)<lb/> Blot: anti-<lb/>Figure 1 Ubiquitinated c-Myc protein is
			phosphorylated on Thr 58, but not<lb/> on Ser 62. Quiescent REF 52 fibroblasts were
			infected with Ad-c-Myc (MOI<lb/> = 50). At 16 h after infection, cells maintained in
			low-serum medium were<lb/> treated with 10 µM lactacystin for 6 h, except for the
			samples shown in<lb/> lanes 1 and 2, which represent 10 and 40 µg of untreated
			Ad-c-Myc-<lb/>infected cell lysate, respectively. Lactacystin-treated cells were
			harvested<lb/> and lysates were divided for immunoprecipitation (IP) with an
			anti-ubiquitin<lb/> antibody or control protein A + G beads. The input lysates (10%
			volume),<lb/> anti-ubiquitin IP, and control IP were then analysed in triplicate by<lb/>
			SDS–PAGE and western blotting with an anti-c-Myc monoclonal antibody<lb/> (C-33/sc-42),
			or one of the two phospho-specific c-Myc antibodies 11 . The<lb/> c-Myc and
			ubiquitinated c-Myc proteins are indicated. The 50K band visible<lb/> in lanes 4 and 10
			is the IgG heavy chain, which is recognized by the anti-<lb/>mouse secondary antibody.
			This band is not visible in lane 7 and only faintly<lb/> visible in lane 10 (longer
			exposure), where anti-rabbit secondary antibodies<lb/> were used.<lb/></figure>

		<p><ref type="figure">Supplementary Information, Fig. S1</ref>).<lb/></p>

		<p>In vitro phosphatase assays were then performed to examine the role<lb/> of PP2A in
			targeting the Ser 62 phosphate. c-Myc protein was<lb/> immunoprecipitated from
			Ad-Myc-infected quiescent REF52 fibrob-<lb/>lasts that were co-infected with Ad-Ras to
			maximize phosphorylation<lb/> of c-Myc <ref type="biblio">10</ref> . Immunoprecipitated
			c-Myc was incubated with either<lb/> buffer only (−), or with purified PP1 or PP2A. Ser
			62 and Thr 58 phos-<lb/>phorylation were assessed by western blotting with the Ser 62 or
			Thr 58<lb/> phospho-specific antibodies. PP2A, but not PP1, removed Ser 62
			phos-<lb/>phate from c-Myc without significantly reducing Thr 58 phosphate<lb/>
			<ref type="figure">(Fig. 3a)</ref>. The ability of PP2A to dephosphorylate the stable
			c-Myc<lb/> mutant, c-Myc T58A , was also evaluated. This mutant has enhanced
			Ser<lb/></p>

		<figure>a<lb/> d<lb/> b<lb/> c<lb/> −<lb/> −<lb/> −<lb/> O k a d a ic<lb/> a c id C y c lo s
			p o r in<lb/> A<lb/> c-Myc<lb/> c-Myc<lb/> c-Myc -<lb/>c-Myc -<lb/>Ad-c-Myc -<lb/>c-Myc
			-<lb/>+ OA<lb/> c-Myc -<lb/>N.S.<lb/> Ponceau S<lb/> 1<lb/> 2<lb/> 3<lb/> 1<lb/> 2<lb/>
			3<lb/> 1<lb/> 2<lb/> 3<lb/> Starved<lb/> Asynchronous<lb/> 1<lb/> 2<lb/> 1<lb/> 2<lb/>
			3<lb/> 4<lb/> + Ad-small T<lb/> + A d -s m a ll T<lb/> Chase time (min)<lb/> 0<lb/> 1
			5<lb/> 3 0<lb/> Chase time (min)<lb/> Chase time (min)<lb/> 0<lb/> 1 0<lb/> 2 0<lb/> 35
			S-c-Myc remaining (percentage)<lb/> 100<lb/> 50<lb/> 10<lb/> 0<lb/> 1 0<lb/> 2 0<lb/> 3
			0<lb/> Endogenous control<lb/> (t 1/2 = 12.6 min)<lb/> Endogenous + OA<lb/> (t 1/2 =
			61.8 min)<lb/> Ectopic control<lb/> (t 1/2 = 12.5 min)<lb/> Figure 2 Accumulation of
			c-Myc is regulated by PP2A. (a) Okadaic acid (OA)<lb/> increases accumulation of c-Myc.
			Quiescent REF52 fibroblasts were<lb/> infected with Ad-c-Myc (MOI = 25) and maintained
			in medium with 0.25%<lb/> serum for an additional 20 h. At 4 h before harvesting,
			infected cells were<lb/> either treated with ethanol (−), 100 nM OA or 5 µM cyclosporin
			A. Extracts<lb/> from an equal number of cells were subjected to western blot analysis
			with<lb/> the c-Myc C-33 antibody. Equal protein loading was verified by Ponceau S<lb/>
			staining. (b) SV40 small T antigen increases Ad-c-Myc protein levels.<lb/> Quiescent
			REF52 cells were infected with Ad-c-Myc (MOI = 25), together<lb/> with a control virus
			(−), Ad-GFP (MOI = 200), or a recombinant adenovirus<lb/> expressing SV40 small T
			antigen, Ad-small T (MOIs = 100, 200 and 400 in<lb/> lanes 2–4), and maintained in
			medium containing 0.25% serum. At 20 h<lb/> after infection, equal cell counts were
			harvested for western blot analysis<lb/> with the C-33 c-Myc antibody. Equal protein
			loading was verified by<lb/> Ponceau S staining of the membrane and the presence of a
			non-specific<lb/> (N.S.) band on the blot. (c) SV40 small T antigen increases endogenous
			c-<lb/>Myc protein levels. Quiescent or asynchronous REF52 cells were infected<lb/> with
			either control Ad-GFP virus (−) or Ad small T antigen, both at<lb/> MOI = 200, and
			maintained in growth medium (asynchronous) or starvation<lb/> medium (starved) for an
			additional 20 h. Equal cell numbers were harvested<lb/> and immunoprecipitations were
			performed to concentrate endogenous<lb/> c-Myc, before western blot analysis with the
			c-Myc C-33 monoclonal<lb/> antibody. (d) Okadaic acid stabilizes endogenous c-Myc
			protein. Quiescent<lb/> REF52 fibroblasts were either infected with Ad-c-Myc (MOI = 50)
			and<lb/> maintained in medium with 0.25% serum for 18 h, or not infected.
			Non-<lb/>infected cells were either mock treated with ethanol (control) or treated
			with<lb/> 100 nM okadaic acid (OA) for 6 h. Cells were pulse-labelled with<lb/> 35
			S-methionine/cysteine and chased with medium containing excess<lb/> unlabelled
			methionine and cysteine in the continued presence of ethanol or<lb/> okadaic acid. 35
			S-labelled c-Myc was immunoprecipitated, separated by<lb/> SDS–PAGE and quantified with
			a phosphorimager. Best-fit exponential lines<lb/> are shown for endogenous c-Myc
			control, endogenous c-Myc + OA and<lb/> ectopic c-Myc control. Calculated half-lives are
			also shown.<lb/></figure>

		<p>62 phosphorylation <ref type="biblio">11,14</ref> . Whereas PP2A effectively
			dephosphorylated<lb/> Ser 62 in wild-type c-Myc <ref type="figure">(Fig. 3b,
			left)</ref>, the c-Myc T58A mutant was not<lb/> dephosphorylated by PP2A <ref
				type="figure">(Fig. 3b, right)</ref>. These results suggest a role<lb/> for Thr 58
			phosphorylation in facilitating removal of Ser 62 phosphate<lb/> by PP2A.<lb/></p>

