PHLDA3 is a novel tumor suppressor of pancreatic ... · Pancreatic neuroendocrine tumors (PanNETs)...

10
PHLDA3 is a novel tumor suppressor of pancreatic neuroendocrine tumors Rieko Ohki a,1 , Kozue Saito a,b , Yu Chen a,b , Tatsuya Kawase c , Nobuyoshi Hiraoka d , Raira Saigawa a,b , Maiko Minegishi a,b , Yukie Aita a , Goichi Yanai e , Hiroko Shimizu f , Shinichi Yachida a , Naoaki Sakata g , Ryuichiro Doi h , Tomoo Kosuge i , Kazuaki Shimada i , Benjamin Tycko j , Toshihiko Tsukada k , Yae Kanai d , Shoichiro Sumi e , Hideo Namiki b , Yoichi Taya c,l , Tatsuhiro Shibata f,2 , and Hitoshi Nakagama a,2 Divisions of a Refractory Cancer Research, c Radiobiology, d Molecular Pathology, f Cancer Genomics, and k Familial Cancer Research, National Cancer Center Research Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan; b Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan; e Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; g Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; h Department of Surgery and Surgical Basic Science, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; i Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan; j Institute for Cancer Genetics, Columbia University, New York, NY 10032; and l Cancer Research Center of Excellence, Center for Life Sciences, National University of Singapore, Singapore 117456 Edited by Douglas Hanahan, Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland, and approved May 1, 2014 (received for review October 25, 2013) The molecular mechanisms underlying the development of pancre- atic neuroendocrine tumors (PanNETs) have not been well defined. We report here that the genomic region of the PHLDA3 gene undergoes loss of heterozygosity (LOH) at a remarkably high fre- quency in human PanNETs, and this genetic change is correlated with disease progression and poor prognosis. We also show that the PHLDA3 locus undergoes methylation in addition to LOH, sug- gesting that a two-hit inactivation of the PHLDA3 gene is required for PanNET development. We demonstrate that PHLDA3 represses Akt activity and Akt-regulated biological processes in pancre- atic endocrine tissues, and that PHLDA3-deficient mice develop islet hyperplasia. In addition, we show that the tumor-suppressing pathway mediated by MEN1, a well-known tumor suppressor of PanNETs, is dependent on the pathway mediated by PHLDA3, and inactivation of PHLDA3 and MEN1 cooperatively contribute to PanNET development. Collectively, these results indicate the existence of a novel PHLDA3-mediated pathway of tumor suppres- sion that is important in the development of PanNETs. p53 | PH domain | everolimus | p53 target gene | mTOR N euroendocrine tumors (NETs) arise from cells of the en- docrine and nervous systems, and are found in tissues such as lung, pancreas and pituitary (13). NETs often produce, store and release biogenic amines and polypeptide hormones, and secretary granules containing these products provide a diagnostic marker for NETs. The mechanisms underlying the development of NETs remain unclear to date, due to the low incidence of these tumors and due to the lack of suitable experimental model systems, including genetically engineered mouse models. Pan- creatic NET (PanNET), which is probably the best-studied NET, is the second-most common pancreatic tumor, having an in- cidence of 1 per 100,000 individuals. Patients having late-stage PanNET often harbor tumors that are unresectable or metastatic and face limited treatment options. Accordingly, the prognosis of patients having metastatic PanNET is the worst among the NET subtypes, with a 5-y survival rate of 2743% (1). Recently, the drug Everolimus has shown promise in the treatment of PanNETs (4), providing a significant improvement in progression-free survival. Everolimus is an inhibitor of mammalian target of rapamycin (mTOR), a downstream mediator of the phosphoi- nositide 3-kinase (PI3K)/protein kinase B (AKT) pathway. The striking efficacy of Everolimus demonstrates the importance of the PI3K/Akt pathway in the pathology of PanNETs. In agreement with these clinical results, studies on pancreatic endocrine cell lines have identified the PI3K/Akt signaling pathway as a major proliferation and survival pathway in these cells (5). Activated Akt phosphorylates substrates such as mTOR and controls various biological processes, including protein syn- thesis, proliferation, cell growth, and survival. Regulation of pancreatic islet β-cell proliferation, cell size, and apoptosis by Akt has been demonstrated using various mouse models. For example, transgenic mice overexpressing constitutively active Akt in β-cells exhibit increased β-cell proliferation and cell size and decreased induction of apoptosis (6). Recently, the results of whole exomic sequencing of 10 Pan- NET specimens were published, revealing several key genetic alterations (7). In particular, genes in the PI3K/Akt pathway, i.e., TSC2, PTEN, and PIK3CA, were mutated in 15% of PanNETs. However, this represents only a subset of PanNETs, and may not fully explain the remarkable clinical results achieved by Ever- olimus in the majority of PanNET patients. Previously, we have shown that Pleckstrin homology-like domain family A, member 3 (PHLDA3) is a novel p53-regu- lated repressor of Akt. The PHLDA3 contains a PH domain that, we showed, competes with the PH domain of Akt for binding to membrane lipids, thereby inhibiting Akt translocation to the cellular membrane and its activation. We also showed that Significance Pancreatic neuroendocrine tumors (PanNETs) are a rare pa- thology, and molecular mechanisms underlying their develop- ment have not been well defined. This article shows that a two-hit inactivation of the PHLDA3 gene is required for PanNET development: methylation of the locus and loss of heterozygosity. PHLDA3 functions as a suppressor of PanNETs via repression of Akt activity and downstream Akt-regulated biological processes. In addition, the tumor-suppressing path- way mediated by MEN1, a well known suppressor of PanNETs, is dependent on the pathway mediated by PHLDA3, and in- activation of PHLDA3 and MEN1 cooperatively contribute to PanNET development. A novel PHLDA3-mediated pathway of tumor suppression that is important in the development of PanNETs is demonstrated, and the findings may contribute to personalized medicine of PanNET patients. Author contributions: R.O. designed research; R.O., K. Saito, Y.C., R.S., M.M., Y.A., and H.S. performed research; T. Kawase, N.H., G.Y., S.Y., N.S., R.D., T. Kosuge, K. Shimada, B.T., T.T., Y.K., and S.S. contributed new reagents/analytic tools; R.O. and K. Saito analyzed data; R.O., H. Namiki, Y.T., T.S. and H. Nakagama supervised the research; and R.O. wrote the paper. The authors declare no conflict of interest. This article is a PNAS Direct Submission. 1 To whom correspondence should be addressed. E-mail: [email protected]. 2 T.S. and H. Nakagama contributed equally to this work. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1319962111/-/DCSupplemental. E2404E2413 | PNAS | Published online May 27, 2014 www.pnas.org/cgi/doi/10.1073/pnas.1319962111

Transcript of PHLDA3 is a novel tumor suppressor of pancreatic ... · Pancreatic neuroendocrine tumors (PanNETs)...

Page 1: PHLDA3 is a novel tumor suppressor of pancreatic ... · Pancreatic neuroendocrine tumors (PanNETs) are a rare pa-thology, and molecular mechanisms underlying their develop-ment have

PHLDA3 is a novel tumor suppressor of pancreaticneuroendocrine tumorsRieko Ohkia,1, Kozue Saitoa,b, Yu Chena,b, Tatsuya Kawasec, Nobuyoshi Hiraokad, Raira Saigawaa,b, Maiko Minegishia,b,Yukie Aitaa, Goichi Yanaie, Hiroko Shimizuf, Shinichi Yachidaa, Naoaki Sakatag, Ryuichiro Doih, Tomoo Kosugei,Kazuaki Shimadai, Benjamin Tyckoj, Toshihiko Tsukadak, Yae Kanaid, Shoichiro Sumie, Hideo Namikib, Yoichi Tayac,l,Tatsuhiro Shibataf,2, and Hitoshi Nakagamaa,2

Divisions of aRefractory Cancer Research, cRadiobiology, dMolecular Pathology, fCancer Genomics, and kFamilial Cancer Research, National Cancer CenterResearch Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan; bGraduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo,Shinjuku-ku, Tokyo 169-8555, Japan; eDepartment of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho,Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; gDivision of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Tohoku University Graduate School ofMedicine, 2-1, Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; hDepartment of Surgery and Surgical Basic Science, Graduate School of Medicine,Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; iHepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital,Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan; jInstitute for Cancer Genetics, Columbia University, New York, NY 10032; and lCancer Research Center ofExcellence, Center for Life Sciences, National University of Singapore, Singapore 117456

Edited by Douglas Hanahan, Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland, and approved May 1, 2014 (received for reviewOctober 25, 2013)

The molecular mechanisms underlying the development of pancre-atic neuroendocrine tumors (PanNETs) have not been well defined.We report here that the genomic region of the PHLDA3 geneundergoes loss of heterozygosity (LOH) at a remarkably high fre-quency in human PanNETs, and this genetic change is correlatedwith disease progression and poor prognosis. We also show thatthe PHLDA3 locus undergoes methylation in addition to LOH, sug-gesting that a two-hit inactivation of the PHLDA3 gene is requiredfor PanNET development. We demonstrate that PHLDA3 repressesAkt activity and Akt-regulated biological processes in pancre-atic endocrine tissues, and that PHLDA3-deficient mice developislet hyperplasia. In addition, we show that the tumor-suppressingpathway mediated by MEN1, a well-known tumor suppressor ofPanNETs, is dependent on the pathway mediated by PHLDA3,and inactivation of PHLDA3 and MEN1 cooperatively contributeto PanNET development. Collectively, these results indicate theexistence of a novel PHLDA3-mediated pathway of tumor suppres-sion that is important in the development of PanNETs.

