11
Matters Arising IGFBP7 Is Not Required for B-RAF-Induced Melanocyte Senescence Lyndee L. Scurr, 1, * Gulietta M. Pupo, 1 Therese M. Becker, 1 Ken Lai, 2,4 David Schrama, 3 Sebastian Haferkamp, 1 Mal Irvine, 1 Richard A. Scolyer, 2,4,5 Graham J. Mann, 1 Ju ¨ rgen C. Becker, 3 Richard F. Kefford, 1,2 and Helen Rizos 1, * 1 Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New South Wales 2145, Australia 2 Melanoma Institute Australia (incorporating the Sydney Melanoma Unit), Sydney, New South Wales 2050, Australia 3 Department of Dermatology, University Hospital Wu ¨ rzburg, Wu ¨ rzburg D-97080, Germany 4 Department of Anatomical Pathology, Royal Prince Alfred Hospital, Camperdown, Sydney, New South Wales 2050, Australia 5 Discipline of Pathology, Faculty of Medicine, University of Sydney, Sydney, New South Wales 2006, Australia *Correspondence: [email protected] (L.L.S.), [email protected] (H.R.) DOI 10.1016/j.cell.2010.04.021 SUMMARY Induction of senescence permanently restricts cellular proliferation after oncogenic stimulation thereby acting as a potent barrier to tumor develop- ment. The relevant effector proteins may therefore be fundamental to cancer development. A recent study identified IGFBP7 as a secreted factor mediating melanocyte senescence induced by oncogenic B-RAF, which is found commonly in cutaneous nevi. In contrast to the previous report, we demon- strate that B-RAF signaling does not induce IGFBP7 expression, nor the expression of the IGFBP7 targets, BNIP3L, SMARCB1, or PEA15, in human melanocytes or fibroblasts. We also found no corre- lation between B-RAF mutational status and IGFBP7 protein expression levels in 22 melanoma cell lines, 90 melanomas, and 46 benign nevi. Furthermore, using a lentiviral silencing strategy we show that B-RAF induces senescence in melanocytes and fibroblasts, irrespective of the presence of IGFBP7. Therefore, we conclude that the secreted protein IGFBP7 is dispensable for B-RAF V600E -induced senescence in human melanocytes. INTRODUCTION Cutaneous melanoma is an aggressive form of cancer that is highly resistant to conventional therapies with fewer than 10% of patients with visceral metastases surviving 2 years (reviewed in Thompson et al., 2005; Herlyn, 2007). It may arise de novo from melanocytes or their precursors in the skin (Kelly et al., 1997) or from common benign clonal expansions of cutaneous melanocytes, known as nevi (Chin et al., 2006; reviewed in Bennett, 2008). Although the presence of high numbers of nevi is strongly associated with melanoma risk, the majority of benign nevi lack proliferative activity and never progress into malig- nancy despite harboring oncogenic alterations. Indeed, consti- tutively activating mutations affecting the N-RAS or B-RAF kinase components of the mitogen-activated protein kinase (MAPK) pathway are found in over 80% of benign nevi (Bauer et al., 2007; Papp et al., 1999; Pollock et al., 2003). The prevailing view is that nevi have initiated a permanent form of proliferative arrest known as oncogene-induced senescence (reviewed in Prieur and Peeper, 2008). Certainly, benign nevi display several markers of senescence growth arrest, including increased senescence-associated b-galactosidase (SA-b-Gal) activity and p16 INK4a expression (Gray-Schopfer et al., 2006; Michaloglou et al., 2005). Although the presence of senescent cells in human melanocytic nevi remains controversial (Cotter et al., 2007), accumulating evidence suggests that senescence occurs in vivo and acts as an effective barrier to tumor formation (reviewed in Prieur and Peeper, 2008). In this regard, identifica- tion of the molecules that enforce oncogene-induced arrest in human melanocytes is of prime importance as their loss may be a critical step in the escape of malignant clones and the consequent development of melanoma. These molecules are also potential therapeutic targets, as reinstating their functions may stimulate entry into senescence. In an attempt to identify these molecules, a genome-wide loss-of-function screen was used to define essential regulators of B-RAF V600E -induced senescence (Wajapeyee et al., 2008). Notably, this oncogenic form of B-RAF occurs in 40%–50% of melanomas (reviewed in Platz et al., 2008) and its expression is sufficient to induce senescence and apoptosis in human diploid fibroblasts and melanocytes (Gray-Schopfer et al., 2006; Michaloglou et al., 2005; Wajapeyee et al., 2008). Seventeen genes were identified and the suppression of each permitted diploid human fibroblasts to proliferate and survive in the pres- ence of oncogenic B-RAF V600E . Among these were p53 and the insulin-like growth factor (IGF) binding protein 7 (IGFBP7). IGFBP7 is a secreted protein that reduces the bioavailability of insulin/IGF in peripheral blood and has been implicated in tumor suppression of breast, colon, lung, and pancreatic cancers (Burger et al., 1998; Chen et al., 2007; Landberg et al., 2001; Ruan et al., 2007; Swisshelm et al., 1995; Ye et al., 2007). B-RAF V600E was also shown to upregulate IGFBP7 expression via the B-RAF-MEK-ERK signaling cascade. IGFBP7 inhibited Cell 141, 717–727, May 14, 2010 ª2010 Elsevier Inc. 717

IGFBP7 Is Not Required for B-RAF-Induced Melanocyte Senescence

  • Upload
    mq

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Matters Arising

IGFBP7 Is Not Required forB-RAF-Induced Melanocyte SenescenceLyndee L. Scurr,1,* Gulietta M. Pupo,1 Therese M. Becker,1 Ken Lai,2,4 David Schrama,3 Sebastian Haferkamp,1

Mal Irvine,1 Richard A. Scolyer,2,4,5 Graham J. Mann,1 Jurgen C. Becker,3 Richard F. Kefford,1,2 and Helen Rizos1,*1Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New

South Wales 2145, Australia2Melanoma Institute Australia (incorporating the Sydney Melanoma Unit), Sydney, New South Wales 2050, Australia3Department of Dermatology, University Hospital Wurzburg, Wurzburg D-97080, Germany4Department of Anatomical Pathology, Royal Prince Alfred Hospital, Camperdown, Sydney, New South Wales 2050, Australia5Discipline of Pathology, Faculty of Medicine, University of Sydney, Sydney, New South Wales 2006, Australia

*Correspondence: [email protected] (L.L.S.), [email protected] (H.R.)DOI 10.1016/j.cell.2010.04.021

SUMMARY

Induction of senescence permanently restrictscellular proliferation after oncogenic stimulationthereby acting as a potent barrier to tumor develop-ment. The relevant effector proteins may therefore befundamental to cancer development. A recent studyidentified IGFBP7 as a secreted factor mediatingmelanocyte senescence induced by oncogenicB-RAF, which is found commonly in cutaneousnevi. In contrast to the previous report, we demon-strate that B-RAF signaling does not induce IGFBP7expression, nor the expression of the IGFBP7targets, BNIP3L, SMARCB1, or PEA15, in humanmelanocytes or fibroblasts. We also found no corre-lation between B-RAF mutational status and IGFBP7protein expression levels in 22 melanoma cell lines,90 melanomas, and 46 benign nevi. Furthermore,using a lentiviral silencing strategy we show thatB-RAF induces senescence in melanocytes andfibroblasts, irrespective of the presence of IGFBP7.Therefore, we conclude that the secreted proteinIGFBP7 is dispensable for B-RAFV600E-inducedsenescence in human melanocytes.