		<p>In addition, accumulation of c-Myc in vivo was assessed using<lb/> RNA interference
			(RNAi) to inhibit endogenous PP2A activity.<lb/> Small-interfering RNA (siRNA) directed
			against the catalytic sub-<lb/>unit (C) of PP2A reduced the level of endogenous C
			subunit, and<lb/> coincident with this reduction there was an increase in the level
			of<lb/> c-Myc protein <ref type="figure">(Fig. 3c)</ref>. In contrast, a scrambled
			sequence siRNA or<lb/> an siRNA targeting luciferase had no effect on the level of the C
			sub-<lb/>unit and did not alter c-Myc levels, although the luciferase siRNA did<lb/>
			inhibit luciferase activity in this experiment (see <ref type="figure"
				>Supplementary<lb/> Information, Fig. S2</ref>). Taken together, these results
			suggest that<lb/> PP2A contributes to degradation of c-Myc by dephosphorylating Ser<lb/>
			62 and imply that this degradation is dependent on Thr 58<lb/> phsophorylation.<lb/></p>

		<figure>a<lb/> b<lb/> c<lb/> Anti-c-Myc-S62-P<lb/> c-Myc (P-S62)<lb/> c-Myc (P-S62)<lb/>
			c-Myc<lb/> c-Myc<lb/> Anti-c-Myc-T58-P<lb/> Anti-c-Myc<lb/> c-Myc<lb/> PP2A C<lb/>
			−<lb/> PP1c<lb/> PP2A<lb/> PP2A:<lb/> −<lb/> +<lb/> +<lb/> +<lb/> c-Myc -<lb/>c-Myc<lb/>
			PP2A:<lb/> −<lb/> +<lb/> +<lb/> +<lb/> c-Myc T58A<lb/> IgG -<lb/>c-Myc -<lb/>IgG
			-<lb/>Time<lb/> (min)<lb/> Time<lb/> (min)<lb/> 20<lb/> 0<lb/> 10<lb/> 1<lb/> 2<lb/>
			3<lb/> 20<lb/> 20<lb/> 0<lb/> 10<lb/> 20<lb/> C o n t r o l R N A i<lb/> L u c if e r a
			s e R N A i<lb/> P P 2 A C<lb/> R N A i<lb/> Blot: anti-<lb/>Blot: anti-<lb/>Blot:
			anti-<lb/> Figure 3 Ser 62 phosphate is a substrate for PP2A. (a) PP2A<lb/>
			dephosphorylates c-Myc at Ser 62. Quiescent REF52 cells were infected<lb/> with Ad-c-Myc
			(MOI = 50) together with Ad-Ras Q61L (MOI = 200),<lb/> maintained in starvation medium
			and harvested 20 h after infection. c-Myc<lb/> was immunoprecipitated from cell lysates
			and incubated with either control<lb/> buffer (−), 1 µg of purified PP1 (human
			recombinant) or 0.1 µg of PP2A<lb/> (bovine kidney), as indicated, at 30 °C for 30 min.
			Samples were run in<lb/> triplicate and immunoblotted with the phospho-Ser 62 antibody,
			the<lb/> phospho-Thr 58 antibody, or the C-33 c-Myc antibody, as indicated. (b) Thr<lb/>
			58 phosphorylation is necessary for PP2A-mediated dephosphorylation of<lb/> Ser 62.
			Quiescent REF52 fibroblasts were infected with wild-type Ad-c-Myc<lb/> or Ad-c-Myc T58A
			virus (MOI = 200) and maintained in low-serum medium<lb/> overnight. At 4 h before
			harvesting, cells were treated with 10 µM MG132<lb/> and 100 nM okadaic acid. c-Myc was
			immunoprecipitated from the cell<lb/> extracts using the C-33 c-Myc monoclonal antibody.
			Immunoprecipitated<lb/> c-Myc samples were incubated with purified PP2A for the
			indicated times.<lb/> Western blot analysis was performed with the phospho-Ser 62
			antibody or<lb/> the C-33 c-Myc antibody. The lower band in the anti-c-Myc blot is IgG.
			(c)<lb/> Specific inhibition of PP2A by siRNA results in accumulation of c-Myc.<lb/>
			293a cells were transfected with 500 ng CMV-c-Myc and 100 ng CMV-β-gal,<lb/> along with
			100nM scrambled siRNA (lane 1), siRNA specific for luciferase<lb/> (lane 2), or pooled
			siRNAs consisting of four different siRNAs specific for<lb/> the PP2A C subunit (lane
			3). Cells transfected with scrambled siRNA and<lb/> luciferase siRNA also received 250
			ng pGL3-luciferase plasmid. Transfected<lb/> cells were serum-starved for 48 h and
			harvested. β-Galactosidase activity<lb/> was measured to adjust protein load volumes for
			transfection efficiency. The<lb/> top half of the western blot was probed with the c-Myc
			antibody (N262) and<lb/> the bottom half with the PP2A C subunit antibody. Luciferase
			assays from<lb/> lane 1 and 2 extracts are presented in Supplementary Information, Fig.
			S2.<lb/></figure>

		<head>The Pin1 prolyl isomerase interacts with c-Myc<lb/></head>

		<p>PP2A may be a conformation-sensitive protein phosphatase, prefer-<lb/>ring the trans
			configuration of a proline residue adjacent to the phos-<lb/>pho-serine or
			phospho-threonine in substrates such as Cdc25 and<lb/> Tau <ref type="biblio">18</ref> .
			In addition, the phosphorylation-directed prolyl isomerase<lb/> Pin1 may catalyse the
			isomerization of proline residues in such phos-<lb/>pho-proteins to promote their
			dephosphorylation by PP2A <ref type="biblio">19</ref> .<lb/> Furthermore, the presence
			of doubly phosphorylated serine/threo-<lb/>nine-proline residues, as found in the motifs
			that comprise the car-<lb/>boxy-terminal domain of RNA polymerase II, increase the
			binding<lb/> affinity and catalytic efficacy of Pin1 towards these substrates
			through<lb/> recognition of one of the two phosphorylated residues by the Pin1-<lb/>WW
			domain <ref type="biblio">21,22</ref> . These findings led us to test whether c-Myc
			could<lb/> bind to Pin1. As an alternative approach, we have used quiescent<lb/> REF52
			fibroblasts infected with Ad-Myc, made extracts from these cells<lb/> and incubated them
			with glutathione S-transferase (GST) alone,<lb/> GST–Pin1 or GST–Pin1 bearing point
			mutations in either the WW<lb/> domain or the catalytic domain <ref type="biblio"
				>30</ref> immobilized on glutathione–agarose<lb/> beads. c-Myc bound preferentially
			to wild-type GST–Pin1 beads, com-<lb/>pared with GST alone <ref type="figure">(Fig. 4a,
				compare lanes 2 and 3)</ref>. c-Myc inter-<lb/>acted poorly with the Pin1 W33A WW
			mutant <ref type="figure">(Fig. 4a, )lane 4</ref>, but<lb/> bound the Pin1 C109A
			catalytic mutant with similar efficacy to wild-type<lb/> Pin1 <ref type="figure">(Fig.
				4a, compare lanes 3 and 5)</ref>.<lb/></p>