p53 | PH domain | everolimus | p53 target gene | mTOR

Neuroendocrine tumors (NETs) arise from cells of the en-docrine and nervous systems, and are found in tissues such

as lung, pancreas and pituitary (1–3). NETs often produce, storeand release biogenic amines and polypeptide hormones, andsecretary granules containing these products provide a diagnosticmarker for NETs. The mechanisms underlying the developmentof NETs remain unclear to date, due to the low incidence ofthese tumors and due to the lack of suitable experimental modelsystems, including genetically engineered mouse models. Pan-creatic NET (PanNET), which is probably the best-studied NET,is the second-most common pancreatic tumor, having an in-cidence of ∼1 per 100,000 individuals. Patients having late-stagePanNET often harbor tumors that are unresectable or metastaticand face limited treatment options. Accordingly, the prognosisof patients having metastatic PanNET is the worst among theNET subtypes, with a 5-y survival rate of 27–43% (1). Recently, thedrug Everolimus has shown promise in the treatment of PanNETs(4), providing a significant improvement in progression-freesurvival. Everolimus is an inhibitor of mammalian target ofrapamycin (mTOR), a downstream mediator of the phosphoi-nositide 3-kinase (PI3K)/protein kinase B (AKT) pathway. Thestriking efficacy of Everolimus demonstrates the importance ofthe PI3K/Akt pathway in the pathology of PanNETs.In agreement with these clinical results, studies on pancreatic

endocrine cell lines have identified the PI3K/Akt signalingpathway as a major proliferation and survival pathway in thesecells (5). Activated Akt phosphorylates substrates such as mTORand controls various biological processes, including protein syn-

thesis, proliferation, cell growth, and survival. Regulation ofpancreatic islet β-cell proliferation, cell size, and apoptosis byAkt has been demonstrated using various mouse models. Forexample, transgenic mice overexpressing constitutively activeAkt in β-cells exhibit increased β-cell proliferation and cell sizeand decreased induction of apoptosis (6).Recently, the results of whole exomic sequencing of 10 Pan-

NET specimens were published, revealing several key geneticalterations (7). In particular, genes in the PI3K/Akt pathway, i.e.,TSC2, PTEN, and PIK3CA, were mutated in 15% of PanNETs.However, this represents only a subset of PanNETs, and may notfully explain the remarkable clinical results achieved by Ever-olimus in the majority of PanNET patients.Previously, we have shown that Pleckstrin homology-like

domain family A, member 3 (PHLDA3) is a novel p53-regu-lated repressor of Akt. The PHLDA3 contains a PH domainthat, we showed, competes with the PH domain of Akt forbinding to membrane lipids, thereby inhibiting Akt translocationto the cellular membrane and its activation. We also showed that

Significance

Pancreatic neuroendocrine tumors (PanNETs) are a rare pa-thology, and molecular mechanisms underlying their develop-ment have not been well defined. This article shows thata two-hit inactivation of the PHLDA3 gene is required forPanNET development: methylation of the locus and loss ofheterozygosity. PHLDA3 functions as a suppressor of PanNETsvia repression of Akt activity and downstream Akt-regulatedbiological processes. In addition, the tumor-suppressing path-way mediated by MEN1, a well known suppressor of PanNETs,is dependent on the pathway mediated by PHLDA3, and in-activation of PHLDA3 and MEN1 cooperatively contribute toPanNET development. A novel PHLDA3-mediated pathway oftumor suppression that is important in the development ofPanNETs is demonstrated, and the findings may contribute topersonalized medicine of PanNET patients.

Author contributions: R.O. designed research; R.O., K. Saito, Y.C., R.S., M.M., Y.A., and H.S.performed research; T. Kawase, N.H., G.Y., S.Y., N.S., R.D., T. Kosuge, K. Shimada, B.T.,T.T., Y.K., and S.S. contributed new reagents/analytic tools; R.O. and K. Saito analyzeddata; R.O., H. Namiki, Y.T., T.S. and H. Nakagama supervised the research; and R.O. wrotethe paper.

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.1To whom correspondence should be addressed. E-mail: [email protected]. and H. Nakagama contributed equally to this work.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1319962111/-/DCSupplemental.

E2404–E2413 | PNAS | Published online May 27, 2014 www.pnas.org/cgi/doi/10.1073/pnas.1319962111

Page 2: PHLDA3 is a novel tumor suppressor of pancreatic ... · Pancreatic neuroendocrine tumors (PanNETs) are a rare pa-thology, and molecular mechanisms underlying their develop-ment have

PHLDA3 may have a tumor suppressive function (8). However,there has hitherto been no reported role for PHLDA3 in humantumors, and its in vivo function has remained elusive. In thisreport, we demonstrate that the PHLDA3 gene is a novel tumorsuppressor, inactivation of which can lead to the development ofPanNETs. We show that the PHLDA3 genomic locus undergoesLOH and that the PHLDA3 promoter is methylated at a highfrequency in PanNETs. Furthermore, analysis of PHLDA3-deficient mice showed that these mice frequently develop islethyperplasia as a result of enhanced islet cell proliferation and anincrease in islet cell size. Collectively, these results indicate thatPHLDA3 functions as a tumor suppressor in PanNETs.

ResultsFrequent LOH at the PHLDA3 Gene Locus in PanNETs. The PHLDA3gene is located at 1q31, a locus that has been reported to havea high frequency of LOH in two NETs derived from pancreas:insulinomas and gastrinomas (9, 10). We therefore speculatedthat the PHLDA3 locus may undergo LOH in PanNETs, andanalyzed the PHLDA3 locus for LOH using microsatellite markerssurrounding the gene in 54 PanNET samples (Fig. 1 A–D; clinicaldiagnosis for each sample is shown in SI Appendix, Fig. S1A). As

shown in Fig. 1B, out of 54 PanNETs, 50 samples were informativeand 36 samples showed LOH at the PHLDA3 locus. The incidenceof LOH at the PHLDA3 locus (72%) is remarkably high, and wascomparable to the reported LOH incidence of the Multiple endo-crine neoplasia type 1 (MEN1) gene, which has the highest reportedincidence of genomic changes in PanNETs (11). Within the re-gion analyzed, the LOH frequency peaks near the PHLDA3 lo-cus, suggesting that LOH of the PHLDA3 gene is critical forPanNET development (Fig. 1D). This tendency becomes clearerwhen samples that exhibit partial LOH within this region (PanNET1–18) were analyzed (Fig. 1D, blue line). A strikingly high in-cidence of LOH at the PHLDA3 locus indicates the impor-tance of this PHLDA3-regulated tumor suppression pathwayin PanNETs. Most of the PanNETs analyzed in this study arenonfunctional, and we found no associations between PHLDA3LOH and specific PanNET type or insulin/glucagon positivity, tothe extent that we examined this (SI Appendix, Fig. S1).

PHLDA3 and MEN1 Cooperatively Suppress PanNET. The most out-standing genomic aberration previously reported in PanNETswas the mutation and LOH of the MEN1 gene, a tumor sup-pressor gene associated with multiple endocrine neoplasia type 1

A

B

C

Chromosome 1q

PHLDA3

200 205 (M)

D1S2622D1S306

D1S510 D1S249

D1S2738

D1S1723

40

50

60

70

80

90

100

LO

H f

req

uen

cy (

%)

200 205201 206202 204203 (M)Chromosome 1q

all sample

sample with partial LOH

D1S249D1S2622

D1S2738

D1S1723

D1S306

D1S510

PHLDA3

Normal

PanNET

D1S306 D

PHLDA3 LOH

PHLDA3 Hetero

no data

D1S2622

D1S2738

D1S1723

D1S306

LOH atPHLDA3

D1S510

D1S249

Pan

NE

T1

Pan

NE

T2

Pan

NE

T3

Pan

NE

T4

Pan

NE

T5

Pan

NE

T6

Pan

NE

T7

Pan

NE

T8

Pan

NE

T9

Pan

NE

T10

Pan

NE

T11

Pan

NE

T12

Pan

NE

T13

Pan

NE

T14

Pan

NE

T15

Pan

NE

T16

Pan

NE

T17

Pan

NE

T18

Pan

NE

T19

Pan

NE

T20

Pan

NE

T21

Pan

NE

T22

Pan

NE

T23

Pan

NE

T24

Pan

NE

T25

Pan

NE

T26

Pan

NE

T27

Pan

NE

T28

Pan

NE

T29

Pan

NE

T30

Pan

NE

T31

Pan

NE

T32

Pan

NE

T33

Pan

NE

T34

Pan

NE

T35

Pan

NE

T36

Pan

NE

T37

Pan

NE

T38

Pan

NE

T39

Pan

NE

T40

Pan

NE

T41

Pan

NE

T42

Pan

NE

T43

Pan

NE

T44

Pan

NE

T45

Pan

NE

T46

Pan

NE

T47

Pan

NE

T48

Pan

NE

T49

Pan

NE

T50

Pan

NE

T51

Pan

NE

T52

Pan

NE

T53

Pan

NE

T54

Fig. 1. Frequency of LOH at the PHLDA3 gene locus in PanNETs. (A) Chromosomal locations of PHLDA3 gene and microsatellite markers used in this study. D1S306 islocated just next to the PHLDA3 gene (32 kb upstream). (B) Microsatellite analysis of the PHLDA3 gene locus region. PanNET samples were analyzed for LOH aroundthe PHLDA3 gene locus. Because D1S306 is located next to the PHLDA3 gene, the LOH status of the PHLDA3 gene was determined from the LOH status of the D1S306locus. For some loci with no data (not informative or data unavailable), the LOH status of the locus was determined from the surrounding LOH status (shown in faintpink and faint blue). (C) Representative microsatellite analysis results. In normal tissues, two peaks derived frommaternal and paternal alleles were detected, whereasin tumors, one allele was lost (shown by orange arrows), indicating LOH at the locus. (D) LOH frequency for each microsatellite marker. Frequencies from all samples(shown by red line) and frequencies from samples showing LOH partially within the analyzed region (PanNET1-18, shown by blue line) are shown.