INTRODUCTION

Cutaneous melanoma is an aggressive form of cancer that is

highly resistant to conventional therapies with fewer than 10%

of patients with visceral metastases surviving 2 years (reviewed

in Thompson et al., 2005; Herlyn, 2007). It may arise de novo

from melanocytes or their precursors in the skin (Kelly et al.,

1997) or from common benign clonal expansions of cutaneous

melanocytes, known as nevi (Chin et al., 2006; reviewed in

Bennett, 2008). Although the presence of high numbers of nevi

is strongly associated with melanoma risk, the majority of benign

nevi lack proliferative activity and never progress into malig-

nancy despite harboring oncogenic alterations. Indeed, consti-

tutively activating mutations affecting the N-RAS or B-RAF

kinase components of the mitogen-activated protein kinase

(MAPK) pathway are found in over 80% of benign nevi (Bauer

et al., 2007; Papp et al., 1999; Pollock et al., 2003). The prevailing

view is that nevi have initiated a permanent form of proliferative

arrest known as oncogene-induced senescence (reviewed in

Prieur and Peeper, 2008). Certainly, benign nevi display several

markers of senescence growth arrest, including increased

senescence-associated b-galactosidase (SA-b-Gal) activity

and p16INK4a expression (Gray-Schopfer et al., 2006;

Michaloglou et al., 2005). Although the presence of senescent

cells in human melanocytic nevi remains controversial (Cotter

et al., 2007), accumulating evidence suggests that senescence

occurs in vivo and acts as an effective barrier to tumor formation

(reviewed in Prieur and Peeper, 2008). In this regard, identifica-

tion of the molecules that enforce oncogene-induced arrest in

human melanocytes is of prime importance as their loss may

be a critical step in the escape of malignant clones and the

consequent development of melanoma. These molecules are

also potential therapeutic targets, as reinstating their functions

may stimulate entry into senescence.

In an attempt to identify these molecules, a genome-wide

loss-of-function screen was used to define essential regulators

of B-RAFV600E-induced senescence (Wajapeyee et al., 2008).

Notably, this oncogenic form of B-RAF occurs in 40%–50% of

melanomas (reviewed in Platz et al., 2008) and its expression is

sufficient to induce senescence and apoptosis in human diploid

fibroblasts and melanocytes (Gray-Schopfer et al., 2006;

Michaloglou et al., 2005; Wajapeyee et al., 2008). Seventeen

genes were identified and the suppression of each permitted

diploid human fibroblasts to proliferate and survive in the pres-

ence of oncogenic B-RAFV600E. Among these were p53 and

the insulin-like growth factor (IGF) binding protein 7 (IGFBP7).

IGFBP7 is a secreted protein that reduces the bioavailability of

insulin/IGF in peripheral blood and has been implicated in tumor

suppression of breast, colon, lung, and pancreatic cancers

(Burger et al., 1998; Chen et al., 2007; Landberg et al., 2001;

Ruan et al., 2007; Swisshelm et al., 1995; Ye et al., 2007).

B-RAFV600E was also shown to upregulate IGFBP7 expression

via the B-RAF-MEK-ERK signaling cascade. IGFBP7 inhibited

Cell 141, 717–727, May 14, 2010 ª2010 Elsevier Inc. 717

Figure 1. Senescence Program Induced by Oncogenic B-RAFV600E in Human Melanocytes and Fibroblasts

(A) Human epidermal melanocytes 1455 (HEM) and human dermal fibroblasts 1314 (HDF) were infected with lentiviruses encoding B-RAFV600E with copGFP (+) or

copGFP alone (�) and the expression levels of total and phosphorylated ERK (p-ERK) were compared to those observed in the NM39 melanoma cell line, which is

heterozygous for B-RAFV600E. All lanes in this analysis were derived from a single gel and western blot. Ectopically expressed B-RAFV600E was detected using an

antibody against the MYC epitope and endogenous B-RAF in the NM39 cells was detected with the antibody against B-RAF.

(B) Human melanocytes and fibroblasts were infected with lentiviruses encoding wild-type B-RAF, B-RAFV600E, or copGFP. The efficiency of transduction was

controlled with the coexpression of copGFP and was consistently above 90%. Cell proliferation (Ki67), chromatin condensation (DAPI), and the appearance of

increased SA-b-Gal activity were analyzed and quantitated 5 and 10 days after infection of melanocytes and fibroblasts, respectively. Cells enlarged to show

DAPI-stained chromatin foci are indicated with arrows. Percentage of cells positive for the indicated marker is shown in histograms, which correspond to the

mean ± standard deviation (SD) of at least two independent transduction experiments from a total of at least 300 cells.

718 Cell 141, 717–727, May 14, 2010 ª2010 Elsevier Inc.

phosphorylation of MEK by B-RAF, initiating a negative-feed-

back loop. Consistent with the hypothesis that IGFBP7

suppresses early B-RAF-driven tumor formations in vivo,

expression of IGFBP7 was highly expressed in B-RAFV600E-posi-

tive nevi and in primary melanomas and melanoma cell lines

lacking activated B-RAF but was lost in B-RAFV600E-positive

melanomas and melanoma cell lines. Furthermore, purified re-

combinant IGFBP7 potently suppressed the growth of

B-RAFV600E-positive human melanomas but had no effect on

tumors with wild-type B-RAF (Wajapeyee et al., 2008). Taken

together these observations suggest that IGFBP7 is essential

in constraining B-RAFV600E-stimulated cell proliferation.

In this work we readdressed the role of IGFBP7 in B-RAFV600E-

induced senescence in diploid human melanocytes. This was

particularly important as two recent reports did not detect

a correlation between B-RAF mutation status and IGFBP7

expression in a panel of primary melanomas and metastases

(Schrama et al., 2009; Wajapeyee et al., 2009). We also found

no correlation between B-RAF mutational status and IGFBP7

expression levels in a series of melanoma cell lines, melanomas,

and benign nevi. Furthermore, in contrast to the previous report

(Wajapeyee et al., 2008), we demonstrate that B-RAF signaling

does not induce IGFBP7 expression in either human melano-

cytes or fibroblasts, and using a lentiviral silencing strategy we

show that B-RAF potently induces senescence in the presence

and absence of IGFBP7. Therefore, we conclude that the

secreted protein IGFBP7 is dispensable for B-RAFV600E-induced

senescence in human melanocytes.

RESULTS

Expression of Oncogenic B-RAF Induces CellularSenescence that Is Associated with IGFBP7 RepressionTo evaluate the response of human cells to oncogenic B-RAF,

the melanoma-associated B-RAFV600E mutant was stably trans-

duced into primary diploid HEM1455 melanocytes and HDF1314

epidermal fibroblasts using lentiviral vectors coexpressing

Copepod GFP (copGFP). Viral titers were selected to provide

an efficiency of infection above 90% and activation of the ERK

pathway that was comparable to human melanoma cells ex-

pressing endogenous B-RAFV600E (Figure 1A).

As expected from previous studies, expression of oncogenic

B-RAF efficiently blocked cellular proliferation and induced SA-

b-Gal activity as early as 5 days after infection (Figure 1B). The

majority of B-RAFV600E-transduced melanocytes and fibroblasts

displayed several markers of oncogene-driven senescence,

increased SA-b-Gal expression, senescence-associated hetero-

chromatin foci (SAHF), and significantly decreased Ki67 expres-

sion (Figure 1B). Each focus in a senescent cell results from

condensation of an individual chromosome and is enriched for

common markers of heterochromatin, including histone H3

methylated at lysine 9 (H3K9Me) and the nonhistone chromatin

(C) Representative examples of chromatin condensation (DAPI) in human epider

HMGA2. Cells showing DAPI-stained chromatin foci are indicated with arrows.