		<p>To investigate the role of Thr 58 and Ser 62 phosphorylation in the<lb/> Pin1–Myc
			interaction, the ability of wild-type c-Myc, c-Myc T58A or<lb/> c-Myc S62A mutants <ref
				type="biblio">11</ref> to interact with Pin1 in vivo was examined. 293a<lb/> cells
			were transfected with vectors to express His 6 -tagged wild-type or<lb/> mutant c-Myc
			proteins. Cells were lysed and His 6 –Myc proteins were<lb/> selected on nickel–NTA
			beads. Bound material was eluted with imidi-<lb/>zole and then analysed by western
			blotting. Pin1 was recovered in asso-<lb/>ciation with wild-type c-Myc <ref
				type="figure">(Fig. 4b, compare lanes 1 and 2)</ref>, but a<lb/> reduced interaction
			was detected with either c-Myc T58A or c-Myc S62A<lb/>
			<ref type="figure">(Fig. 4b, lanes 3 and 4)</ref>. As c-Myc T58A remains highly
			phosphorylated<lb/> on Ser 62, whereas c-Myc S62A lacks phosphorylation at either Thr 58
			or<lb/> Ser 62 <ref type="biblio">(refs 11–14)</ref>, these results suggest that the
			phosphorylation of<lb/> c-Myc at Thr 58 is important for Pin1 binding.<lb/></p>

		<head>Pin1 facilitates c-Myc protein degradation<lb/></head>

		<p>We next examined whether Pin1 expression affected the level of c-Myc<lb/> protein by
			utilizing primary mouse embryo fibroblasts (MEFs) derived<lb/> from Pin1 −/− embryos.
			Heterozygous Pin1 mice, maintained in an iso-<lb/>genic C57BL/6J background, were bred,
			and sibling 13.5-day embryos<lb/> were used to prepare primary MEFs with either +/+ or
			−/− genotypes <ref type="biblio">23</ref> .<lb/> Substantially more c-Myc protein
			accumulated in Pin1 −/− cells, com-<lb/>pared with wild-type MEFs after Ad-Myc infection
				<ref type="figure">(Fig. 5a, compare<lb/> )lanes 1 and 2</ref>. This increase in
			c-Myc levels was caused by the absence<lb/> of Pin1, as re-introduction of Pin1 (using
			Ad-Pin1) into the Pin1 −/−<lb/> MEFs reduced c-Myc levels to those detected in wild-type
			MEFs <ref type="figure">(Fig.<lb/> 5a, compare lanes 1 and 3)</ref>. Furthermore, a
			pulse-chase experiment<lb/> demonstrated that the increased accumulation of c-Myc in the
			absence<lb/> of Pin1 was associated with an increased half-life of c-Myc in Pin1
			−/−<lb/> MEFs <ref type="figure">(Fig. 5b)</ref>. Moreover, additional pulse-chase
			assays demonstrated<lb/> that re-introduction of Pin1 into Pin −/− cells reduced the
			half-life of<lb/> c-Myc in these cells (see <ref type="figure">Supplementary
				Information, Fig. S3</ref>).<lb/></p>

		<p>The accumulation of c-Myc observed in the absence of Pin1 was not<lb/> significantly
			affected by inhibition of PP2A through expression of<lb/> small T antigen <ref
				type="figure">(Fig. 5c, lanes 2 and 4)</ref>, when compared with the effects<lb/> of
			small T antigen expression on c-Myc in Pin1 +/+ cells <ref type="figure">(Fig. 5c, lanes
				1<lb/> and 3)</ref>. This result suggests a similar role for the actions of Pin1
			and<lb/> PP2A on c-Myc. Moreover, the enhancement of wild-type c-Myc accu-<lb/>mulation
			in the absence of Pin1 <ref type="figure">(Fig. 5d, top)</ref> was not observed for
			the<lb/> c-Myc T58A mutant <ref type="figure">(Fig. 5d, bottom)</ref>, consistent with
			the observation<lb/> that this mutant shows reduced binding to Pin1 and displays
			increased<lb/> Ser 62 phosphorylation <ref type="biblio">11</ref> . Taken together,
			these results suggest a com-<lb/>plementary role for Pin1 and PP2A in controlling c-Myc
			turnover.<lb/></p>

		<head>Pin1 ensures transient accumulation of c-Myc after growth<lb/> stimulation<lb/></head>

		<p> Given the role of Pin1 in the degradation of c-Myc, we investigated the<lb/>
			contribution of Pin1 to the normal regulation of c-Myc levels after<lb/> growth
			stimulation. Wild-type or Pin1 −/− primary MEFs were density<lb/> arrested and then
			plated into low-serum medium for 24 h. The quies-<lb/>cent MEFs were then stimulated
			with 20% foetal calf serum and<lb/> endogenous c-Myc protein levels were determined by
			western blotting<lb/> at 0, 3, 6 and 24 h after serum addition. Endogenous c-Myc in
			wild-type<lb/></p>

		<figure>a<lb/> b<lb/> In p u t<lb/> G S T<lb/> G S T − P in 1<lb/> G S T − P in 1 -W W<lb/>
			H is − c -M y c T 5 8 A<lb/> H is − c -M y c S 6 2 A<lb/> G S T − P in 1<lb/> (c a t a
			ly t ic<lb/> m u t a n t )<lb/> c-Myc<lb/> 1<lb/> 2<lb/> 3<lb/> 4<lb/> 1<lb/> 2<lb/>
			3<lb/> 4<lb/> 5<lb/> Blot: anti-c-Myc<lb/> c-Myc<lb/> Pin1<lb/> −<lb/> H is − c -M y c W
			T<lb/> Blot: anti-<lb/>Figure 4 Pin1 interacts with c-Myc. (a) Interaction between c-Myc
			and Pin1<lb/> requires the Pin1 WW domain. Quiescent REF52 fibroblasts were
			infected<lb/> with Ad-c-Myc (MOI = 25 ). Infected cells were maintained in medium
			with<lb/> 0.25% serum for 20 h and then harvested in low-stringency buffer. Cell<lb/>
			lysates from equal cell counts were incubated with either GST, or GST–Pin1<lb/>
			wild-type or mutant proteins bound to glutathione–sepharose beads, as<lb/> indicated.
			Precipitated proteins were washed four times in low-stringency<lb/> buffer and then
			released with SDS sample buffer. Samples plus a 10%<lb/> volume of input lysate were
			subjected to western blot analysis with the C-33<lb/> c-Myc antibody. (b) c-Myc and Pin1
			interact in vivo. 293a cells were<lb/> transfected with 2 µg pD40-His-c-Myc,
			pD40-His-c-Myc T58A , pD40-His-c-<lb/>Myc S62A or empty pD40-His vector (−), as
			indicated, plus 2 µg Ad-Trk-Pin1<lb/> and 0.5 µg CMV-β-gal. Cells were serum-starved for
			48 h and then<lb/> harvested by lysis in low-stringency co-immunoprecipitation
			buffer.<lb/> Supernatants were normalized for β-galactosidase levels and incubated
			with<lb/> nickel–NTA agarose beads at 4 °C for 3–4 h to precipitate His 6 -tagged<lb/>
			c-Myc proteins. Precipitated proteins were washed with co-<lb/>immunoprecipitation
			buffer containing 20 mM imidazole and eluted with<lb/> co-immunoprecipitation buffer
			containing 250 mM imidazole. Eluted<lb/> proteins were analysed by western blotting. The
			upper half of the western<lb/> blot was probed with anti-Myc (N262) and the bottom half
			with anti-Pin1,<lb/> as indicated.<lb/></figure>