Ohki et al. PNAS | Published online May 27, 2014 | E2405

MED

ICALSC

IENCE

SPN

ASPL

US

Page 3: PHLDA3 is a novel tumor suppressor of pancreatic ... · Pancreatic neuroendocrine tumors (PanNETs) are a rare pa-thology, and molecular mechanisms underlying their develop-ment have

(7, 11). It has been reported that ∼50% of PanNET cases exhibitLOH of the MEN1 gene. Therefore, we analyzed whether MEN1LOH is observed in our PanNET samples. As shown in Fig. 2 Aand B, 32 samples out of 48 informative samples showed LOH atthe MEN1 locus. Frequent LOH was observed at the MEN1 locusin our samples (67%), confirming previous studies. We next com-bined the LOH data for the PHLDA3 and MEN1 loci. As shown inFig. 2C, LOH at the PHLDA3 and MEN1 loci did not show a mu-tually exclusive pattern, which would be expected if PHLDA3 andMEN1 were on the same tumor suppressing pathway. Interestingly,we observed a significant frequency of double LOH, i.e., occurringat both the PHLDA3 andMEN1 loci (25 of 45 samples, P < 0.05 byFisher’s exact test). These data suggest that development of Pan-NET involves the functional loss of both pathways.

LOH at the PHLDA3 Locus Is Correlated to Poor Prognosis in PanNETPatients. To select the proper treatment for each PanNET pa-tient, prediction of disease prognosis is important. We observedthat LOH at the PHLDA3 locus was associated with advancedstage PanNETs, whereas absence of LOH was associated withlower tumor grades. These associations were statistically signif-icant (P < 0.01, by Fisher’s exact test), suggesting that the LOH isassociated with a malignant phenotype in PanNETs (Fig. 3A).We next analyzed the relationship between LOH at the PHLDA3locus and the prognosis of PanNET patients. As shown in Fig.3B, patients exhibiting LOH at the PHLDA3 locus seemed tohave a poorer prognosis compared with the patients withoutLOH. However, the observed difference did not achieve statis-tical significance, probably due to the relatively small numbers ofpatients analyzed. On the other hand, LOH at the MEN1 locushad no influence on tumor grade or patient survival, as has beenreported (7) (Fig. 3 C and D). It will be necessary to extend thesestudies with larger numbers of patients before reaching a firmconclusion, however these results suggest that the PHLDA3-

regulated pathway, but not the MEN1-regulated pathway, maybe a critical determinant of the prognosis of PanNET patients.

The PHLDA3 Gene Undergoes Aberrant Methylation in Addition toLOH in PanNETs. Because we found frequent LOH at the PHLDA3locus in PanNETs, we next examined how the remaining alleleis inactivated. The PHLDA3 gene is mutated in several can-cers: 11 mutations in lung, urinary, and large intestine cancerare reported in the COSMIC database (http://cancer.sanger.ac.uk/cancergenome/projects/cosmic), and mutation in lung cancerwas reported by Yoo et al. (12). Therefore, we analyzed thePHLDA3 ORF genomic sequence in our PanNET samples, butdid not find any mutations within the coding regions (SI Appendix,Fig. S2). We next analyzed PHLDA3 mRNA expression levels inthese PanNETs. As shown in Fig. 4A, PHLDA3 expression wassignificantly lower in samples showing LOH at the PHLDA3locus compared with samples without LOH. We noticed a CpGisland overlapping the promoter region and the first exon of thePHLDA3 gene (Fig. 4B). We therefore analyzed whether themethylation status of the PHLDA3 gene is related to PHLDA3transcription levels. Specifically, we analyzed DNA methylationlevels within the first exon of the PHLDA3 gene, because it hasbeen reported that methylation of the first exon is tightly linkedto transcriptional silencing (13). Analysis of PHLDA3 mRNAexpression levels in four cancer cell lines revealed that PHLDA3is highly expressed in LNCaP and MDA-MB-M468 cells, whereasexpression is very low in DLD1 and H1299 cells (Fig. 4C). Analysisof DNA methylation levels by methylation-specific PCR revealeddetectable methylation only in DLD1 and H1299 cells, cell lineswith low PHLDA3 expression (Fig. 4D). It is of note that, inH1299 cells, a human lung NET cell line, PHLDA3 mRNA ex-pression was remarkably low and methylation was remarkablyhigh. We further treated H1299 cells with 5-aza-C to demethy-late the PHLDA3 gene. As shown in Fig. 4E, 5-aza-C treatmentresulted in decreased methylation at the PHLDA3 gene and

A

B

Chromosome 11qD11S4940

MEN1 gene

D11S4946PYGM

64.5 64.6 64.7(M)

MEN1 HeteroMEN1 LOH

no data

D11S4940

D11S4946

LOH atMEN1

PYGM

Pan

NE

T1

Pan

NE

T2

Pan

NE

T3

Pan

NE

T4

Pan

NE

T5

Pan

NE

T6

Pan

NE

T7

Pan

NE

T8

Pan

NE

T9

Pan

NE

T10

Pan

NE

T11

Pan

NE

T12

Pan

NE

T13

Pan

NE

T14

Pan

NE

T15

Pan

NE

T16

Pan

NE

T17

Pan

NE

T18

Pan

NE

T19

Pan

NE

T20

Pan

NE

T21

Pan

NE

T22

Pan

NE

T23

Pan

NE

T24

Pan

NE

T25

Pan

NE

T26

Pan

NE

T27

Pan

NE

T28

Pan

NE

T29

Pan

NE

T30

Pan

NE

T31

Pan

NE

T32

Pan

NE

T33

Pan

NE

T34

Pan

NE

T35

Pan

NE

T36

Pan

NE

T37

Pan

NE

T38

Pan

NE

T39

Pan

NE

T40

Pan

NE

T41

Pan

NE

T42

Pan

NE

T43

Pan

NE

T44

Pan

NE

T45

Pan

NE

T46

Pan

NE

T47

Pan

NE

T48

Pan

NE

T49

Pan

NE

T50

Pan

NE

T51

Pan

NE

T52

Pan

NE

T53

Pan

NE

T54

PHLDA3 LOH

Hetero

MEN1 LOH

no data

MEN1

PHLDA3

Pan

NE

T1

Pan

NE

T2

Pan

NE

T3

Pan

NE

T4

Pan

NE

T5

Pan

NE

T6

Pan

NE

T7

Pan

NE

T8

Pan

NE

T9

Pan

NE

T10

Pan

NE

T11

Pan

NE

T12

Pan

NE

T13

Pan

NE

T14

Pan

NE

T15

Pan

NE

T16

Pan

NE

T17

Pan

NE

T18

Pan

NE

T19

Pan

NE

T20

Pan

NE

T21

Pan

NE

T22

Pan

NE

T23

Pan

NE

T24

Pan

NE

T25

Pan

NE

T26

Pan

NE

T27

Pan

NE

T28

Pan

NE

T29

Pan

NE

T30

Pan

NE

T31

Pan

NE

T32

Pan

NE

T33

Pan

NE

T34

Pan

NE

T35

Pan

NE

T36

Pan

NE

T37

Pan

NE

T38

Pan

NE

T39

Pan

NE

T40

Pan

NE

T41

Pan

NE

T42

Pan

NE

T43

Pan

NE

T44

Pan

NE

T45

Pan

NE

T46

Pan

NE

T47

Pan

NE

T48

Pan

NE

T49

Pan

NE

T50

Pan

NE

T51

Pan

NE

T52

Pan

NE

T53

Pan

NE

T54C

PHLDA3 LOH (32/45) PHLDA3 hetero (13/45)

MEN1 LOH 25 5

MEN1 hetero 7 8

(30/45)

(15/45) (Fisher's exact test, p<0.05)

Fig. 2. Relationship between PHLDA3 and MEN1 tumor-suppressing pathways. (A) Chromosomal locations of the MEN1 gene and microsatellite markersused in this study. (B) Analysis of LOH at the MEN1 locus. The LOH status of the MEN1 gene was determined from the LOH status of either of the informativemarkers. (C) Relationship between LOH status of the PHLDA3 and MEN1 loci. In total, 45 samples informative for both PHLDA3 and MEN1 loci were analyzed.