(D) Expression of the indicated proteins was determined by western blot analys

copGFP, wild-type B-RAF, or B-RAFV600E.

protein HMGA2 (Figure 1C) (reviewed in Adams, 2007). The

accumulation of oncogenic B-RAFV600E in both melanocytes

and fibroblasts was associated with increased levels of phos-

phorylated ERK and induction of the CDK inhibitor p16INK4a. In

addition, the levels of p53 were significantly repressed and

expression of the p53-transcription target p21Waf1 remained

low or was reduced by the accumulation of B-RAFV600E

(Figure 1D).

It has been suggested that senescence triggered by onco-

genic B-RAF is associated with the transcriptional upregulation

of IGFBP7 (Wajapeyee et al., 2008). We therefore reanalyzed

the expression of IGFBP7 and conclusively show that oncogenic

B-RAF represses, rather than induces, IGFBP7 expression in

both human melanocytes and fibroblasts (Figure 2A and

Figure S1 available online). Indeed, B-RAFV600E-transduced

melanocytes and fibroblasts displayed diminished levels of

cellular and secreted IGFBP7 compared to the control

copGFP-transduced cells (Figure 2A and Figure S1). The deple-

tion of IGFBP7 was routinely observed as early as 2 days post-B-

RAFV600E transduction and was maintained for the duration of

the transduction experiments (5 days and 15 days post-trans-

duction of melanocytes and fibroblasts, respectively). The slight

increase in secreted IGFBP7 in melanocytes, 2 days post-trans-

duction with oncogenic B-RAFV600E, was not consistent over

replicate experiments and did not correspond with an increase

in cellular IGFBP7 at this time point (Figure 2A). The depletion

of IGFBP7 occurred earlier than the onset of BRAFV600E-induced

senescence (see Figure 1B). Furthermore, the specific silencing

of oncogenic B-RAF or the addition of a MEK inhibitor led to the

accumulation of IGFBP7 in melanoma cell lines expressing the

B-RAFV600E variant (Figure 2B).

B-RAF Mutation Status Does Not Correlate with IGFBP7ExpressionTo further scrutinize the correlation of IGFBP7 expression with

B-RAF mutation status, we analyzed a series of 22 melanoma

cell lines for IGFBP7 expression. In this series, we did not find

a correlation between IGFBP7 expression and B-RAF mutation

status, as IGFBP7 was detected in 10/14 B-RAF mutant and

3/8 B-RAF wild-type tumor lines (Figure 2C). Further, we found

no correlation between B-RAF mutation status and IGFBP7 tran-

script levels in gene expression microarray analyses of 99 meta-

static melanoma tumors (Figure 3A, Table 1). This was confirmed

at the protein level by tissue microarray analyses of 90 of these

tumors (Figure 3B, Table 2). Consistent with these data, we did

not observe increased expression of several IGFBP7-regulated

gene products, including SMARCB1, BNIP3L, and PEA15

(Wajapeyee et al., 2008), in B-RAFV600E-transduced fibroblasts

or melanocytes (Figure 3C), nor did the expression of these

IGFBP7-regulated transcripts correlate with B-RAF mutation

status in our microarray analyses of 99 metastatic melanomas

(Figure 3C, Table 1). Finally, IGFBP7 protein expression did not

mal melanocytes expressing B-RAFV600E (day 5) and staining for H3K9Me and

is after infection of melanocytes and fibroblasts with lentiviruses expressing

Cell 141, 717–727, May 14, 2010 ª2010 Elsevier Inc. 719

Figure 2. Oncogenic B-RAFV600E Expres-

sion Does Not Correlate with IGFBP7 Accu-

mulation

(A) Immunoblot analysis monitoring the accumula-

tion of IGFBP7 in human melanocytes and

fibroblasts infected with copGFP control lentivirus

(�) or lentivirus expressing B-RAFV600E (+). Protein

expression was analyzed at 2, 3, 5, 10, and 15

days post-transduction (PT), as shown (see also

Figure S1).

(B) Immunoblot analysis monitoring cellular

IGFBP7 expression in melanoma cell lines 5 days

post-transduction with control (�) or BRAFV600E-

specific (+) shRNA molecules, as indicated. This

figure is compiled from duplicate immunoblots.

Cells were also treated with the MEK inhibitor

PD98059 for 72 hr, as shown. The WMM1175

melanoma cells carry wild-type B-RAF, and the

ME1042 and NM39 melanoma cells are heterozy-

gous for B-RAFV600E.

(C) Immunoblot analysis monitoring cellular

IGFBP7 levels in a panel of human melanoma cell

lines, primary human melanocytes (HEM1455),

three fibroblasts (HDF1314, BJ, and WS1), and

three other cancer cell lines. The mutation status

of B-RAF and N-RAS is indicated.

correlate with B-RAF mutation status in a series of human benign

nevi (Figure 3D, Table 2). Collectively these data indicate that

B-RAFV600E does not upregulate IGFBP7 expression in fibro-

blasts or melanocytes nor is the loss of IGFBP7 required for

the development of B-RAFV600E-positive melanomas.

IGFBP7 Is Not Required for B-RAFV600E-InducedSenescenceTo specifically examine the contribution of IGFBP7 to B-RAF-

mediated senescence, we applied lentiviral shRNA vectors that

specifically suppress the expression of IGFBP7. To minimize

confounding effects of shRNA off-target silencing two indepen-

dent silencing molecules targeting IGFBP7 were generated

(Wajapeyee et al., 2008). These silencing constructs were highly

effective in silencing the expression of IGFBP7. The role of

720 Cell 141, 717–727, May 14, 2010 ª2010 Elsevier Inc.

IGFBP7 in oncogene-induced senes-

cence was evaluated in human melano-

cytes (HEM1455, nHEM) and fibroblasts

(HDF1314, WS1, and BJ), and the results

for HDF1314 and HEM1455 are depicted

in detail. Cells were transduced with an

IGFBP7 shRNA molecule coexpressing

copGFP and 3 days post-infection were

retransduced with lentiviral vectors

expressing B-RAFV600E with copGFP

or copGFP alone. All experiments

included a negative control shRNA mole-

cule without homology to any human

gene. Just as in our previous single trans-

duction experiments, transduction

efficiencies reached at least 90% in all

cells (Figure S2A) and there was no

need for antibiotic selection. This strategy proved effective as

the depletion of pRb along with p53 using highly effective

short-hairpin RNAs (Haferkamp et al., 2009a) overcame

B-RAFV600E-induced melanocyte cell-cycle arrest (Figure S2B).

As expected, all melanocytes and fibroblasts transduced with

the combination of control shRNA and B-RAFV600E expressed

markers associated with the onset of senescence. These

included significantly reduced levels of the proliferation marker

Ki67, accumulation of SAHF, and increased SA-b-Gal activity

(Figures 4A and 4B). Importantly, inhibition of IGFBP7 expression

did not alter the growth arrest induced by oncogenic

B-RAFV600E, and cells were arrested after infection, stained posi-

tive for SA-b-Gal, and accumulated DAPI-stained foci regardless

of the expression level of IGFBP7 (Figures 4A and 4B). In addi-

tion, the induction of p16INK4a associated with oncogenic

Figure 3. Oncogenic B-RAFV600E Expression Does Not Correlate with IGFBP7 Accumulation in Melanoma Tumors or Benign Nevi

(A) The relationship between B-RAF mutation status and IGFBP7 gene expression levels was determined using microarray analyses of 99 metastatic melanoma

tumors. The horizontal bars indicate the median expression values.

(B) Tissue microarray results of IGFBP7 staining in 90 metastatic melanomas. Scatter plot analysis showing the percentage of cells positive for IGFBP7 in B-RAF

mutant and B-RAF wild-type melanomas. The median expression value was zero for both sets.