		<p>MEFs exhibits a typical temporal pattern <ref type="biblio">11</ref> : that is, a low
			level in quies-<lb/>cent cells <ref type="figure">(Fig. 6, lane 1)</ref>, a peak
			accumulation at 3 h <ref type="figure">(Fig. 6, lane 2)</ref>, a<lb/> decline at 6 h
				<ref type="figure">(Fig. 6, lane 3)</ref> and a further decrease to a basal
			steady-<lb/>state level by 24 h <ref type="figure">(Fig. 6, lane 4)</ref>. Quiescent
			MEFs lacking Pin1 also<lb/> showed undetectable levels of c-Myc protein before serum
			stimulation,<lb/> because of very low levels of c-myc gene transcription in
			growth-<lb/>arrested cells <ref type="figure">(Fig. 6, lane 5)</ref>. Endogenous c-Myc
			was induced at 3 h<lb/> after serum stimulation in Pin1 −/− cells <ref type="figure"
				>(Fig. 6, lane 6)</ref>. In contrast to<lb/> wild-type MEFs, however, c-Myc levels
			increased continuously with<lb/> time, reaching maximal levels at 24 h after serum
			stimulation <ref type="figure">(Fig. 6,<lb/> lane 8)</ref>. This result is very similar
			to that resulting from inhibition of<lb/> GSK-3β activity <ref type="biblio">11</ref>
			and is consistent with stabilization of c-Myc in the<lb/> Pin1 −/− cells. In addition,
			the increased levels of c-Myc in the Pin1 −/−<lb/> cells correlated with enhanced Ser 62
			phosphorylation <ref type="figure">(Fig. 6, second<lb/> panel)</ref>. This is
			particularly notable at the 6-h time point, when total lev-<lb/>els of c-Myc are largely
			equivalent between wild-type and Pin1 −/−<lb/> MEFs, but when phospho-Ser 62 levels are
			much higher in the Pin1 −/−<lb/> MEFs <ref type="figure">(Fig. 6, compare lanes 3 and
				7)</ref>. This result was also supported by<lb/> treatment of wild-type MEFs with
			okadaic acid, which also increased<lb/> Ser 62 phosphorylation and prolonged c-Myc
			expression in response<lb/> to growth stimulation (data not shown). Examination of Pin1
			levels in<lb/> the same samples demonstrates that Pin1 expression in wild-type<lb/> MEFs
			is not affected by serum stimulation <ref type="figure">(Fig. 6)</ref>. These results
			sug-<lb/>gest that one way in which Pin1 promotes a decrease in c-Myc levels is<lb/>
			through enhancing Ser 62 dephosphorylation.<lb/></p>

		<head>Control of c-Myc phosphorylation affects c-Myc transactivation<lb/>
			capacity<lb/></head>

		<p>A primary function of c-Myc is to activate transcription of a large<lb/> number of target
			genes encoding proteins important for cell growth. To<lb/> assess the functional
			significance of the regulation of c-Myc phospho-<lb/>rylation and turnover by PP2A, the
			transcriptional activation capacity<lb/> of c-Myc was assessed in the absence or
			presence of small T antigen.<lb/> Previously, we demonstrated a role for c-Myc in
			activation of the E2F2<lb/> promoter that is dependent on the presence of c-Myc-binding
			sites<lb/> (E-box elements) in the promoter <ref type="biblio">24</ref> . To examine how
			controlling<lb/> c-Myc degradation affects its function, we reduced the level of
			Ad-Myc<lb/> sufficiently to minimally activate the E2F2 promoter (<ref type="figure"
				>Fig. 7</ref>; E2F2-Luc,<lb/> compare control with c-Myc). We also observed only
			minimal effects<lb/> on the activity of the E2F2 promoter as a result of expressing
			small T<lb/></p>

		<figure>WT<lb/> Pin1 −/−<lb/> Pin1 −/−<lb/> Pin1 +/+<lb/> Pin1<lb/> +/+<lb/> Pin1<lb/>
			+/+<lb/> Pin1<lb/> −/−<lb/> Pin1<lb/> −/−<lb/> Pin1<lb/> +/+<lb/> Pin1<lb/> −/−<lb/>
			Pin1 +/ ++<lb/> (t 1/2 = 9.2 min)<lb/> Pin1 −/−<lb/> (t 1/2 =137.7 min)<lb/> + C o n t r
			o l<lb/> + C o n t r o l<lb/> + P in 1<lb/> c-Myc<lb/> c-Myc<lb/> c-Myc<lb/> c-Myc<lb/>
			c-Myc<lb/> c-Myc T58A<lb/> Ponceau S<lb/> 1<lb/> 0<lb/> 1<lb/> 2<lb/> 3<lb/> 4<lb/>
			1<lb/> 2<lb/> 3<lb/> 4<lb/> 7<lb/> 15<lb/> 30<lb/> 2<lb/> 3<lb/> Chase time (min)<lb/>
			Chase time (min)<lb/> a<lb/> c<lb/> d<lb/> b<lb/> 100<lb/> 1<lb/> 0<lb/> 1 0<lb/> 3
			0<lb/> 20<lb/> 10<lb/> Ad-small T antigen<lb/> 35 S-c-Myc remaining (percentage)<lb/>
			Figure 5 Accumulation of c-Myc is regulated by Pin1. (a) c-Myc levels<lb/> are elevated
			in Pin1 −/− cells. Passage-3 primary MEFs from siblings with<lb/> either wild-type (WT)
			or Pin1 −/− genotypes were made quiescent by<lb/> serum starvation for 48 h and then
			infected with Ad-c-Myc (MOI = 100)<lb/> together with either Ad-GFP (control) or Ad-Pin1
			(both at MOI = 400).<lb/> Infected cells were maintained in medium with 0.25% serum for
			20 h<lb/> and extracts prepared from an equal number of cells for each condition<lb/>
			were subjected to western blot analysis with the C-33 c-Myc antibody.<lb/> (b) c-Myc
			half-life is increased in the absence of Pin1. Quiescent<lb/> primary Pin1 +/+ or Pin1
			−/− MEFs were infected with Ad-c-Myc<lb/> (MOI = 100) and maintained in low-serum
			medium. At 18 h after<lb/> infection, cells were labelled with 35 S-methionine/cysteine
			for 30 min<lb/> and chased in medium with 0.20% serum containing excess unlabelled<lb/>
			methionine and cysteine for the indicated times. Labelled c-Myc was<lb/>
			immunoprecipitated from equal cell numbers at each time, analysed by<lb/> SDS–PAGE and
			then quantified with a phosphorimager. Best-fit<lb/> exponential lines are shown for
			c-Myc expressed in Pin1 +/+ and Pin1 −/−<lb/> MEFs. (c) c-Myc protein levels in Pin1 −/−
			MEFS are not affected by small<lb/> T antigen. Quiescent primary Pin1 +/+ or Pin1 −/−
			MEFs were infected with<lb/> Ad-c-Myc (MOI = 100) and either Ad-GFP (control; MOI = 400)
			or Ad-<lb/>small T (MOI = 400). Infected cells were treated as described in a.<lb/>
			Equal protein loading was confirmed by Ponceau S staining and<lb/> measurement of a
			reference band. (d) The c-Myc T58A mutant is not<lb/> affected by deletion of Pin1.
			Quiescent primary Pin1 +/+ or Pin1 −/− MEFs<lb/> (passage 3) were infected with Ad-c-Myc
			or Ad-c-Myc T58A (MOI = 100)<lb/> and treated as described in a.<lb/></figure>

		<p>antigen. In contrast, co-expression of small T antigen and c-Myc<lb/> resulted in a
			substantial increase in E2F2 promoter activity, and this<lb/> activation was dependent
			on the presence of intact c-Myc-binding sites<lb/> in the promoter (<ref type="figure"
				>Fig. 7</ref>; E2F2(-Ebox)-Luc, c-Myc + tAg). We conclude<lb/> that the control of
			c-Myc phosphorylation not only regulates the stabil-<lb/>ity of c-Myc, but also has a
			direct impact on c-Myc function.<lb/></p>

		<head>Stabilized c-Myc can replace small T antigen in transformation<lb/> of human
			cells<lb/></head>