E2406 | www.pnas.org/cgi/doi/10.1073/pnas.1319962111 Ohki et al.

Page 4: PHLDA3 is a novel tumor suppressor of pancreatic ... · Pancreatic neuroendocrine tumors (PanNETs) are a rare pa-thology, and molecular mechanisms underlying their develop-ment have

enhanced expression of PHLDA3 in a 5-aza-C concentration-dependent manner. A similar result was obtained using a humanPanNET cell line A99 (Fig. 4F) and the mouse insulinoma cellline MIN6 that has very low PHLDA3 expression (SI Appendix,Fig. S3 A and B). These results indicate that methylation of thePHLDA3 gene affects PHLDA3 transcription levels. We thenanalyzed the methylation status of the PHLDA3 promoter inPanNET samples that had undergone LOH at the PHLDA3locus. As shown in Fig. 4G, methylation was not detected innormal pancreas or islets, whereas significant methylation wasdetected in all LOH+ samples (seven of seven) analyzed. Theseresults show that the PHLDA3 gene can undergo methylationin addition to LOH in PanNETs, indicating that a two-hit in-activation of the PHLDA3 gene may often occur. We also analyzed

LOH− samples and found detectable levels of methylation in two offour samples (SI Appendix, Fig. S4). Thus, the repression ofPHLDA3 expression in LOH− samples due to methylation mayalso contribute to tumor progression in PanNETs.

A

B

0

0.2

0.4

0.6

0.8

1

0 1000 2000 3000 4000 5000

Su

rviv

al r

ate

Days

MEN1 LOH - (n = 12)

MEN1 LOH + (n = 30)

p = 0.85

C

0

0.2

0.4

0.6

0.8

1

0 1000 2000 3000 4000 5000 6000

Su

rviv

al r

ate

Days

PHLDA3 LOH - (n = 14)

PHLDA3 LOH + (n = 31)

p = 0.11

(p<0.01, Fisher’s exact test)

grade 1or 2 grade 3 or 4PHLDA3 LOH + 10 21PHLDA3 LOH- 11 3

grade 1or 2 grade 3 or 4MEN1 LOH + 19 19MEN1 LOH- 4 3

D

(p=0.73, Fisher’s exact test)

Fig. 3. LOH at the PHLDA3 and MEN1 gene locus and progression of Pan-NETs. (A) LOH at the PHLDA3 gene locus and grade of PanNETs. The P valuewas calculated by Fisher’s exact test. (B) Kaplan–Meier plots of overall sur-vival of patients with PanNETs. Fourteen or 31 patients without or withPHLDA3 LOH were analyzed. Wilcoxon test was used to determine theP value. (C) LOH at the MEN1 gene locus and grade of PanNETs. (D) Kaplan–Meier plot for overall survival of patients with PanNETs. Twelve or 30patients without or with MEN1 LOH were analyzed as in B.

methyl + unmethylPHLDA3 locus

methylatedPHLDA3 locus

DL

D1

no

DN

A

H12

99

MD

A-M

B-4

68

LN

Cap

fully

met

hyl

ated

D

NA

lowPHLDA3

highPHLDA3

methyl + unmethyl

methyl

#1no

DN

A

#2 #3 #5fully

met

hyl

ated

DN

A

PanNET samples

#1 #2 #1 #2 #3 #4 #6 #7

pan

crea

s

isle

t

A

B

C

G

D

E

F

0 0.15-aza-CE

xpre

ssio

n o

f P

HL

DA

3 m

RN

A0.01

(μM)

- 0.10.015-aza-C

methyl + unmethylPHLDA3 locus

methylatedPHLDA3 locus

(μM) 0.3

0

0.5

1

1.5

2

2.5

0.3

0

1

2

3

4

5

6

7

8

0 0.15-aza-C

Exp

ress

ion

of

PH

LD

A3

mR

NA

0.01(μM)

- 0.10.015-aza-C

methyl + unmethylPHLDA3 locus

methylatedPHLDA3 locus

(μM)

DL

D1

H12

99

MD

A-M

B-4

68

LN

Cap

0

10

20

30

40

50

60

70

Exp

ress

ion

of

PH

LD

A3

mR

NA

normalPancreas

*p<0.05

Exp

ress

ion

of

PH

LD

A3

mR

NA

PanNET

- +PHLDA3

LOHPHLDA3

LOH

0

0.2

0.4

0.6

0.8

1

1.2

+386+1 +2994 +3677+831

-508 +944

ORF

08 9CpG island

region amplified by MSP

*p<0.05

*p<0.05**p<0.01

**

*

*

Fig. 4. PHLDA3 expression and promoter region methylation status inPanNETs. (A) PHLDA3 gene expression in PanNETs. Total RNAs were pre-pared from normal pancreas and PanNETs. RNA was pooled from 5 normalpancreases for the normal controls. RNA was isolated from PanNET sampleswith (10 samples) or without LOH (7 samples). Gene expression was quan-titated by RT-PCR and normalized to GAPDH. (B) Genomic organization ofPHLDA3 promoter region and location of CpG island. Wide lines indicateexonic regions of the PHLDA3 gene (+1 to +831 and +2994 to +3677). (C)PHLDA3 gene expression in cell lines. Gene expression was analyzed as inA. (D) DNA methylation of the PHLDA3 promoter. Genomic DNAs from theindicated cell lines were analyzed by methylation-specific PCR. Positions ofthe primers used in the assay are shown in B. Primers designed to amplifymethylated DNA (upper panel) or DNA with or without DNA methylation(lower panel) were used. Fully methylated DNA was used as a control. (E andF) 5-aza-C treatment of Lung NET H1299 cells (E) or PanNET A99 cells (F).Both genomic DNAs and total RNAs were isolated, and analyzed as in Dand A, respectively. (G) Methylation status of PHLDA3 promoter in normalpancreas, normal isolated islets and PanNETs (samples showing LOH atthe PHLDA3 locus was analyzed). Genomic DNAs were prepared and analyzedas in D.

Ohki et al. PNAS | Published online May 27, 2014 | E2407

MED

ICALSC

IENCE

SPN

ASPL

US

Page 5: PHLDA3 is a novel tumor suppressor of pancreatic ... · Pancreatic neuroendocrine tumors (PanNETs) are a rare pa-thology, and molecular mechanisms underlying their develop-ment have

PHLDA3 Controls Akt Activity, Cell Proliferation, and Apoptosis ofIslet Cells. PanNETs are derived from pancreatic islet endocrinecells. It is well known that, in islet β cells, Akt signaling playsa central role in promoting cell growth and inhibiting apoptosis (5).Therefore, in β cells, loss of PHLDA3 function may result in thehyperactivation of Akt oncogenic signaling, and thus lead to tumorprogression. To analyze the function of PHLDA3 in islet β cells, weexamined RIN and MIN6 cells, cell lines derived from pancreatic βcells (Fig. 5 A–E). Whereas RIN cells have detectable levels ofPHLDA3 expression, MIN6 has very low PHLDA3 expression (SIAppendix, Fig. S3A). We first used a gain-of-function approach toconfirm that PHLDA3 functions as a repressor of Akt in MIN6cells. As shown in Fig. 5A, expression of PHLDA3 resulted indecreased Akt activation levels and decreased phosphorylationof signaling molecules downstream of Akt. Similar results wereobtained using PHLDA3−/−mouse embryonic fibroblasts (MEFs, SIAppendix, Fig. S5). Next, we knocked down PHLDA3 expression inRIN cells using siRNA against PHLDA3 (Fig. 5 B and C), andobserved increased Akt activation and cell proliferation (Fig. 5 Dand E). We observed similar results in normal primary rat islet cells,

i.e., knockdown of PHLDA3 expression resulted in activation of Akt(with or without glucose stimulation) and significant enhancementof cell proliferation (Fig. 5 F–H and SI Appendix, Fig. S6). Next, weanalyzed the effect of PHLDA3 expression on the apoptosis of isletcells induced by Streptozotocin (STZ), a chemical that is particularlytoxic to insulin-producing β cells (14). We observed that althoughinhibition of PHLDA3 expression by siRNA was relatively poor inisolated rat islets (Fig. 5I), this knockdown significantly reduced thenumber of apoptotic cells caused by STZ treatment (Fig. 5J). Col-lectively, these results demonstrate that PHLDA3 controls Akt ac-tivity, cell growth, and the apoptosis of islet cells.

Development of Hyperplastic Islets in PHLDA3-Deficient Mice. Toanalyze the effect of PHLDA3 deficiency on islets in vivo, weexamined the pancreases of PHLDA3-deficient mice. Differencesin islet sizes were not detected in 3-mo-old PHLDA3+/+ orPHLDA3−/− mice (SI Appendix, Fig. S7). However, in 6-mo-oldPHLDA3−/− mice, we found significantly fewer islets that weresmaller than 0.01 mm2 and significantly more islets that were largerthan 0.01 mm2, compared with PHLDA3+/+ mice (Fig. 6 A and C).