(C) The relationship between B-RAF mutation status and the expression of the IGFBP7-regulated transcripts BNIP3L, SMARCB1, and PEA15 was analyzed. Left

panel: Immunoblot analysis monitoring the accumulation of BNIP3L, SMARCB1, and PEA15 in human fibroblasts and melanocytes post-infection with copGFP

control lentivirus (�) or lentivirus expressing B-RAFV600E (+), respectively. Protein expression was analyzed at 5, 10, and 15 days post-infection of fibroblasts

(representative examples at 5 days post-infection of melanocytes and 15 days post-infection of fibroblasts are shown). Right panels: The relationship between

B-RAF mutation status and BNIP3L, SMARCB1, and PEA15 gene expression levels was determined using microarray analyses of 99 metastatic melanoma

tumors. The horizontal bars indicate the median expression values.

(D) The relationship between B-RAF mutation status and IGFBP7 staining was determined using immunohistochemistry analysis of 44 benign human nevi. The

horizontal bar indicates the median expression values (left panel). Representative examples of nevi carrying wild-type B-RAF or B-RAFV600E mutations demon-

strating low IGFBP7 expression (left side panels) or IGFBP7 expression (right side panels) are shown. Scale bar: 50 mM.

B-RAF expression was also maintained in melanocytes and

fibroblasts regardless of IGFBP7 expression (Figure 5).

Similar results were obtained when IGFBP7-silenced fibro-

blasts (data not shown) and melanocytes were selected using

puromycin prior to reinfection with oncogenic B-RAFV600E

(Figure S2C). This antibiotic selection strategy resembled

the experimental design employed by Wajapeyee et al.

(2008) and confirmed that IGFBP7 was not essential for

Cell 141, 717–727, May 14, 2010 ª2010 Elsevier Inc. 721

Table 1. Median and Interquartile Ranges of Transcript Expression by B-RAF Status in Metastatic Melanomas Together with

Associated p Values (Mann-Whitney Test)

B-RAF Status

Wild-Type Mutant

Transcripts Median (LQ, UQ) Median (LQ, UQ) p Value

BNIP3L 818 402, 1556 768 262, 1447 0.591

IGFBP7 7461 5057, 14312 8290 4978, 12647 0.886

PEA15 7423 5369, 9105 6125 4057, 9973 0.129

SMARCB1 1070 667, 2032 772 562, 1528 0.109

TP53 301 181, 586 219 135, 520 0.124

UQ, upper quartile; LQ, lower quartile.

B-RAFV600E-induced senescence as its depletion did not prevent

cell-cycle arrest, formation of DAPI-stained foci, or the induction

of SA-b-Gal activity (Figure S2C).

DISCUSSION

The detection of senescent cells in vivo suggests that senes-

cence is a genuine, physiological tumor suppressor mechanism,

and this notion has intensified the search for the molecules that

initiate and maintain this proliferative arrest.

In a landmark study, a genome-wide shRNA screen identified

17 genes that were essential for B-RAFV600E-induced senes-

cence in human fibroblasts and melanocytes. Two of these

essential candidates were p53 and the secreted protein IGFBP7

(Wajapeyee et al., 2008).

We and others have shown that p53 levels are not induced by

oncogenic B-RAF in human melanocytes (Gray-Schopfer et al.,

2006; Zhuang et al., 2008) or in senescent normal melanocytes

(Sviderskaya et al., 2003). Furthermore, several reports have

confirmed that inactivation of p53 alone does not prevent B-

RAF-induced senescence in melanocytes (Denoyelle et al.,

2006; Zhuang et al., 2008). In keeping with these findings,

immortal melanocytes accumulating oncogenic B-RAF and

silenced for p53 expression did arise in one investigation.

However, these immortal cells had acquired a substantial dele-

tion in chromosome 13, which encodes the pRb1 tumor

suppressor gene (Yu et al., 2009). Indeed, loss of both pRb

and p53 is sufficient to overcome both N-RASQ61K- (Haferkamp

et al., 2009a) and B-RAFV600E-induced (Figure S2B) senescence

in melanocytes. Collectively these data strongly indicate that p53

is not required for B-RAF-induced senescence in melanocytes

in vitro and in vivo.

Table 2. Median and Interquartile Ranges of IGFBP7 Protein Expres

Together with Associated p Values (Mann-Whitney Test)

B-RAF status

Wild type

IGFBP7 Protein Median (LQ, UQ)

Melanoma 0 0, 38

Nevi 70 40, 100

UQ, upper quartile; LQ, lower quartile.

722 Cell 141, 717–727, May 14, 2010 ª2010 Elsevier Inc.

Because IGFBP7 might also have clinical potential, we used

four independent experimental strategies to reanalyze the

reported contribution of IGFBP7 to B-RAF-induced senescence.

First, we examined the regulation of IGFBP7 by oncogenic

B-RAF in several melanocyte and fibroblast cells. We did not

detect any significant upregulation of IGFBP7 in response

to oncogenic B-RAF. In fact we saw significant reduction in

IGFBP7 in all primary cells tested, and a slight IGFBP7 upregula-

tion was only seen in response to silencing of B-RAFV600E

or inhibiting the MAPK pathway using a MEK inhibitor in mela-

noma cell lines harboring oncogenic B-RAFV600E. IGFBP7 is tran-

scriptionally regulated by p53 (Suzuki et al., 2009), and the

depletion of IGFBP7 in response to oncogenic B-RAF may be

due to the transcriptional repression of p53. Second, two inde-

pendent and highly effective IGFBP7 shRNA molecules were

employed to examine the role of IGFBP7 in B-RAF-induced

senescence. We found that B-RAF effectively induced senes-

cence regardless of IGFBP7 expression in the two melanocyte

and three fibroblast strains tested. Third, there was no correla-

tion between IGFBP7 expression and B-RAF mutation status in

a panel of 22 melanoma cell lines. Fourth, immunohistochemical

analysis of human nevi and melanoma samples with known

B-RAF mutation status confirmed that IGFBP7 expression was

not related to B-RAF activation in vivo. The interpretation of the

melanoma tumor data is not straightforward, however, as it

is not possible to rule out the influence of environmental

and genetic risk factors present in the Australian melanoma

patients. The Australian population is generally exposed to

high levels of ultraviolet radiation and develops melanocytic

nevi early in life and in large numbers (Harrison et al., 2008).

There is evidence that interactions among phenotypic, genetic,

and environmental risk factors may influence the mechanisms

sion by B-RAF Status in Metastatic Melanomas or Benign Nevi

Mutant

Median (LQ, UQ) P Value

0 0, 50 0.894

70 40, 100 0.655

Figure 4. IGFBP7 Is Not Required for

B-RAF-Induced Senescence

(A) Melanocytes were infected with lentiviruses

containing the indicated shRNA constructs (see

Figure S2A). Three days post-infection the cells

were reinfected with lentiviruses encoding

B-RAFV600E or copGFP, as shown. Cell prolifera-

tion (Ki67), chromatin condensation (DAPI), and

SA-b-Gal activity were analyzed. Representative

examples at 5 days after B-RAF infection are

shown. Cell counts for each of these markers are

shown as histograms, which correspond to

the mean ± SD of at least two independent trans-

duction experiments from a total of at least 300

cells (see also Figures S2B and S2C).

(B) Fibroblasts were transduced with lentiviruses

containing the indicated shRNA constructs (see

Figure S2A). Three days post-infection the cells

were retransduced with copGFP control lentivirus

(�) or lentiviruses expressing B-RAFV600E or

copGFP, as shown. Cell proliferation (Ki67), chro-

matin condensation (DAPI), and the appearance of

SA-b-Gal activity were analyzed as detailed above

on days 5, 10, and 15 post-second transduction.