		<p>Human fibroblasts can be transformed by a combination of Ras,<lb/> telomerase and SV40
			large T and small T antigens <ref type="biblio">25</ref> . Given the role of<lb/> small
			T antigen in regulating PP2A, together with the role for PP2A in<lb/> mediating the
			destabilization of c-Myc, we evaluated whether a stabi-<lb/>lized form of c-Myc could
			replace small T antigen as a necessary<lb/> requirement for transformation and
			tumorigenesis. Human embry-<lb/>onic kidney (HEK) cells expressing large T antigen, the
			catalytic sub-<lb/>unit of human telomerase (hTERT) and Ras G12V (hereby referred to
			as<lb/> HEK-TER cells) cannot grow in soft agar (a measure of transformed<lb/> cell
			growth) or form tumours in mice, unless small T antigen is intro-<lb/>duced <ref
				type="biblio">26</ref> . We ectopically expressed the stable c-Myc T58A mutant in
			the<lb/> HEK-TER cells or independently expressed small T antigen (as a posi-<lb/>tive
			control), wild-type c-Myc or c-Myc S62A (which cannot be phos-<lb/>phorylated and
			consequently is rapidly degraded 11 ) as negative<lb/> controls, and developed cell
			populations that expressed each of the four<lb/> proteins. These cell populations were
			then assayed for anchorage-inde-<lb/>pendent growth in soft agar, the best in vitro
			corollary to tumorigenic<lb/> growth in vivo. Addition of small T antigen promoted
			aggressive trans-<lb/>formed growth, as previously reported <ref type="biblio">26</ref>
			, whereas cells expressing wild-<lb/>type c-Myc or c-Myc S62A failed to grow in an
			anchorage-independent<lb/> manner <ref type="figure">(Fig. 8a)</ref>. However, the cells
			expressing stabilized c-Myc T58A<lb/> protein grew in soft agar identically to cells
			expressing small T antigen<lb/>
			<ref type="figure">(Fig. 8a)</ref>. Similar transformation trends for the c-Myc S62A
			and<lb/> c-Myc T58A mutants have been reported previously in rodent cells <ref
				type="biblio">12,13</ref> .<lb/> To confirm our result, mixed populations were
			examined, as well as<lb/> clonally isolated cell lines expressing c-Myc T58A . All these
			cell lines<lb/> exhibited growth in soft agar, whereas none of the wild-type c-Myc<lb/>
			populations were able to grow (see <ref type="figure">Supplementary Information,
				Fig.<lb/> S4</ref>). Finally, c-Myc T58A was tested in a second human cell type —
			BJ<lb/> fibroblasts also containing small T antigen, hTERT and Ras G12V — and<lb/>
			compared with small T antigen as a positive control or vector alone as a<lb/> negative
			control. c-Myc T58A -expressing cells grew as well as the small-<lb/>T-antigen -positive
			control cells <ref type="figure">(Fig. 8a)</ref>. Thus, a stable, and therefore<lb/>
			oncogenic, c-Myc can functionally replace small T antigen in trans-<lb/>formed cell
			growth of human cells expressing a defined set of genes.<lb/></p>

		<p>We then questioned whether c-Myc T58A could also replace small T<lb/> antigen in the most
			stringent test of human oncogenesis: in vivo tumour<lb/> growth. The four HEK-TER cell
			populations described above were<lb/> assayed for tumour growth in immuno-compromised
			mice. As previ-<lb/>ously observed, small T antigen was essential for primary human
			cells<lb/> expressing large T antigen, hTERT and Ras G12V to form tumours <ref
				type="figure">(Fig.<lb/> 8b)</ref>. Consistent with soft agar assays, mice injected
			with cells expressing<lb/> wild-type c-Myc or the c-Myc S62A mutant completely failed to
			exhibit<lb/> tumour growth for up to 35 days, which is three times longer than it<lb/>
			takes positive control cells expressing small T antigen to form tumours.<lb/>
			Strikingly, cells expressing c-Myc T58A formed tumours in an almost<lb/> identical
			manner to cells expressing small T antigen <ref type="figure">(Fig. 8b)</ref>. On
			the<lb/> basis of these results, we conclude that the oncogenic transformation of<lb/>
			human cells can be accomplished by the combined action of large T<lb/> antigen, Ras,
			telomerase, and an activated, stabilized, c-Myc. Thus, at<lb/> least one functional
			consequence of c-Myc stabilization in human cells<lb/> is to sensitize the cells to
			transformation and tumorigenesis.<lb/></p>

		<head>DISCUSSION<lb/></head>

		<p>Although an increase in protein levels could be achieved in principle<lb/></p>

		<figure>WT<lb/> Pin −/−<lb/> 0<lb/> 3<lb/> 6<lb/> 2 4<lb/> 0<lb/> 3<lb/> 6<lb/> 2 4<lb/>
			c-Myc<lb/> c-Myc-P-S62<lb/> α-tubulin<lb/> Pin1<lb/> 1<lb/> 2<lb/> 3<lb/> 4<lb/> 5<lb/>
			6<lb/> 7<lb/> 8<lb/> (h)<lb/> Time<lb/> Figure 6 Pin1 is essential for normal control of
			c-Myc accumulation.<lb/> Accumulation of endogenous c-Myc is prolonged in Pin1 −/−
			fibroblasts and<lb/> shows increased Ser 62 phosphorylation. Primary Pin1 +/+ or Pin1
			−/− MEFs<lb/> (passage 2 ) were density growth-arrested, plated at low density into
			medium<lb/> with 0.25% serum and incubated for 24 h. These quiescent cells were
			then<lb/> stimulated by the addition of 20% FCS for the indicated times. Equal cell<lb/>
			counts from serum-starved (0 h) or stimulated (3, 6, and 24 h) cells were<lb/> harvested
			and subjected to western blot analysis with the mouse monoclonal<lb/> C33 c-Myc antibody
			(top). The blot was stripped and re-probed with the<lb/> rabbit polyclonal phosphoSer62
			antibody (second panel) or the α-tubulin<lb/> antibody (third panel). Samples were then
			re-run and probed with the anti-<lb/>Pin1 antibody (bottom).<lb/></figure>

		<p>through transcriptional activation coupled with a short half-life of the<lb/> protein,
			the action of Ras provides a mechanism to amplify c-Myc<lb/> accumulation by stabilizing
			this otherwise short-lived protein. We sug-<lb/>gest that the role of the inter-related
			phosphorylation and dephospho-<lb/>rylation events is to ensure that Ras-mediated
			amplification of c-Myc<lb/> protein levels is indeed transient and self-limited. The
			Raf–MEK–ERK<lb/> pathway stabilizes c-Myc by enhancing Ser 62 phosphorylation, and<lb/>
			the PI(3)K/Akt pathway prevents the subsequent phosphorylation of c-<lb/>Myc at Thr 58
			by inhibiting GSK-3β. However, as Ras activity declines<lb/> after cessation of the
			growth stimulus, PI(3)K and Akt activities also<lb/> decline, resulting in reactivation
			of GSK-3β and phosphorylation of c-<lb/>Myc on Thr 58. Our data suggest that
			phosphorylation of Thr 58 is<lb/> important for recognition of c-Myc by the Pin1 prolyl
			isomerase, the<lb/> actions of which facilitate c-Myc dephosphorylation at Ser 62 by
			PP2A,<lb/> which then promotes c-Myc turnover by the ubiquitin-proteasome<lb/> pathway.
			Thus, the very mechanism that stabilizes and amplifies c-Myc<lb/> accumulation — c-Myc
			phosphorylation at Ser 62 — also triggers the<lb/> subsequent phosphorylation at Thr 58
			and the series of events that cul-<lb/>minate in degradation of c-Myc.<lb/></p>