B D

E G

H

PH

LD

A3

con

tro

l

siRNA

γ-ray - + - +PHLDA3β-actin

I

P-Akt

total Akt

PH

LD

A3

con

tro

l

siRNA

β-actin

Nu

mb

ers

of

Brd

U p

osi

tive

cel

ls

0

0.5

1

1.5

2

2.5

3

control PHLDA3siRNA

*p<0.05

F

0

0.5

1

1.5

2

2.5

P-A

kt/t

ota

l Akt

control PHLDA3siRNA

P-Akt- +glucose

cont

rol

total Akt

- +

PH

LDA

3siRNA

β-actin

P-A

kt/t

ota

l Akt

0

2

4

6

8

- +glucose - +

control PHLDA3siRNA

0

5

10

15

20

25

30

35

40

TU

NE

L p

osi

tive

cel

ls (

%)

STZ0.2 mg/ml

cont

rol

PH

LDA

3

cont

rol

PH

LDA

3

- +

**p<0.01

siRNA

C

J

0

0.5

1

1.5

2

Nu

mb

ers

of

Brd

U p

osi

tive

cel

ls

control PHLDA3siRNA

*p<0.05

A

0

0.2

0.4

0.6

0.8

1

1.2

Exp

ress

ion

of

PH

LD

A3

mR

NA

control PHLDA3siRNA

**p<0.01

control PHLDA3siRNA

Exp

ress

ion

of

PH

LD

A3

mR

NA

0

0.2

0.4

0.6

0.8

1

1.2**p<0.01

Exp

ress

ion

of

PH

LD

A3

mR

NA

0

0.5

1

1.5

2

STZ0.2 mg/ml

cont

rol

PH

LDA

3

cont

rol

PH

LDA

3

- +

siRNA

**p<0.01

*p<0.05

Ad

-Lac

Z

Ad

-PH

LD

A3

P-Akt

total Akt

P-Mdm2

P-S6

PHLDA3

β-actin

P-p70 S6K1.00 0.12

1.00 0.22

1.00 0.97

0

0.2

0.4

0.6

0.8

1

1.2

LacZ PHLDA3

P-A

kt/t

ota

l Akt

Fig. 5. Effect of PHLDA3 expression on Akt activity,cell proliferation and apoptosis of islet cells. (A) Effectof PHLDA3 expression on Akt activity in MIN6 cells.MIN6 cells were transduced with Ad-LacZ or Ad-PHLDA3 at a moi of 35, and harvested 30 h post-infection. Akt activation and phosphorylation of Aktdownstream signaling molecules were analyzed byWestern blotting and quantified by normalization tototal Akt levels (P-Akt) or by β-actin levels (P-p70 S6K,P-S6, P-Mdm2). (B and C) Efficiency of siRNA inhibitionof PHLDA3 expression in RIN cells. RIN cells weretransfected with control or PHLDA3 siRNAs. PHLDA3mRNA levels were analyzed 31 h posttransfection byquantitative RT-PCR, standardized against β-actin (B).PHLDA3 protein levels were also determined 48 hposttransfection by Western blotting (C), using cellssubjected to γ-ray irradiation (20 Gy) versus untreated.The γ-ray irradiated samples were included to helpidentify the band representing PHLDA3 protein(PHLDA3 is induced by p53 activation). (D) Effect ofPHLDA3 expression on Akt activation in RIN cells. RINcells were transfected as in B and Akt activation wasanalyzed by Western blotting 31 h posttransfection(left) and quantified by normalization to total Aktlevels (right). (E) Effect of PHLDA3 expression on RINcell proliferation. RIN cells were transfected as in B,and labeled with BrdU for 3 h and harvested 28 h posttransfection. BrdU positive cells were quantified byusing Ziva Ultrasensitive BrdU assay. (F) siRNA sup-pression of PHLDA3 expression in primary islet cells.Isolated primary islet cells were transfected with con-trol or PHLDA3 siRNA, harvested 30 h post trans-fection, and PHLDA3 mRNA levels were analyzed byquantitative RT-PCR as in B. (G) Effect of PHLDA3 ex-pression on Akt activation in primary islet cells. Cellswere transfected with siRNA as in F and 48 h post-transfection were treated with glucose (30 mM) for 20min. Levels of Akt activation were analyzed as inD. (H)Effect of PHLDA3 expression on primary islet cell pro-liferation. Cells were transfected as in F, labeled withBrdU for 4 h and harvested 30 h post transfection.BrdU positive cells were analyzed as in E. (I and J) Ef-fect of PHLDA3 expression on STZ-induced apoptosisof primary rat islets. Isolated islets were pooled fromthree rats, and transfected with control or PHLDA3siRNA. At 75 h posttransfection, islets were treatedwith STZ (20 μg/mL) for 30 min. Islets were then cul-tured overnight, and total RNAs were prepared andPHLDA3 expression was analyzed by quantitative RT-PCR as in B (I), or subjected to TUNEL staining andanalyzed by FACS (J).

E2408 | www.pnas.org/cgi/doi/10.1073/pnas.1319962111 Ohki et al.

Page 6: PHLDA3 is a novel tumor suppressor of pancreatic ... · Pancreatic neuroendocrine tumors (PanNETs) are a rare pa-thology, and molecular mechanisms underlying their develop-ment have

However, the overall average islet size at 6 mo was not significantlydifferent between PHLDA3+/+ and PHLDA3−/− mice (Fig. 6E). At10 mo, abnormally large islets (larger than 0.08 mm2) were fre-quently found (Fig. 6 B and D), and the average islet sizes weresignificantly larger in 10-mo-old PHLDA3+/− and PHLDA3−/− micecompared with PHLDA3+/+ mice (Fig. 6E). Thus, a difference inislet sizes between PHLDA3+/+ and PHLDA3−/− mice appears toemerge between 3 and 10 mo of age. We stained the islets with Ki67antibody and found significantly more Ki67-positive cells inPHLDA3−/− islets compared with the PHLDA3+/+ islets (Fig. 6F;representative images are shown in SI Appendix, Fig. S8). Thesedata show that loss of PHLDA3 expression results in enhancedproliferation of islet cells.

Enhanced Proliferation of β Cells and Altered Glucose Metabolism inPHLDA3-Deficient Mice. The islets were further stained with anti-insulin and anti-glucagon antibodies to determine the numbers

of β and α cells, respectively, within the islets. Normal murine isletshave β cells in the center of the islet and α cells at the peripherysurrounding the β cells (as shown in PHLDA3+/+ islets in Fig. 7A).However, in the hyperplastic islets of PHLDA3+/− and PHLDA3−/−

mice, huge numbers of β cells and relatively small numbers of α cellswere often observed, and the hyperplastic islets often showed ab-normal islet architecture, with few α cells at the periphery of theislets (Fig. 7A). We calculated the areas occupied by α and β celltypes to determine which of these had increased. As shown in Fig.7B, the mean percentage area occupied by β cells was significantlyhigher in PHLDA3−/− islets. The area occupied by β cells was alsosignificantly high in large PHLDA3−/− islets (larger than 0.05 mm2)compared with that seen in large islets infrequently found inPHLDA3+/+ mice (Fig. 7B). Because we had found a significantlyhigher number of proliferating cells in PHLDA3−/− islets (Fig. 6F),we stained the islets with anti-Ki67, -insulin, and -glucagonantibodies to identify which cell types within the islets wereproliferating. As shown in Fig. 7C, we found that most Ki67-

A

E

PHLDA3 +/+ PHLDA3 -/-B

C

PHLDA3 +/- PHLDA3 -/-PHLDA3 +/+

D

F

0

0.05

0.1

0.15

0.2

0.25

0.3

Ki6

7+ c

ells

/isle

t ce

lls (

%)

**p<0.01

+/+n=5

-/-n=5

/ /

300 μm

0 - 0.009 0.010 - 0.079

p va

lue

+/+ vs -/- 0.017 0.025 0.182

islet size (mm2)

Islet size (mm2)

30

40

50

60

70

80

90

0

PHLDA3 +/+ (n=17)

PHLDA3 +/− (n=16)

PHLDA3 −/− (n=17)

20

2

0

30

40

50

60

70

80

90

0 - 0.009 0.010 - 0.079 0.080

Islet size (mm2)

Isle

t p

erce

nta

ge

(%) *

*

PHLDA3 +/+ (n=6)PHLDA3 −/− (n=6)

20

2

0 - 0.009 0.010 - 0.079 0.080

*

0 - 0.009 0.010 - 0.079 0.080

+/+ vs +/- 0.004 0.002 0.035

+/+ vs -/- 0.024 0.027 0.045

+/- vs -/- 0.225 0.214 0.291

islet size (mm2)

p va

lue

*p<0.05**p<0.01

*

*

*

**

**

*p<0.05

n=17 n=16 n=17n=6 n=6

Ave

rag

e o

f is

let

size

(m

m2 )

*

0

0.003

0.006

0.009

0.012

0.015

0.018

+/+ -/- +/+ +/- -/-

6 M 10 M

*

Isle

t p

erce

nta

ge

(%)