Representative examples at 10 days after B-RAF

infection are shown. Cell counts for each of the

markers are shown as histograms, which corre-

spond to the mean ± SD of at least two indepen-

dent transduction experiments from a total of at

least 300 cells.

of melanoma development (Curtin et al., 2006; Falchi et al.,

2009). Thus, the lack of correlation between IGFBP7 expression

and B-RAF mutation status in this cohort may reflect differences

in genetic characteristics and environmental risk factors. Never-

Cell 141, 717–

theless, two recent reports also failed to

detect a correlation between IGFBP7

expression and B-RAF status in mela-

noma metastases (Schrama et al., 2009;

Wajapeyee et al., 2009). From all obser-

vations, we conclude that IGFBP7

expression is not induced by oncogenic

B-RAF. Moreover, IGFBP7 expression

seems dispensable for B-RAF-induced

senescence in human melanocytes,

fibroblasts, and nevi. It is also notable

that although IGFBP7 is thought to act

as a tumor suppressor in some cancers

(Burger et al., 1998; Chen et al., 2007;

Landberg et al., 2001; Ruan et al., 2007;

Swisshelm et al., 1995; Ye et al., 2007),

it is overexpressed in glioblastomas and

gastric, prostate, and colorectal cancers

(Akaogi et al., 1996; Degeorges et al.,

1999; Jiang et al., 2008; Pen et al.,

2007, 2008; Shao et al., 2004; Takeno

et al., 2008), which argues against it

having a purely tumor suppressive role.

Because the primary goal of this study

was to analyze the contribution of

IGFBP7 in B-RAF-mediated senescence, we did not analyze

the effect of recombinant IGFBP7 on primary melanocytes,

especially given that we could not confirm any correlation

between IGFBP7 expression and B-RAF mutation status.

727, May 14, 2010 ª2010 Elsevier Inc. 723

Figure 5. Oncogenic B-RAF Signaling Induces

ERK Activation and p16INK4a Expression and

Downregulates p53 in the Presence and Absence

of IGFBP7 Expression

Expression of the indicated proteins was determined by

western blot analysis after infection of human epidermal

melanocytes and fibroblasts with the indicated shRNA

constructs and either lentiviruses encoding B-RAFV600E

or the copGFP (�) control. Representative examples at

5 and 10 days after B-RAF infection of melanocytes and

fibroblasts, respectively, are shown.

However, although recombinant IGFBP7 induced apoptosis in

BRAF mutation-positive cancer cell lines (Wajapeyee et al.,

2008), it also promoted cell death in a subset of human cancer

cells with wild-type B-RAF that did not respond to the MEK inhib-

itor U0126 (Wajapeyee et al., 2009). These findings suggest that

the impact of recombinant IGFBP7 may be independent of the

MAPK pathway.

The most likely explanation for the disparate results reported

in the previous study by Wajapeyee et al. (2008) is that the tissue

culture conditions required for the loss-of-function screen re-

sulted in the selective outgrowth of antibiotic-resistant cell

clones that had escaped oncogene-induced senescence. Active

oncogenes promote DNA hyper-replication and damage

(Bartkova et al., 2006; Di Micco et al., 2006), and immortal

melanocytic clones harboring oncogenes have been shown to

accumulate additional secondary genetic changes (Sviderskaya

et al., 2003; Yu et al., 2009). Consequently, we have limited the

contribution of additional genetic alterations in our study by

(1) excluding long-term antibiotic selection steps and reducing

the duration of our experiments, (2) analyzing pooled cell

populations rather than cell clones, and (3) repeating our short-

term experiments in several different primary human

cell cultures. In support of this approach, p53-deficient melano-

cytes behaved as expected and responded to oncogenic B-RAF

by inducing p16INK4a (Zhuang et al., 2008), whereas the equiva-

lent antibiotic-selected cell clones generated by Wajapeyee

et al. (2008) showed no induction of p16INK4a. As there is no

evidence that p53 loss interrupts the regulation of p16INK4a

by the MAPK pathway, we are led to speculate that

the p16INK4a gene had been altered in those clones, especially

given that both wild-type and p53 null melanocytes should

respond to oncogenic N-RAS by induction of p16INK4a expres-

sion (Haferkamp et al., 2009a).

In conclusion, despite the use of several independent experi-

mental strategies, we were unable to find evidence to support

a critical role of IGFBP7 in mediating B-RAF-induced senes-

cence in melanocytes, in contrast to the recent report in this

journal. There is little doubt, however, that secreted factors

play a significant role in senescence. Key components of the

Wnt1, transforming growth factor b, plasmin, and interleukin

signaling cascades reinforce senescence and potentially sensi-

tize neighboring cells to proliferative arrest (reviewed in Acosta

724 Cell 141, 717–727, May 14, 2010 ª2010 Elsevier Inc.

et al., 2008; Kuilman and Peeper, 2009). Paradoxically, some

of these secreted factors can also exert protumorigenic effects

depending on the cell type and tumor stage. This has important

implications when considering the therapeutic applicability of

agonists that promote senescence, and it is critical that the

contribution and in vivo interactions of these secreted effectors

are defined.

EXPERIMENTAL PROCEDURES

Specimen Collection

Melanoma patients with available fresh-frozen metastatic lymph node disease

were identified from the Sydney Melanoma Unit Biospecimen Bank/Depart-

ment of Anatomical Pathology, Royal Prince Alfred Hospital. Specimen

collection is from routine surgical resections. All tissue specimens are snap

frozen in liquid nitrogen and stored at �80�C until required for use. Melanoma

tumors were obtained between 1996–2006 under a sample procurement

protocol approved by the Sydney South West Area Health Service institutional

ethics review committee (RPAH Zone) Protocol No. X08-0155 and HREC

Ref. 08/RPAH/262—‘‘Histological and Immunohistological Analysis of Mela-

nocytic Tumours.’’ The histopathologic features of each sample were reviewed

by a pathologist (R.A.S.) to confirm diagnosis and tumor content.

Paraffin0embedded samples from 19 junctional, 10 compound, and 17

congenital nevi were obtained by surgical excision. All nevi have undergone

routine histology for diagnosis.

RNA Extraction and Microarray Gene Expression Analysis

Total RNA was extracted from 10–20 mg of tumor. Tissue samples were

homogenized using a high-speed agitation Polytron blender (Kinematica,

Luzern, Switzerland) in the presence of Trizol (Invitrogen Life Technologies,

Carlsbad, CA, USA). Following homogenization, chloroform was added and

the sample centrifuged. The upper phase was removed and mixed with 70%

ethanol. The RNA was then isolated and purified with an RNeasy purification

kit with DNase I digestion on the column (QIAGEN, Valencia, CA, USA). The

quality of the RNA preparations was assessed using a Agilent 2100 Bioana-

lyser (Agilent Technologies, Palo Alto, CA, USA). The extracted total RNA

was of high quality (RNA integrity number: 8–10). cRNA amplification and

labeling with biotin were performed using the Illumina TotalPrep RNA amplifi-

cation kit (Ambion, Austin, TX, USA) according to the manufacturer’s direc-

tions starting with 250 ng total RNA. Gene expression analysis was performed

using the Sentrix HumanRef-6 v.3.0 Expression BeadChip (Illumina,

San Diego, CA, USA) and BeadStation system from Illumina according to

manufacturer’s instructions.