		<p>Considerable evidence suggests that Thr 58 phosphorylation is criti-<lb/>cal for ensuring
			the transient and timely degradation of c-Myc. All v-<lb/>myc genes recovered in
			transforming retroviruses harbour a mutation<lb/> at Thr 58 <ref type="biblio">(ref.
				27)</ref>. Similarly, a large number of c-myc genes amplified<lb/> in Burkitt&apos;s
			lymphoma carry a mutation at Thr 58, as well as in other<lb/> residues between amino
			acids 57 and 63 <ref type="biblio">(refs 12, 13, 16, 17 and 28)</ref>.<lb/> Assay of
			these mutants generally demonstrates their increased onco-<lb/>genic potential in both
			cell transformation assays and animals <ref type="biblio">12,13,29</ref> . In<lb/>
			conjunction with other studies, our results confirm that mutation of<lb/> c-Myc at Thr
			58 increases the stability of c-Myc, highlighting a key role<lb/> for this
			phosphorylation site in controlling c-Myc degradation <ref type="biblio">11,16,17</ref>
			.<lb/> These results are consistent with a model in which the Pin1 WW<lb/> domain binds
			to c-Myc phosphorylated at Thr 58 in a manner that<lb/> could result in a conformational
			change that makes phospho-Ser 62 an<lb/> ideal site for dephosphorylation by PP2A. Our
			additional finding that a<lb/> stabilized c-Myc protein can completely substitute for
			small T antigen<lb/> by collaborating with Ras, hTERT and small T antigen in the
			transfor-<lb/>mation and oncogenic conversion of primary human cells, whereas<lb/>
			wild-type c-Myc cannot do so, establishes the importance of c-Myc sta-<lb/>bility in
			human cancer.<lb/></p>

		<head>METHODS<lb/></head>

		<p>Antibodies. The C-33 and N262 c-Myc antibodies are from Santa Cruz<lb/> Biotechnology
			(Santa Cruz, CA). The ubiquitin antibody is from Zymed (South<lb/> San Francisco, CA).
			The Thr 58 phosphospecific antibody is from Upstate Cell<lb/> Signaling (Waltham, MA).
			The Ser 62 phosphospecific antiserum was prepared<lb/> as described previously <ref
				type="biblio">11</ref> . The Pin1 antibody was prepared as described
			previ-<lb/>ously <ref type="biblio">30</ref> . The α-tubulin antibody is from Sigma (St
			Louis, MO). The PP2A cat-<lb/>alytic unit antibody is from BD Biosciences (San Jose,
			CA).<lb/></p>

		<p>Plasmids. CMV -Myc, CMV-β-gal, E2F2-Luc, and E2F2(-Ebox3)-Luc plasmids<lb/> have been
			previously described <ref type="biblio">24</ref> . His 6 -tagged c-Myc was created by
			PCR ampli-<lb/>fication of the coding sequence for wild-type murine c-Myc2 from the
			CMV-<lb/>Myc plasmid with a CACC 5′ extension added to the 5′ primer for cloning
			into<lb/> the TOPO entry clone from Invitrogen (Carlsbad, CA). This sequence was
			then<lb/> moved into the pDEST-40 mammalian expression vector using Gateway
			tech-<lb/>nology (Invitrogen), creating pD40-His-c-Myc. His 6 -tagged c-Myc T58A
			and<lb/> His 6 -tagged c-Myc S62A were constructed similarly, except that PCR
			amplifica-<lb/>tion was from the CMV-Myc T58A and CMV-Myc S62A plasmids previously<lb/>
			described <ref type="biblio">11</ref> , creating pD40-His-c-Myc(T58A) and pD40-His-c-Myc
			S62A . The<lb/> Ad-Trk-Pin1 plasmid was constructed as described below.<lb/></p>

		<p>Cells. REF52 cells were grown in DMEM containing 5% foetal calf serum (FCS)<lb/> and 5%
			calf serum (CS). To bring REF52 cells to quiescence, cells were plated at<lb/> ~3,500
			cells cm −2 and incubated overnight. The next day the culture medium<lb/> was replaced
			with DMEM containing 0.25% serum (FCS/CS) and cells were<lb/> incubated for an
			additional 48 h. Primary MEFs were grown in DMEM contain-<lb/>ing 15% heat-inactivated
			FCS. MEFs were made quiescent either by plating pas-<lb/>sage-3 MEFs at a density of
			approximately 2,000 cells cm −2 and the next day<lb/> replacing the medium with DMEM
			containing 0.2% heat-inactivated FCS for<lb/> 48 h, or by allowing passage-2 MEFs to
			grow for 24 h after they had reached con-<lb/>fluence and then plating them directly
			into DMEM/0.2% serum at 4,000 cells<lb/> cm −2 for 24 h. Where indicated, cells were
			serum-stimulated by adding FCS<lb/> directly to the starvation media to a final
			concentration of 20%. 293 cells<lb/> obtained from the American Type Culture Collection
			were grown in DMEM<lb/> containing 10% FCS. For serum-starvation conditions,
			exponentially growing<lb/> 293 cells were washed with DMEM, refed with DMEM containing
			2% FCS and<lb/> incubated for 48 h.<lb/></p>

		<p>Viruses. Stocks of purified virus were created as described previously <ref type="biblio"
				>31</ref> . Viral<lb/> titres (MOI) were determined in 293 cells using an indirect
			immunofluores-<lb/>cence microscopy assay specific for the viral 72K E2 gene product,
			as<lb/> described <ref type="biblio">32</ref> . Experimental comparisons demonstrate
			that REF52 fibroblasts are<lb/> approximately 400-fold less susceptible to infection
			than 293 cells, and MEFs are<lb/> approximately 1,200-fold less susceptible. Quiescent
			REF52 or MEFs were<lb/> infected with virus by incubation in DMEM containing 20 mM Hepes
			at pH 7.2<lb/> for 75 min at 37 °C at a cell-to-volume ratio of 5 × 10 5 cells ml −1 .
			After infec-<lb/>tion, four volumes of DMEM/0.25% (REF52) or 0.2% (MEF) serum were
			added<lb/> to each plate and the cells were further incubated at 37 °C for 18–20 h
			before<lb/> harvest and analysis. The construction of Ad-c-Myc, Ad-c-Myc T58A and
			Ad-c-<lb/>Myc S62A has been described 11 . Ad-GFP has been described previously <ref
				type="biblio">33</ref> . Ad<lb/> small T antigen was constructed by digesting the
			pCEP small T plasmid with<lb/> SmaI and HindIII, and ligating this fragment containing
			the SV40 small T anti-<lb/>gen cDNA into the HindIII/EcoRV sites in the AdTrack-CMV
			plasmid used to<lb/></p>

		<figure>E2F2-Luc<lb/> E2F2(-Ebox)-Luc<lb/> 4.00<lb/> 3.00<lb/> 2.00<lb/> 1.00<lb/> 0.00<lb/>
			C o n t r o l<lb/> c -M y c<lb/> t -A g<lb/> c -M y c + t -A g<lb/> C o n t r o l<lb/> c
			-M y c<lb/> t -A g<lb/> c -M y c + t -A g<lb/> Fold luciferase activity<lb/> Figure 7
			Small T antigen enhances the transactivation function of c-Myc.<lb/> REF52 fibroblasts
			were transfected by calcium phosphate co-precipitation<lb/> with either wild-type
			E2F2-luciferase construct (E2F2-Luc) or mutant<lb/> E2F2-luciferase construct containing
			point mutations in the three E-box<lb/> c-Myc-binding sites (which eliminates c-Myc
			binding to the E2F2<lb/> promoter; E2F2(-Ebox)-Luc), plus CMV-β-gal as an internal
			control. After<lb/> 16 h, cells were placed in DMEM containing 0.25% FCS for 48 h.<lb/>
			Transfected cells were then infected with Ad-GFP (MOI = 200) (Ctrl), Ad-<lb/>c-Myc (MOI
			= 100), Ad-small T antigen (MOI = 100) or Ad-c-Myc<lb/> (MOI = 100) + Ad-small T antigen
			(MOI = 100), as indicated. Cells were<lb/> harvested 20 h after infection, and
			luciferase and β-galactosidase<lb/> activities were measured. Luciferase activity was
			normalized to β-<lb/>galactosidase activity, and data are presented as fold activation
			over<lb/> control. Transfections were performed in two separate experiments and<lb/> the
			mean fold induction from three separate data points per condition is<lb/> shown, with
			error bars indicating standard deviations.<lb/></figure>