*p<0.05

300 μm 300 μm 300 μm 300 μm

60 μm 60 μm 60 μm 60 μm 60 μm

0.080≥

p value

6 M 0.145

0.0280.043

0.20810 M

+/+ vs +/-+/+ vs -/-+/- vs -/-

+/+ vs -/-

Fig. 6. Hyperplastic islets in PHLDA3-deficient mice.Six-month-old (21- to 31-wk-old) (A, C, and E) and10-mo-old (39- to 49-wk-old) (B and D–F) mice wereanalyzed. (A and B) Hematoxylin and eosin stainingof islets from wild-type, heterozygote and PHLDA3-deficient mice (A, 6-mo; B, 10-mo). (C–E) Calculationof islet sizes. Islet areas were calculated from he-matoxylin and eosin-stained pancreas sections. One(6-mo-old mice) or three pancreas sections (10-mo-old mice; sections separated by more than 20 μm zaxis) were analyzed per mouse. Islet areas from in-dicated numbers (n) of mice were analyzed. Sizedistributions of islets (C, 6-mo; D, 10-mo) and aver-age islet sizes (E; 6- and 10-mo-old) are shown.The P values between PHLDA3+/+ and PHLDA3+/− orPHLDA3−/− were calculated and are shown (*P <0.05; **P < 0.01). (F) Quantitation of Ki67-positivecells. Pancreas sections from indicated numbers(n) of mice were analyzed. Islet nuclei number andKi67-positive nuclei number were counted and thepercentage of Ki67-positive cells was calculated.(**P < 0.01) Representative images are shown in SIAppendix, Fig. S4.

Ohki et al. PNAS | Published online May 27, 2014 | E2409

MED

ICALSC

IENCE

SPN

ASPL

US

Page 7: PHLDA3 is a novel tumor suppressor of pancreatic ... · Pancreatic neuroendocrine tumors (PanNETs) are a rare pa-thology, and molecular mechanisms underlying their develop-ment have

positive cells were β cells in PHLDA3−/− islets. Taken together,these data show that islet β cell proliferation is enhanced inPHLDA3-deficient islets.It would be expected that abnormal proliferation of β cells

would result in the excess production of insulin, leading to al-tered glucose metabolism in PHLDA3-deficient mice. As shown inFig. 7D, although the PHLDA3 genotype had no influence on bodyweights, 10-mo-old PHLDA3+/− and PHLDA3−/− mice had higherplasma insulin and lower blood glucose levels compared with age-matched wild-type mice under fed conditions. Similar results wereobtained in 6-mo-old fed mice (SI Appendix, Fig. S9A) and in 10-mo-old fasted mice (SI Appendix, Fig. S9B). Therefore, we performedglucose tolerance tests and found significant enhancement of glucosetolerance in PHLDA3+/− and PHLDA3−/− mice (Fig. 7E). Thesedata show that loss of PHLDA3 expression results in enhancedproliferation of islet cells, especially β cells, and consequentialincrease of insulin secretion.We further characterized PHLDA3−/− islets using several β cell

markers: the endocrine cell marker chromogranin A (SI Appendix,Fig. S10), the β cell differentiation marker Glut2 (SI Appendix,

Fig. S11), and deposition of IAPP (islet amyloid polypeptide; SIAppendix, Fig. S12). None of these markers showed significantdifference compared with PHLDA3+/+ islets, indicating thatPHLDA3−/− islets retain normal β cell characteristics, to theextent we examined.

PHLDA3-Deficient Islets Have Enhanced Akt Activity and Larger CellSize, and Are Resistant to Apoptosis.We further analyzed the effects ofPHLDA3 deficiency on several Akt-regulated biological processes.We first confirmed reduction or loss of PHLDA3 expression inPHLDA3+/− and PHLDA3−/− mice islets (Fig. 8A). We found thatactivation of Akt and phosphorylation of its downstream moleculeswere augmented in PHLDA3−/− islets (Fig. 8B). It has been reportedthat enhanced Akt activity has an effect on islet cell size (5, 6). Wetherefore analyzed islet cell size in PHLDA3+/+, PHLDA3+/−, andPHLDA3−/− mice. As shown in Fig. 8C, islet cell size was signifi-cantly increased in PHLDA3−/− and PHLDA3+/− mice. We nextanalyzed the sensitivity of PHLDA3−/− islets to apoptosis. Wild-typeor PHLDA3−/− mice were treated with STZ for 5 consecutive daysto produce β cell injury. It is known that STZ-induced apoptosis of β

PHLDA3 +/+(n=25)

PHLDA3 +/−(n=24)

PHLDA3 −/−(n=21)

A B

C

E

HEKi67Red; insulin

Green; glucagon

60 μm

PHLDA3 +/+

Bo

dy

wei

gt

(g)

n=27 n=32 n=25

p value0.3640.1230.155

Blo

od

glu

cose

leve

ls (

mg

/dL

)

p value0.0380.0030.228

n=27 n=32 n=25

**p<0.01

0

0.5

1

1.5

2

2.5

3

+/+ +/- -/-Pla

sma

insu

lin le

vels

(n

g/m

L)

p value0.0460.0340.265

*p<0.05 *p<0.05

n=21 n=24 n=21

0

50

100

150

200

250

300

350

400

450500

0 15 30 45 60 90 120post-fasting

**

**

****

**

****

**

*

*

Blo

od

glu

cose

leve

ls (

mg

/dL

)

Time (min)

*p<0.05**p<0.01

PHLDA3 +/- PHLDA3 -/-

D

0

5

10

15

20

25

30

35

40

45

+/+ +/- -/-

80

100

120

140

160

180

200

220

+/+ +/- -/-

*p<0.05

post-fasting 0 15 30 45 60 90 120

0.038 0.396 0.287 0.018 0.001 0.001 0.002 0.014

0.003 0.220 0.116 0.005 0.0001 0.00005 0.00001 0.0003

0.228 0.314 0.275 0.272 0.139 0.146 0.063 0.066

p value

80

84

88

92

96

100

+/+ -/- +/+ -/-

All islets 0.05 mm2

islets

β ce

lls a

rea

/ isl

et a

rea

(%)

n=182 n=257 n=24n=4

*

*

*p<0.05

>

15 μm 15 μm 15 μm

15 μm 15 μm 15 μm

+/+ vs +/-+/+ vs -/-+/- vs -/-

+/+ vs +/-+/+ vs -/-+/- vs -/-

+/+ vs +/-+/+ vs -/-+/- vs -/-

60 μm60 μm

+/+ vs +/-

+/+ vs -/-

+/- vs -/-

Fig. 7. Proliferation of β cells in the islets ofPHLDA3-deficient mice. Ten-mo-old (39- to 49-wk-old) mice were analyzed. (A) Distribution of β and αcells in pancreas sections. Pancreas sections werestained with antibodies to identify β cells (anti-in-sulin; red) and α cells (anti-glucagon; green). Rep-resentative images of hyperplastic islets fromPHLDA3+/− and PHLDA3−/− mice are shown. (B) Per-cent area occupied by β cells. Pancreas sections werestained as inA, and areas occupied by α- and β-cells inindicated numbers of islets from PHLDA3+/+ (fivemice were analyzed) and PHLDA3−/− (six mice wereanalyzed) were calculated and percentages of β-cellareas in the islets were determined. Semihyperplasicislets and hyperplastic islets (larger than 0.05 mm2

islet area) were separately calculated and shownat the right. (*P < 0.05; **P < 0.01). (C) Proliferationin pancreas sections. Serial pancreas sections ofPHLDA3−/− mice were stained with anti-insulin andanti-glucagon, anti-Ki67 and HE, to determine thecell types that are Ki67 positive. Representativeimages are shown. (D) Body weight, blood glucoseand insulin levels. Indicated numbers (n) of mice wereanalyzed. Plasma insulin levels and blood glucoselevels were determined in blood from tail vein sam-ples, and were determined twice for each mouse.Mice were fed ad libitum. (*P < 0.05; **P < 0.01). (E)Glucose tolerance test (GTT). Indicated numbers (n) ofovernight-fasted mice were subjected to GTT by i.p.injection of glucose (2 mg/g body weight). GTT wasperformed twice for each mouse. The P values be-tween PHLDA3+/+ and PHLDA3+/− or PHLDA3−/− werecalculated and are shown in the panels (*P < 0.05;**P < 0.01). The differences between PHLDA3+/− andPHLDA3−/− were not significant (P > 0.05).

E2410 | www.pnas.org/cgi/doi/10.1073/pnas.1319962111 Ohki et al.

Page 8: PHLDA3 is a novel tumor suppressor of pancreatic ... · Pancreatic neuroendocrine tumors (PanNETs) are a rare pa-thology, and molecular mechanisms underlying their develop-ment have

cell results in the elevation of blood glucose, and can thereby ex-perimentally induce type I diabetes in mice. We analyzed bloodglucose levels at the indicated times following STZ treatment, andSTZ administration resulted in the effective elevation of bloodglucose levels in wild-type mice, whereas elevation was diminished inPHLDA3−/− mice (Fig. 8D). When we calculated β cell and α cell

areas in STZ-treated mice, we observed a significant increase in thearea occupied by β cells in PHLDA3-deficient mice, showing thatPHLDA3 is required for the efficient induction of β cell apoptosis bySTZ (Fig. 8E). These results demonstrate that PHLDA3 controlsAkt activity, cell size, and apoptosis of islet cells in vivo.