DNA Extraction and Genotyping

DNA was extracted from a 10–20 mg tumor tissue sample using the QIAGEN

QIAamp DNA mini kit (QIAGEN). Briefly tissue was pulverized using liquid

nitrogen, then incubated with ALT buffer (QIAGEN) and proteinase K for 96 hr

at 56�C for complete digestion. The following B-RAF and N-RAS single-nucle-

otide polymorphisms (SNPs) were genotyped in tumors and cell lines as part of

the Sequenom OncoCarta Assay Panel v1.0: B-RAF SNPs: G464R, G464V/E,

G466R, F468C, G469S, G469E, G469A, G469V, G469R, D594V/G, F595L,

G596R, L597S, L597R, L597Q, L597V, T599I, V600E, V600K, V600R, V600L,

K601N, K601E. N-RAS SNPs: G12V/A/D, G12C/R/S, G13V/A/D, G13C/R/S,

A18T, Q61L/R/P, Q61H, Q61E/K.

DNA of nevi from three consecutive sections from the paraffin blocks was

isolated using a DNA isolation kit (QIAGEN) according to the manufacturer’s

recommendations. Subsequently, the B-RAFV600E mutation was detected by

real-time PCR as previously described (Benlloch et al., 2006).

Cell Culture

Human neonatal dermal fibroblast (HDF1314) and human neonatal epidermal

melanocytes (HEM1455) were obtained from Cell Applications (San Diego, CA,

USA). Melanocytes, including nHEM cells (Invitrogen), were grown in HAM’s

F10 media, supplemented with ITS premix (Becton Dickinson, Franklin Lakes,

NJ, USA), 12-O-tetradecanoylphorbol-13-acetate (TPA; Sigma-Aldrich, St.

Louis, MO, USA), IBMX (Becton Dickinson), cholera toxin (List Biological Labo-

ratories, Campbell, CA, USA), 20% fetal bovine serum, and glutamine (GIBCO

BRL, Carlsbad, CA, USA) (modified from Halaban et al., 1986). HEK293T, fibro-

blasts, and all melanoma cell lines were grown in Dulbecco’s modified Eagle’s

medium (GIBCO BRL) supplemented with 10% fetal bovine serum, HEPES,

and glutamine. All cells were cultured in a 37�C incubator with 5% CO2. Cells

were treated with media containing 40 mM PD98059 for 72 hr and the media

with MEK inhibitor were replenished every 24 hr.

Lentiviral Transductions

Lentiviruses were produced in HEK293T cells as described previously

(Haferkamp et al., 2009b). Cells were infected using a multiplicity of infection

(moi) between 5 and 10 to provide an efficiency of infection above 90%.

Constructs

The wild-type and mutant B-RAF cDNAs were kindly provided by Professor

R. Marais (The Institute of Cancer Research, UK). An additional missense

mutation in the wild-type B-RAF clone (C > G at nucleotide 634;

NM_004333) was repaired using PCR overlap mutagenesis. The MYC-tagged

wild-type and mutant B-RAF cDNAs were each cloned into the pCDH-CMV-

MCS-EF1-copGFP lentiviral vector, which coexpresses copGFP (System

Biosciences, Mountain View, CA, USA). The IGFBP7 shRNA sequences corre-

spond to nucleotides 963–983 and 865–885 (NM_001553; Wajapeyee et al.,

2008). The V600E-specific B-RAF shRNA sequence corresponds to nucleo-

tides 1853–1871 (NM_004333). The pRb- and p53-specific shRNAs have

been described previously (Haferkamp et al., 2009a). All shRNAs were cloned

into the pSIH-H1-EF1-copGFP and pSIH-H1-Puro lentiviral vectors, which

express copGFP or puromycin resistance, respectively (System Biosciences).

The nonsilencing negative control shRNA did not show complete homology to

any known human transcript and had the following sequence: 50-TTAGAGGC-

GAGCAAGACTA-30.

Western Blotting

Total cellular proteins were extracted at 4�C using RIPA lysis buffer containing

protease inhibitors (Roche, Basel, Switzerland). Proteins (30–50 mg) were

resolved on 12% SDS-polyacrylamide gels and transferred to Immobilon-P

membranes (Millipore, Bedford, MA, USA). Secreted proteins were collected

from the culture media, which were initially cleared by centrifugation. Four

milliliters of the cleared media were centrifuged through 15 ml centricon

centrifugal filter units (50 kDa cut off) (Millipore) at 4000 3 g for 30 min and

the resulting supernatants were loaded onto 10 kDa centricon units and recen-

trifuged. The concentrated sample (100–200 ml) containing soluble IGFBP7

(�37 kDa) was collected and proteins (40 mg) were resolved on 12% SDS-poly-

acrylamide gels and transferred to Immobilon P-membranes (Millipore).

Western blots were probed with antibodies against p16INK4a (N20; Santa

Cruz, Santa Cruz, CA, USA), IGFBP7 (W18R; Santa Cruz), BNIP3L (aa 77–92

from human; ProSci Incorporated, Poway, CA, USA), SMARCB1 (3E10; Ab-

nova, Taipei, Taiwan), PEA15 (H80; Santa Cruz), p21Waf1 (C-19; Santa Cruz),

p53 (DO-1; Santa Cruz), c-MYC (A14; Santa Cruz), phosphorylated ERK (E4;

Santa Cruz), total ERK (137F5; Cell Signaling, Danvers, MA, USA), B-RAF

(L12G7, Cell Signaling), pRb (G3-245; Becton Dickinson), and b-actin

(AC-74; Sigma-Aldrich).

Indirect Immunofluorescence

Cells were seeded on coverslips in 12-well plates at 3 3 104 cells per well at

each time point and incubated overnight. Cells were washed in phosphate-

buffered saline (PBS) and fixed with 4% formaldehyde/PBS for 15 min at

room temperature. Cells were rinsed three times with PBS, then permeabilized

with 0.2% Triton X-100/PBS for 10 min, then rinsed and blocked in 10%

FCS/PBS for 1 hr. Cells were incubated with primary antibodies for 50 min,

washed then incubated with Alexa Fluor 594-conjugated secondary IgG

(Molecular Probes, Carlsbad, CA, USA) and 1 mg/ml of the nuclear DNA stain

40,6-diamidino-2-phenylindole (DAPI; Sigma-Aldrich) for 50 min. Primary anti-

bodies used were Ki67 (MIB-1; DAKO, Glostrup, Denmark), trimethyl-histone

H3 (Lys 9) (07-442; Millipore), and HMGA2 (HMG1-C, FL109; Santa Cruz).

SA-b-Gal activity was detected as described previously (Dimri et al., 1995).

Immunohistochemistry

Tissue arrays of nevi or melanomas were dewaxed before they were rehy-

drated by washing twice with absolute ethanol, two times with 70% ethanol,

and finally by one rinse with bi-distilled water. Samples were antigen retrieved

by heating at 90�C for 20 min in antigen retrieval solution (pH 6) (S1699; DAKO)

or by microwaving for 10 min in 10 mM sodium citrate (pH 6.0) and then cooled

for 20 min. After blocking endogenous peroxidase the slides were incubated

with goat anti-IGFBP7 antibody (C16; Santa Cruz) diluted 1:100 for 25 min to

2 hr, then washed with PBS. Subsequently, the slides were incubated with bio-

tinylated anti-goat antibody (BA-5000; Vector Laboratories, Burlingame, USA

or P0449; DAKO) for 25 min, slides were washed with PBS, incubated in strep-

tavidin-HRP (K0690; DAKO) or streptavidin/biotin-HRP (Invitrogen) for 25 min,

and placed in Vector NovaRed or 3,30-diaminobenzidine substrate (DAB) for

10–15 min (Vector Laboratories). Sections were counterstained with hematox-

ylin and mounted. A pathologist performed the evaluation of the staining in

a blinded manner.

Statistical Analysis

Scatter plots were used to illustrate the distribution of gene expression by

B-RAF mutation status. Medians and interquartile ranges were applied

to summarize the distributions, and the Mann-Whitney test was used to deter-

mine the differences between the B-RAF wild-type and B-RAF mutant

populations.