		<p>construct recombinant adenoviruses as described <ref type="biblio">34</ref> . Ad-Pin was
			similarly con-<lb/>structed by digesting the pET28-Pin1 plasmid with BamHI and XhoI and
			then<lb/> ligating this fragment containing the human Pin1 cDNA into the BglII/XhoI<lb/>
			sites in AdTrack-CMV, creating the plasmid Ad-Trk-Pin1 used to construct the<lb/>
			Ad-Pin1 virus.<lb/></p>

		<p>Detection of ubiquitinated Myc. Cells were washed once with cold PBS and<lb/> lysed with
			RIPA buffer containing standard protease and phosphatase<lb/> inhibitors, as described
				<ref type="biblio">10</ref> , with the addition of the de-ubiquitinase inhibitor
			5<lb/> mM N-ethylmaleimide (NEM; Sigma) at a cell-to-volume ratio of 1 × 10 6 cells<lb/>
			ml −1 . Ubiquitinated proteins were immunoprecipitated from cell lysates with an<lb/>
			anti-ubiquitin antibody from Zymed. After precipitation with the protein<lb/> A/protein
			G–agarose beads, immunoprecipitates were washed once with low-<lb/>stringency buffer
			containing PBS, 0.1% NP40 (Sigma) and all inhibitors used<lb/> with the RIPA buffer.
			Immunoprecipitated proteins were then separated by 10%<lb/> SDS–PAGE, blotted to
			immobilon-P membrane and detected with the indicated<lb/> c-Myc antibodies.<lb/></p>

		<p>Western blotting. Cells were harvested by scraping into hot 1× SDS sample<lb/> buffer at
			a cell-to-volume ratio of 2.5 × 10 6 cells ml −1 . Protein concentrations<lb/> were
			determined for normalization purposes using the BCA solutions (Sigma)<lb/> according to
			the manufacturer&apos;s instructions. Equal protein for each sample was<lb/> separated
			by SDS–PAGE and blotted to Immobilon-P membrane (Millipore,<lb/> Billerica, MA). Primary
			antibodies were in 5% non-fat milk, PBS, 0.1%<lb/> Tween20, except anti-phospho-Ser 62
			antibody which was in 5% heat-inacti-<lb/>vated horse serum/5% BSA, PBS, 0.1% Tween20.
			Primary antibodies were<lb/> detected with HRP-conjugated secondary antibodies using the
			ECL reagents<lb/> from Amersham according to the manufacturer&apos;s
			instructions.<lb/></p>

		<p>Immunoprecipitations. Cells were harvested by washing once in cold PBS, fol-<lb/>lowed by
			lysis in cold c-Myc antibody lysis buffer containing protease and phos-<lb/>phatase
			inhibitors as previously described <ref type="biblio">10</ref> . c-Myc proteins
			were<lb/> immunoprecipitated from equal numbers of cells for each sample at a
			cell-to-<lb/>volume ratio of 1 × 10 6 cells ml −1 lysis buffer using a 1:100 dilution of
			the c-Myc<lb/> monoclonal antibody C-33 (Santa Cruz Biotechnology, Santa Cruz, CA).<lb/>
			Antibody incubation was performed at 4 °C for 14–18 h followed by
			immuno-<lb/>precipitation with protein A/protein G–agarose beads and washing with
			lysis<lb/> buffer containing protease and phosphatase inhibitors.<lb/></p>

		<p>Pulse-chase assays. MEFs or REFs were pre-starved for methionine and cysteine<lb/> by
			replacing the culture medium with DMEM lacking L-Methionine and L -<lb/>Cysteine for 15
			min. Cells were labelled in vivo with <ref type="biblio">35</ref>
			S-methionine/cysteine<lb/> EXPRESS protein labelling mix from Perkin Elmer (Boston, MA)
			using 500 µCi<lb/> ml −1 at a cell-to-volume ratio of 7.5 × 10 5 cells ml −1 for 20–30
			min. After<lb/> labelling, cells were immediately washed once with DMEM containing 5
			mM<lb/> L-methionine, 3 mM L-cysteine and 0.2% serum, and then incubated in the<lb/>
			same media for the indicated chase times. Cells were harvested and labelled<lb/> c-Myc
			proteins were immunoprecipitated as described above. Labelled c-Myc<lb/> was visualized
			by autoradiography and quantified with a phosphorimager.<lb/></p>

		<p>Myc–GST–Pin1 interaction assays. GST and GST–Pin1 fusion proteins with<lb/> either
			wild-type Xenopus laevis Pin1, a X. laevis Pin1 WW-domain mutant with<lb/> substitutions
			at W11A and W34A, or a X. laevis Pin1 catalytic-deficient mutant<lb/> with a
			substitution at C109A were prepared as described <ref type="biblio">30</ref> . The GST
			and<lb/> GST–Pin1 recombinant proteins were bound to glutathione–agarose beads as<lb/>
			follows: 10 µg of recombinant protein were incubated with 25 µl of a 50% slurry<lb/> of
			glutathione–agarose beads in 500 µl of a low-stringency buffer containing<lb/> 1× PBS,
			0.5% NP40 and 5% glycerol with standard protease and phosphatase<lb/></p>

		<figure>a<lb/> b<lb/> 400<lb/> 450<lb/> 400<lb/> 350<lb/> 300<lb/> 250<lb/> 200<lb/>
			150<lb/> 100<lb/> 50<lb/> 0<lb/> 0<lb/> 50<lb/> 100<lb/> 150<lb/> 200<lb/> 250<lb/>
			300<lb/> 350<lb/> t-Ag<lb/> c-Myc<lb/> T58A<lb/> S62A<lb/> Control<lb/> t-Ag<lb/>
			T58A<lb/> HEK-TER cells<lb/> BJ-TER cells<lb/> 3<lb/> 2.5<lb/> 2<lb/> 1.5<lb/> 1<lb/>
			0.5<lb/> 0<lb/> 0<lb/> 3 0<lb/> 25<lb/> 20<lb/> 15<lb/> 10<lb/> 5<lb/> 3 5<lb/> Time
			(days)<lb/> Cell type<lb/> Tumour number<lb/> t-Ag<lb/> c-Myc WT<lb/> c-Myc T58A<lb/>
			c-Myc S62A<lb/> 6/6<lb/> 0/6<lb/> 6/6<lb/> 0/6<lb/> Tumour growth (cm<lb/> 3 )<lb/>
			Colony number<lb/> Colony number<lb/> Figure 8 Stabilized c-Myc cooperates with Ras and
			telomerase to transform<lb/> human fibroblasts. (a) c-Myc T58A has transforming
			properties. Anchorage-<lb/>independent growth of the indicated cells stably infected
			with retroviruses<lb/> encoding small T antigen, c-Myc, c-Myc S62A or c-Myc T58A ,
			calculated from<lb/> the average number of colonies observed in three plates. (b) c-Myc
			T58A<lb/> replaces small T antigen function in tumorigenesis assays. Mean ±
			standard<lb/> error of tumour volumes (cm 3 ) from six mice injected with HEK cells<lb/>
			expressing hTERT, T-ag, Ras G12V and either small T antigen, c-Myc, c-<lb/>Myc S62A or
			c-Myc T58A . n = 6 for each injection.<lb/></figure>