DiscussionIn this report, we have shown that PHLDA3 is a candidate tumorsuppressor in PanNETs. We observed that the LOH of thePHLDA3 gene locus in human PanNETs occurs at a remarkablyhigh frequency and is comparable to that of the MEN1 genelocus, the gene reported to be most frequently affected in PanNETs(15). We also showed that LOH at the PHLDA3 gene locus isassociated with disease progression and poor prognosis inPanNETs. In contrast, LOH/mutation of MEN1 was foundto be unrelated to the prognosis of PanNET patients, both inthis and in a previous study (7). These results show that thePHLDA3-regulated tumor suppression pathway is relevant tothe progression and malignant phenotype of PanNETs and thepoor prognosis of PanNET patients. Because PHLDA3 is a re-pressor of Akt, we propose that inhibition of the Akt pathwaywould improve the prognosis of PanNET patients exhibitingLOH of PHLDA3. This possibility is supported by the reportedclinical efficacy of the Akt pathway inhibitor Everolimus, whichhas been shown to improve the survival of PanNET patientssignificantly. Everolimus treatment was approved in Japan onlyfrom 2011, and therefore none of the patients in our study hadreceived Everolimus. We speculate that patients who are positivefor PHLDA3 LOH and have a poor prognosis may benefitmost from Everolimus treatment. Furthermore, determination ofPHLDA3 LOH status could serve as a diagnostic measure toselect patients who should receive Everolimus. Thus, it will beimportant to conduct a prospective study to analyze the effect ofEverolimus in patients with or without PHLDA3 LOH.We have shown that PHLDA3 regulates Akt activity and

various Akt-regulated biological processes, i.e., cell proliferation,cell size, and apoptosis, in cultured cell lines and in vivo endo-crine cells. Several laboratories have generated transgenic micethat specifically express active Akt in β cells (6, 16). These micedeveloped hyperplastic islets, but did not develop malignantPanNETs (17). Likewise, we have shown that loss of PHLDA3and the resulting activation of Akt leads to hyperplastic isletdevelopment, but not to the development of PanNETs. AlthoughAkt activation is observed in various cancers and contributes totumorigenesis and tumor progression, the conclusion derivedfrom these experiments is that activation of Akt pathway alone isinsufficient to cause PanNETs unless combined with an onco-genic event in a second pathway. In human PanNETs, doubleLOH of PHLDA3 and MEN1 is frequently observed. In thesecells, loss of PHLDA3 function, i.e., activation of Akt, is com-bined with loss of MEN1 function to promote PanNET tumor-igenesis, and loss of PHLDA3 function particularly contributesto the progression of PanNETs. It would therefore be interestingto generate mice that are doubly deficient in PHLDA3 andMEN1 to analyze whether the islet phenotypes are more pro-nounced and the hyperplastic islets more prone to malignancy.We observed hyperplastic islets in PHLDA3 heterozygote

(+/−) mice as well as PHLDA3-deficient (−/−) mice. This ob-servation suggests that PHLDA3 is haplo-insufficient for thesuppression of endocrine cell proliferation. We have previouslyshown that PHLDA3 expression represses Akt activity in a dose-dependent manner, consistent with the possibility that loss ofa single PHLDA3 allele could lead to enhanced Akt activationand enhanced proliferation of endocrine cells (8). However, wehave also shown that in human PanNETs, the PHLDA3 locusundergoes methylation in addition to LOH, suggesting that twohits on the PHLDA3 gene is required for human PanNET de-velopment. We assume that loss of both PHLDA3 alleles, andconsequent stronger activation of Akt, may be required to exerta tumorigenic phenotype. Future studies should examine the isletphenotypes of PHLDA3+/− and PHLDA3−/− in a MEN1−/− back-

A B

C

D E

p value0.0140.0010.019

0

20

40

60

80

100

120

Isle

t ce

ll si

ze ( μ

m )2

n=5

*p<0.05**p<0.01

+/+ +/- -/-n=5 n=5

PHLDA3 -/-PHLDA3 +/+

20 μm

PHLDA3 +/-

20 μm 20 μm

PHLDA3 +/+ PHLDA3 -/-

Red; insulinGreen; glucagon

60 μm

Days

PHLDA3+/+ (n=11)

PHLDA3-/- (n=5)

Blo

od

glu

cose

leve

ls (m

g/d

L)

*

0

100

200

300

400

500

600

700

0 10 20 30 40 50 60 70

p<0.05

p<0.01**

*****

Exp

ress

ion

of

PH

LD

A3

mR

NA

0

0.2

0.4

0.6

0.8

1

1.2

+/+ +/- -/-

60 μm

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1.6

1.8

+/+ -/-

P-A

kt/t

ota

l Akt

+/+ -/-

P-Akt

total Akt

P-Mdm2

P-Gsk3β

P-S6

P-p70 S6K

β-actin

1.00

1.00

1.00

1.00

1.00 1.00

2.99

1.41

1.41

1.32

**p<0.01

+/+ -/-

Are

a (m

m2 )

*p<0.05

n=3 n=3

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7 α cellβ cell

+/+ vs +/-+/+ vs -/-+/- vs -/-

Fig. 8. Akt activation in islets of PHLDA3-deficient mice. (A) PHLDA3 mRNAexpression in isolated islets. Islets were isolated from PHLDA3+/+, PHLDA3+/−,and PHLDA3−/− mice, and analyzed for PHLDA3 expression by quantitativeRT-PCR, normalized to β-actin. (B) Akt activity and phosphorylation of Aktdownstream signaling molecules in isolated islets. Akt activation and phos-phorylation of Akt downstream signaling molecules were analyzed byWestern blotting and quantified by normalization to total Akt levels (P-Akt,Right) or by β-actin levels (P-p70 S6K, P-S6, P-GSK3β, and P-Mdm2). (C) Isletcell size in PHLDA3-deficient mice. (Left) Indirect immunofluorescencestaining of Glut2 in pancreas section from 10-mo-old mice. (Right) Islet areasand nuclei numbers were calculated, and mean islet cell sizes were de-termined from one hematoxylin- and eosin-stained pancreas sectionsper indicated numbers (n) of mice. The P values between PHLDA3+/+ andPHLDA3+/− or PHLDA3−/− were calculated and shown in the panels (*P <0.05; **P < 0.01). The difference between PHLDA3+/− and PHLDA3−/− wasalso significant (P < 0.05). (D) Blood glucose levels in streptozotocin-induceddiabetic mice. Indicated numbers (n) of PHLDA3+/+ or PHLDA3−/− mice wereinjected i.p. with STZ for 5 consecutive days. Blood glucose levels were de-termined at different time points as indicated after administration of STZ.(*P < 0.05; **P < 0.01). (E) Distribution of β and α cells in STZ-treatedPHLDA3+/+ and PHLDA3−/− mice. (Left) Sections were stained with antibodyagainst insulin (β cell marker; red) and glucagon (α cell marker; green)and representative images are shown. Pancreas sections of three PHLDA3+/+

and three PHLDA3−/− mice were analyzed. (Right) For each mouse, α-cell andβ-cell areas were calculated from one pancreas section. (*P < 0.05.)

Ohki et al. PNAS | Published online May 27, 2014 | E2411

MED

ICALSC

IENCE

SPN

ASPL

US

Page 9: PHLDA3 is a novel tumor suppressor of pancreatic ... · Pancreatic neuroendocrine tumors (PanNETs) are a rare pa-thology, and molecular mechanisms underlying their develop-ment have

ground. We expect that, in a MEN1−/− background, PHLDA3−/−

mice would develop PanNETs more frequently than in PHLDA3+/−

mice. In addition, because we found association of LOH at thePHLDA3 locus in both nonfunctional and functional humanPanNETs, whether nonfunctional PanNETs develop in thesemice is an interesting issue.We previously identified PHLDA3 as a p53 target gene. In the

present study, we show that the PHLDA3 locus undergoes fre-quent LOH in PanNETs, a tumor type in which p53 mutationsare rare (3, 7). Recently it has been reported that among thevarious target genes of p53, PHLDA3 displays prominent tumorsuppressor activity (18). Therefore, in tumors harboring a p53mutation, loss of p53-regulated PHLDA3 expression may sig-nificantly contribute to tumor progression, whereas in tumorswithout a p53 mutation, loss of the PHLDA3 gene itself maydrive oncogenesis. Interestingly, global methylation profiling ofprostate cancer specimens has revealed significant methylationof the PHLDA3 gene in these cancers (19). In addition, in theCOSMIC database, 11 PHLDA3 mutations in several cancersare reported. All mutations are located within the PH domain ofPHLDA3, and nonsynonymous mutations may result in loss ofAkt repressing ability. Therefore, in addition to inactivation bymethylation, these and other mutations that result in a functionalloss of PHLDA3 may contribute to some cancers.In summary, our results show that PHLDA3 is a novel tumor

suppressor of PanNET, and PHLDA3 and MEN1 cooperativelysuppress its development. PHLDA3 represses Akt activity in isletcells and hyperplastic islets are found in both PHLDA3+/− andPHLDA3−/− mice. Collectively, our data illustrates the importanceof the PHLDA3-regulated tumor suppression pathway in PanNETs.