SUPPLEMENTAL INFORMATION

Supplemental Information includes Extended Experimental Procedures and

two figures and can be found with this article online at doi:10.1016/j.cell.

2010.04.021.

ACKNOWLEDGMENTS

This work is supported by Program Grant 402761, project grants of the

National Health and Medical Research Council of Australia (NHMRC), and an

infrastructure grant to Westmead Millennium Institute by the Health Depart-

ment of NSW through Sydney West Area Health Service. Westmead Institute

for Cancer Research is the recipient of capital grant funding from the Australian

Cancer Research Foundation. L.L.S. is a Cameron Melanoma Research

Fellow, supported by the Melanoma and Skin Cancer Research Institute,

University of Sydney. H.R. is a Cancer Institute New South Wales Research

Fellow. R.A.S. is a Cancer Institute New South Wales Clinical Research Fellow.

We thank Eva-Maria Stuhl, Claudia Siedel, James Wilmott, Suzanah Philipsz,

Branka Mijatov, Svetlana Pianova, and Jason Todd for excellent technical

assistance and Dr. Hermann Kneitz for staining evaluation. The support of

the Melanoma Foundation of the University of Sydney and colleagues from

the Melanoma Institute Australia (incorporating the Sydney Melanoma Unit)

Cell 141, 717–727, May 14, 2010 ª2010 Elsevier Inc. 725

and the Department of Anatomical Pathology at the Royal Prince Alfred

Hospital are also gratefully acknowledged.

Received: July 25, 2009

Revised: November 21, 2009

Accepted: April 15, 2010

Published: May 13, 2010

REFERENCES

Acosta, J.C., O’Loghlen, A., Banito, A., Raguz, S., and Gil, J. (2008). Control of

senescence by CXCR2 and its ligands. Cell Cycle 7, 2956–2959.

Adams, P.D. (2007). Remodeling of chromatin structure in senescent cells and

its potential impact on tumor suppression and aging. Gene 397, 84–93.

Akaogi, K., Sato, J., Okabe, Y., Sakamoto, Y., Yasumitsu, H., and Miyazaki, K.

(1996). Synergistic growth stimulation of mouse fibroblasts by tumor-derived

adhesion factor with insulin-like growth factors and insulin. Cell Growth Differ

7, 1671–1677.

Bartkova, J., Rezaei, N., Liontos, M., Karakaidos, P., Kletsas, D., Issaeva, N.,

Vassiliou, L.V., Kolettas, E., Niforou, K., Zoumpourlis, V.C., et al. (2006). Onco-

gene-induced senescence is part of the tumorigenesis barrier imposed by

DNA damage checkpoints. Nature 444, 633–637.

Bauer, J., Curtin, J.A., Pinkel, D., and Bastian, B.C. (2007). Congenital melano-

cytic nevi frequently harbor NRAS mutations but no BRAF mutations. J. Invest.

Dermatol. 127, 179–182.

Benlloch, S., Paya, A., Alenda, C., Bessa, X., Andreu, M., Jover, R., Castells,

A., Llor, X., Aranda, F.I., and Massuti, B. (2006). Detection of BRAF V600E

mutation in colorectal cancer: comparison of automatic sequencing and

real-time chemistry methodology. J. Mol. Diagn. 8, 540–543.

Bennett, D.C. (2008). How to make a melanoma: what do we know of the

primary clonal events? Pigment Cell Melanoma Res. 21, 27–38.

Burger, A.M., Zhang, X., Li, H., Ostrowski, J.L., Beatty, B., Venanzoni, M.,

Papas, T., and Seth, A. (1998). Down-regulation of T1A12/mac25, a novel

insulin-like growth factor binding protein related gene, is associated with

disease progression in breast carcinomas. Oncogene 16, 2459–2467.

Chen, Y., Pacyna-Gengelbach, M., Ye, F., Knosel, T., Lund, P., Deutschmann,

N., Schluns, K., Kotb, W.F., Sers, C., Yasumoto, H., et al. (2007). Insulin-like

growth factor binding protein-related protein 1 (IGFBP-rP1) has potential

tumour-suppressive activity in human lung cancer. J. Pathol. 211, 431–438.

Chin, L., Garraway, L.A., and Fisher, D.E. (2006). Malignant melanoma:

genetics and therapeutics in the genomic era. Genes Dev. 20, 2149–2182.

Cotter, M.A., Florell, S.R., Leachman, S.A., and Grossman, D. (2007). Absence

of senescence-associated beta-galactosidase activity in human melanocytic

nevi in vivo. J. Invest. Dermatol. 127, 2469–2471.

Curtin, J.A., Busam, K., Pinkel, D., and Bastian, B.C. (2006). Somatic activation

of KIT in distinct subtypes of melanoma. J. Clin. Oncol. 24, 4340–4346.

Degeorges, A., Wang, F., Frierson, H.F., Jr., Seth, A., Chung, L.W., and Sikes,

R.A. (1999). Human prostate cancer expresses the low affinity insulin-like

growth factor binding protein IGFBP-rP1. Cancer Res. 59, 2787–2790.

Denoyelle, C., Abou-Rjaily, G., Bezrookove, V., Verhaegen, M., Johnson, T.M.,

Fullen, D.R., Pointer, J.N., Gruber, S.B., Su, L.D., Nikiforov, M.A., et al. (2006).

Anti-oncogenic role of the endoplasmic reticulum differentially activated by

mutations in the MAPK pathway. Nat. Cell Biol. 8, 1053–1063.

Di Micco, R., Fumagalli, M., Cicalese, A., Piccinin, S., Gasparini, P., Luise, C.,

Schurra, C., Garre, M., Nuciforo, P.G., Bensimon, A., et al. (2006). Oncogene-

induced senescence is a DNA damage response triggered by DNA hyper-

replication. Nature 444, 638–642.

Dimri, G.P., Lee, X., Basile, G., Acosta, M., Scott, G., Roskelley, C., Medrano,

E.E., Linskens, M., Rubelj, I., Pereira-Smith, O., et al. (1995). A biomarker that

identifies senescent human cells in culture and in aging skin in vivo. Proc. Natl.

Acad. Sci. USA 92, 9363–9367.

Falchi, M., Bataille, V., Hayward, N.K., Duffy, D.L., Bishop, J.A., Pastinen, T.,

Cervino, A., Zhao, Z.Z., Deloukas, P., Soranzo, N., et al. (2009). Genome-

726 Cell 141, 717–727, May 14, 2010 ª2010 Elsevier Inc.

wide association study identifies variants at 9p21 and 22q13 associated

with development of cutaneous nevi. Nat. Genet. 41, 915–919.

Gray-Schopfer, V.C., Cheong, S.C., Chong, H., Chow, J., Moss, T.,

Abdel-Malek, Z.A., Marais, R., Wynford-Thomas, D., and Bennett, D.C.

(2006). Cellular senescence in naevi and immortalisation in melanoma: a role

for p16? Br. J. Cancer 95, 496–505.

Haferkamp, S., Tran, S., Becker, T.M., Scurr, L.L., Kefford, R.F., and Rizos, H.

(2009a). The relative contributions of the p53 and pRb pathways in oncogene-

induced melanocyte senescence. Aging 1, 542–556.

Haferkamp, S., Scurr, L.L., Becker, T.M., Frausto, M., Kefford, R.F., and Rizos,

H. (2009b). Oncogene-induced senescence does not require the p16(INK4a) or

p14ARF melanoma tumor suppressors. J. Invest. Dermatol. 128, 1983–1991.

Halaban, R., Ghosh, S., Duray, P., Kirkwood, J.M., and Lerner, A.B. (1986).