		<p>inhibitors <ref type="biblio">10</ref> . Binding was performed at 4 °C for 1.5 h. GST
			beads were then<lb/> washed four times in 600 µl of low-stringency buffer. REF52 cells
			infected with<lb/> Ad-c-Myc were lysed in low-stringency buffer at a cell-to-volume
			ratio of<lb/> 5 × 10 5 cells ml −1 . 500 µl of cell lysate containing c-Myc was then
			incubated with<lb/> the various GST–Pin1 beads for 4 h at 4 °C. Samples were then washed
			three<lb/> times in 600 µl of low-stringency buffer and precipitated proteins were
			sub-<lb/>jected to western blot analysis.<lb/></p>

		<p>Co-precipitation of Myc and Pin1. 293 cells were transfected with expression<lb/>
			plasmids for wild-type or mutant His 6 -tagged c-Myc proteins, Pin1 and<lb/>
			β-galactosidase. Cells were lysed in co-immunoprecipitation buffer (50mM<lb/> Hepes at
			pH 7.2, 125 mM potassium acetate, 0.5 mM EDTA, 0.5 mM EGTA,<lb/> 0.1% Tween-20 and 12.5%
			glycerol) containing protease and phosphatase<lb/> inhibitors. Extracts were normalized
			for transfection efficiency by β-galactosi-<lb/>dase assay and incubated with
			nickel–NTA–agarose beads (Qiagen, Valencia,<lb/> CA) for 3–4 h at 4 °C followed by three
			washes in co-immunoprecipitation<lb/> buffer containing protease and phosphatase
			inhibitors and 20 mM imidazole.<lb/> Precipitated proteins were released with
			co-immunoprecipitation buffer con-<lb/>taining 250 mM imidazole and then analysed by
			western blotting.<lb/></p>

		<p>Primary wild-type and Pin1 −/− MEFs. Pin1 −/− mice were generated as previ-<lb/>ously
			described <ref type="biblio">35</ref> . The Pin1 gene deletion was transferred into an
			isogenic<lb/> C57Bl6 background using marker-assisted speed congenic breeding by
			Jackson<lb/> Laboratory (Bar Harbor, ME). Pin1 +/+ and Pin1 −/− embryonic fibroblasts
			were<lb/> isolated from the isogenic strain as detailed previously 36 and processed
			as<lb/> described above.<lb/></p>

		<p>Assay of endogenous c-Myc. Asynchronous or quiescent REF52 cells were<lb/> infected with
			Ad-small T antigen or control Ad-GFP, and endogenous c-Myc was<lb/> immunoprecipitated
			as described under Immunoprecipitations, except that cells<lb/> were lysed in antibody
			lysis buffer at 2 × 10 6 cells ml −1 and precipitates were not<lb/> washed, but loaded
			directly onto 8% gels and fractionated by SDS–PAGE.<lb/> Endogenous c-Myc from primary
			MEFs was assayed after cells were density<lb/> growth-arrested in DMEM containing 15%
			heat-inactivated FCS for 24 h after<lb/> they reached confluence, before&amp; plating at
			~4,000 cells cm −2 in DMEM con-<lb/>taining 0.2% heat-inactivated FCS. After 24 h, cells
			were serum-stimulated by<lb/> adding FCS to 20% volume. Samples were then scraped into
			hot SDS sample<lb/> buffer at the indicated times and analysed by western blotting as
			described above.<lb/></p>

		<p>Transfections. REF52 fibroblasts were transfected using calcium phosphate
			co-<lb/>precipitation, as previously described <ref type="biblio">24</ref> . Cells were
			plated the day before trans-<lb/>fection in 100-mm tissue culture dishes at ~3,500cells
			cm −2 . Cells were exposed<lb/> to the CaPO 4 –DNA precipitate containing 10–20 µg of
			total DNA for 18–20 h.<lb/> Transfected cells were washed once with DMEM and placed into
			starve media<lb/> (DMEM containing 0.25% FCS) for 48 h. Luciferase activity was
			measured<lb/> using a Promega (Madison, WI) luciferase assay system kit, as specified by
			the<lb/> manufacturer, and a Berthold luminometer (Bundoora, Australia).
			β-galactosi-<lb/>dase activity was measured as described previously <ref type="biblio"
				>24</ref> . 293a cells were plated at<lb/> ~5,000 cells cm −2 into tissue culture
			dishes and incubated overnight. Cells at<lb/> 60–80% confluency were then transfected
			with Perfectin (Gene Therapy<lb/> Systems, San Diego, CA) or TransIT-TKO (Mirus Co.,
			Madison, WI) at a 4:1<lb/> ratio of lipid to DNA, in low-serum medium (2% FCS) according
			to the manu-<lb/>facturer&apos;s specifications. Transfected cells were maintained in
			DMEM contain-<lb/>ing 2% FCS and harvested 48 h after transfection.<lb/></p>

		<p>RNAi experiments. 293 cells were transfected with expression vectors for c-Myc,<lb/>
			β-galactosidase, and Luciferase, along with either pooled siRNAs directed<lb/> against
			the PP2A C subunit (Dharmacon, Lafayette, CO) or control siRNAs<lb/> including a
			scrambled siRNA or siRNA directed against luciferase from the<lb/> siSTARTER Luciferase
			Kit (Dharmacon) using TransIT-TKO (Mirus Co.). Cells<lb/> were collected 48 h after
			transfection and analysed for β-galactosidase activity as<lb/> previously described.
			Both luciferase activity and protein load volumes for west-<lb/>ern blot analysis were
			adjusted for transfection efficiency on the basis of β-galac-<lb/>tosidase
			activity.<lb/></p>

		<p>Transformation and tumorigenesis assays. The retroviral constructs containing<lb/> c-Myc,
			c-Myc T58A , and c-Myc S62A were created by PCR amplification of cDNA<lb/> from the
			pRC-CMV vector 37 using T7 and SP6 universal primers with BamHI<lb/> and EcoRI sites
			engineered onto the ends. The DNAs were then ligated into the<lb/> pWZL-Blast vector
			digested with the same enzymes to create pWZL-Blast<lb/> c-Myc, c-Myc T58A and c-Myc
			S62A . The HEK and BJ cell lines containing hTERT,<lb/> Ras, large T antigen and small T
			antigen in varying combinations were created<lb/> by retroviral transduction of parental
			cells as previously described <ref type="biblio">26</ref> . The subse-<lb/>quent cell
			lines containing the various c-Myc constructs were generated by<lb/> amphotrophic
			retroviral infection and polyclonal selection with 2.5 µg ml blas-<lb/>ticidin S (EMD
			Biosciences, San Diego, CA).<lb/></p>

		<p>The ability of cells to undergo anchorage-independent growth was tested as<lb/>
			previously described <ref type="biblio">38</ref> . Briefly, 5 × 10 4 cells were plated
			in 0.3% agar for 12 days<lb/> (HEK cells) or 21 days (BJ cells) and assessed for colony
			growth in soft agar. The<lb/> mean and standard deviation represents two independent
			experiments performed<lb/> in triplicate. Tumour formation in immunodeficient mice was
			determined by<lb/> sub-cutaneous injection of 1 × 10 7 cells per animal and was
			represented as the<lb/> number of tumours/number of animals (1 injection site per
			animal).<lb/></p>

	</text>
</tei>