Human melanocytes cultured from nevi and melanomas. J. Invest. Dermatol.

87, 95–101.

Harrison, S.L., MacLennan, R., and Buettner, P.G. (2008). Sun exposure and

the incidence of melanocytic nevi in young Australian children. Cancer Epide-

miol. Biomarkers Prev. 17, 2318–2324.

Herlyn, M. (2007). Farming cells to rebuild skin and melanoma. Cancer Biol.

Ther. 6, 467–471.

Jiang, W., Xiang, C., Cazacu, S., Brodie, C., and Mikkelsen, T. (2008). Insulin-

like growth factor binding protein 7 mediates glioma cell growth and migration.

Neoplasia 10, 1335–1342.

Kelly, J.W., Yeatman, J.M., Regalia, C., Mason, G., and Henham, A.P. (1997).

A high incidence of melanoma found in patients with multiple dysplastic naevi

by photographic surveillance. Med. J. Aust. 167, 191–194.

Kuilman, T., and Peeper, D.S. (2009). Senescence-messaging secretome:

SMS-ing cellular stress. Nat. Rev. Cancer 9, 81–94.

Landberg, G., Ostlund, H., Nielsen, N.H., Roos, G., Emdin, S., Burger, A.M.,

and Seth, A. (2001). Downregulation of the potential suppressor gene

IGFBP-rP1 in human breast cancer is associated with inactivation of the reti-

noblastoma protein, cyclin E overexpression and increased proliferation in

estrogen receptor negative tumors. Oncogene 20, 3497–3505.

Michaloglou, C., Vredeveld, L.C., Soengas, M.S., Denoyelle, C., Kuilman, T.,

van der Horst, C.M., Majoor, D.M., Shay, J.W., Mooi, W.J., and Peeper, D.S.

(2005). BRAFE600-associated senescence-like cell cycle arrest of human

naevi. Nature 436, 720–724.

Papp, T., Pemsel, H., Zimmermann, R., Bastrop, R., Weiss, D.G., and Schiff-

mann, D. (1999). Mutational analysis of the N-ras, p53, p16INK4a, CDK4,

and MC1R genes in human congenital melanocytic naevi. J. Med. Genet.

36, 610–614.

Pen, A., Moreno, M.J., Martin, J., and Stanimirovic, D.B. (2007). Molecular

markers of extracellular matrix remodeling in glioblastoma vessels: microarray

study of laser-captured glioblastoma vessels. Glia 55, 559–572.

Pen, A., Moreno, M.J., Durocher, Y., Deb-Rinker, P., and Stanimirovic, D.B.

(2008). Glioblastoma-secreted factors induce IGFBP7 and angiogenesis by

modulating Smad-2-dependent TGF-beta signaling. Oncogene 27,

6834–6844.

Platz, A., Egyhazi, S., Ringborg, U., and Hansson, J. (2008). Human cutaneous

melanoma; a review of NRAS and BRAF mutation frequencies in relation to

histogenetic subclass and body site. Mol. Oncol. 1, 395–405.

Pollock, P.M., Harper, U.L., Hansen, K.S., Yudt, L.M., Stark, M., Robbins,

C.M., Moses, T.Y., Hostetter, G., Wagner, U., Kakareka, J., et al. (2003).

High frequency of BRAF mutations in nevi. Nat. Genet. 33, 19–20.

Prieur, A., and Peeper, D.S. (2008). Cellular senescence in vivo: a barrier to

tumorigenesis. Curr. Opin. Cell Biol. 20, 150–155.

Ruan, W., Xu, E., Xu, F., Ma, Y., Deng, H., Huang, Q., Lv, B., Hu, H., Lin, J.,

Cui, J., et al. (2007). IGFBP7 plays a potential tumor suppressor role in colo-

rectal carcinogenesis. Cancer Biol. Ther. 6, 354–359.

Schrama, D., Kneitz, H., Willmes, C., Adam, C., Houben, R., and Becker, J.C.

(2009). Lack of correlation between IGFBP7 expression and BRAF mutational

status in melanoma. J. Invest. Dermatol. 130, 897–898.

Shao, L., Huang, Q., Luo, M., and Lai, M. (2004). Detection of the differentially

expressed gene IGF-binding protein-related protein-1 and analysis of its rela-

tionship to fasting glucose in Chinese colorectal cancer patients. Endocr.

Relat. Cancer 11, 141–148.

Suzuki, H., Igarashi, S., Nojima, M., Maruyama, R., Yamamoto, E., Kai, M.,

Akashi, H., Watanabe, Y., Yamamoto, H., Sasaki, Y., et al. (2009). IGFBP7 is

a p53 responsive gene specifically silenced in colorectal cancer with CpG

island methylator phenotype. Carcinogenesis 31, 342–349.

Sviderskaya, E.V., Gray-Schopfer, V.C., Hill, S.P., Smit, N.P., Evans-Whipp,

T.J., Bond, J., Hill, L., Bataille, V., Peters, G., Kipling, D., et al. (2003). p16/Cy-

clin-dependent kinase inhibitor 2A deficiency in human melanocyte senes-

cence, apoptosis, and immortalization: possible implications for melanoma

progression. J. Natl. Cancer Inst. 95, 723–732.

Swisshelm, K., Ryan, K., Tsuchiya, K., and Sager, R. (1995). Enhanced expres-

sion of an insulin growth factor-like binding protein (mac25) in senescent

human mammary epithelial cells and induced expression with retinoic acid.

Proc. Natl. Acad. Sci. USA 92, 4472–4476.

Takeno, A., Takemasa, I., Doki, Y., Yamasaki, M., Miyata, H., Takiguchi, S.,

Fujiwara, Y., Matsubara, K., and Monden, M. (2008). Integrative approach

for differentially overexpressed genes in gastric cancer by combining large-

scale gene expression profiling and network analysis. Br. J. Cancer 99,

1307–1315.

Thompson, J.F., Scolyer, R.A., and Kefford, R.F. (2005). Cutaneous mela-

noma. Lancet 365, 687–701.

Wajapeyee, N., Serra, R.W., Zhu, X., Mahalingam, M., and Green, M.R. (2008).

Oncogenic BRAF induces senescence and apoptosis through pathways medi-

ated by the secreted protein IGFBP7. Cell 132, 363–374.

Wajapeyee, N., Kapoor, V., Mahalingam, M., and Green, M.R. (2009). Efficacy

of IGFBP7 for treatment of metastatic melanoma and other cancers in mouse

models and human cell lines. Mol. Cancer Ther. 8, 3009–3014.

Ye, F., Chen, Y., Knosel, T., Schluns, K., Pacyna-Gengelbach, M., Deutsch-

mann, N., Lai, M., and Petersen, I. (2007). Decreased expression of insulin-

like growth factor binding protein 7 in human colorectal carcinoma is related

to DNA methylation. J. Cancer Res. Clin. Oncol. 133, 305–314.

Yu, H., McDaid, R., Lee, J., Possik, P., Li, L., Kumar, S.M., Elder, D.E., Van

Belle, P., Gimotty, P., Guerra, M., et al. (2009). The role of BRAF mutation

and p53 inactivation during transformation of a subpopulation of primary

human melanocytes. Am. J. Pathol. 74, 2367–2377.

Zhuang, D., Mannava, S., Grachtchouk, V., Tang, W.H., Patil, S., Wawrzyniak,

J.A., Berman, A.E., Giordano, T.J., Prochownik, E.V., Soengas, M.S., et al.

(2008). C-MYC overexpression is required for continuous suppression of

oncogene-induced senescence in melanoma cells. Oncogene 27, 6623–6634.

Cell 141, 717–727, May 14, 2010 ª2010 Elsevier Inc. 727