Materials and MethodsCell Lines, Cell Culture, Transfection, Adenovirus Infection, and Soft AgarColony Formation Assay. Cell lines used in this studywere: LNCaP (humanprostatecancer), MDA-MB-M468 (human breast cancer), DLD1 (human colorectal cancer),H1299 (human lung NET), A99 (human PanNET, ref. 20), RIN (rat pancreatic β cell),and MIN6 (mouse pancreatic β cell). Cell culture and transfection was performedas described (21). The siRNAs were introduced using RNAiMAX (Invitrogen). ON-target plus control and ON-target plus PHLDA3-targeting siRNAs were purchasedfrom Dharmacon Research. Adenovirus infection was performed using previouslydescribed adenoviruses expressing LacZ or N-terminally HA-tagged PHLDA3 (8).Everolimus (AdooQ BioScience) was added to cultures at the indicatedconcentrations. For soft agar colony formation assays, cells were seeded in 3-cm dishes with a bottom layer of 0.5% agarose and a top layer of 0.33%agarose, both in complete media. The assay was performed in triplicate foreach sample. Colonies were photographed after 9 (H1299) or 10 (MIN6) daysof incubation. Colonies were counted in three to five different views fromeach plate to calculate average values. The total numbers of colonies largerthan 100 (MIN6) or 200 (H1299) pixels were counted per view.

Western Blotting Analysis. Cells were lysed in lysis buffer [50 mM Tris·HCl (pH8.0), 1% Nonidet P-40, 250 mM NaCl, 50 mM NaF, 1 mM Na3VO4, 1 mMprotease inhibitor (PMSF, aprotinin, leupeptin) and 1 mM DDT]. Whole celllysates were subjected to protein quantification and analyzed by Westernblotting. To detect PHLDA3, 20 μg of whole cell lysates (WCL) were loaded.To detect other proteins, 5 μg of WCL were loaded. Antibodies used in thisstudy were: anti-Akt rabbit polyclonal antibody, anti-phospho-Akt (S473)rabbit monoclonal antibody, anti-p70S6K rabbit monoclonal antibody, anti-phospho-p70S6K (T389) rabbit polyclonal antibody, anti-phospho-S6 (S240/244) rabbit monoclonal antibody, anti-phospho-Gsk3β (S9) rabbit polyclonalantibody, anti-phospho-Mdm2 (S166) rabbit polyclonal antibody from CellSignaling Technology, anti-actin mouse monoclonal antibody from SIGMAfrom Santa Cruz Biotechnology, anti-PHLDA3 goat polyclonal antibody fromAbcam, anti-HA monoclonal antibody (clone 12CA) from Roche Diagnostics.

Reverse Transcription and Real-Time PCR. Reverse transcription was carried outusing kits from Invitrogen following the manufacturer’s instructions (SuperScriptFirst-Strand Synthesis System for RT-PCR). Total RNA (0.2–5 μg) was usedfor reverse transcription. Reverse-transcribed cDNAs were subjected to real-time PCR, which was performed with a LightCycler 480 Instrument (RocheDiagnostics). For the detection of PHLDA3 (human; Hs00385313_m1, mouse;Mm00449846_m1, rat; rat; Rn01483684_m1), beta actin (mouse; Mm00607939_s1,

rat; Rn00667869_m1) and GAPDH (human; Hs02758991_g1, mouse;Mm99999915_g1), TaqMan probe from Applied Biosystems was used.

BrdU Incorporation Assay and Measurement of Cell Number. BrdU-positive cellswere quantified using the Ziva Ultrasensitive BrdU assay (Jaden BioScience).This assay detects the incorporation of BrdU using a labeled anti-BrdU an-tibody and detection substrate in an ELISA format. By this assay, a smallfraction of proliferating cells within a large population of nonproliferatingcells can be detected. Cell numbers were analyzed by using CellTiter-GloLuminescent Cell Viability Assay (Promega). Using this assay, viable cells aredetermined by quantitation of ATP, an indicator of metabolically active cells.

Flow Cytometry and TUNEL Assay. Apoptosis of islet cells was quantitated bythe TUNEL reaction using the In Situ Cell Death Detection kit, Fluorescein(Roche Diagnostics) followed by flow cytometry. Flow cytometry analysis wasperformed using a FACS Calibur instrument (Becton Dickinson).

Tumor Samples Used in the Study, and DNA and RNA Extraction from PrimaryTumor Samples. The tumor samples used in this study were surgically resectedat the National Cancer Center Hospital (44 samples; PanNET1-18, 24–47, 52,53), Kyoto University Hospital (6 samples; PanNET19-23, 51), or KagawaUniversity Hospital (4 samples; PanNET 48–50, 54) between 1993 and 2012. His-tological grading of the tumors were determined based on the classification ofWorld Health Organization 2004. This study was approved by the InstitutionalReview Board of the National Cancer Center, Tokyo. Clinical and pathologicaldata were obtained through a detailed retrospective review of the medicalrecords of all patients with PanNET. Five-micrometer sections of paraffin-em-bedded tissues were subjected to DNA extraction. Total RNA was extracted fromfrozen normal pancreas and tumor samples using an RNeasy mini kit (Qiagen).

Microsatellite Analysis. Microsatellite analysis was performed basically asdescribed (8). We used six primer pairs labeled with FAM that amplifymicrosatellite loci to achieve accurate detection of LOH at the PHLDA3 locus(Fig. 1A). For the MEN1 locus, three primer pairs were used (Fig. 2A). Am-plified PCR products were analyzed with a 3100 automated sequencer(Applied Biosystems). Collected data were analyzed with GeneScan andGenotyper software (Applied Biosystems), and allele sizes and peak heightswere calculated. The genotype was determined to be heterozygous if twobands of different sizes were obtained from normal tissues. A ratio of the twopeaks in tumor DNA of less than 0.7 in comparison with the correspondingratio of the two peaks in nontumor DNA was considered as allelic loss.

5-aza-dC Treatment and Methylation-Specific PCR. Cells were seeded at adensity of 3 × 105 cells (H1299 and A99) or 3 × 106 (MIN6) cells per 10-cm dishon day 0 and treated with freshly prepared 5-aza-dC (Sigma-Aldrich) for 24 hon days 1, 3, and 5. After each treatment, cells were placed in fresh mediumand harvested on day 6. Genomic DNAwas extracted and subjected to bisulfiteconversion using EZ DNA Methylation kit (Zymo research). Fully methylatedcontrols were prepared by methylating genomic DNA with SssI methylase(New England Biolabs). Methylation-specific PCR (MSP) was performed basi-cally as described (22). MSP was performed using the specific primer setsshown below. BS-F and R primers were used to amplify DNA with or withoutmethylation. MSP-F and R primers were used to amplify methylated DNA.

BS-F: GTAGATAGAGTTTAGGGGAGTAAGAG

BS- R: CTCTACCCCAACTAACCCAACCC

MSP-F: GAGGGTTGGTTAGGGTAGGAATGTG

MSP-R: ACTCCCCTAAACTCTATCTACACAC

Mice Used in This Study. Generation of PHLDA3-deficient mice was reported(23). Briefly, a PHLDA3neo targeting vector was obtained by cloning a 6-kbupstream KpnI fragment and a 6-kb downstream HindIII/NotI fragment intopPNT1. The resulting deletion (nucleotides 579–2,096 of GenBank accessionno. AF151099) eliminates all of exon 1 and part of exon 2, thus deleting theentire PHLDA3 coding region. PHLDA3−/− mice were generated by crossingthe heterozygotes. MEFs were isolated and maintained as described (24).Mouse experiments were performed in a specific pathogen-free environ-ment at the National Cancer Center animal facility according to institutionalguidelines, and all of the animal experiments were approved by the Com-mittee for Ethics in Animal Experimentation at the National Cancer Center.

Blood Glucose, Plasma Insulin Measurement, and Glucose Tolerance Tests.Blood glucose levels were determined with blood samples from tail vein

E2412 | www.pnas.org/cgi/doi/10.1073/pnas.1319962111 Ohki et al.

Page 10: PHLDA3 is a novel tumor suppressor of pancreatic ... · Pancreatic neuroendocrine tumors (PanNETs) are a rare pa-thology, and molecular mechanisms underlying their develop-ment have

punctures in mice using Glucose pilot (Aventir Biotech), according to theprocedures specified by the manufacturer. Plasma insulin levels were de-termined with blood samples from tail vein punctures or inferior vena cavain mice by ELISA (Morinaga Institute of Biological Science), according to theprocedures specified by the manufacturer. For glucose tolerance tests, micewere fasted overnight and blood was drawn from tail vein at 0, 15, 30, 45, 60,90, and 120 min after i.p. injection of D-glucose (2 mg/g of body weight).

Quantitative Measurement of Islet Morphology. Islet area and islet nucleinumber were measured from hematoxylin and eosin-stained pancreas sec-tions and Ki67-positive cells were counted from immunohistochemicallystained pancreas sections using TissueFAXS (TissueGnostics). Chromogranin Asignal intensity was also measured using TissueFAXS. α and β cells areas werequantified by the number of pixels in each immunohistochemically stainedarea in images taken by fluorescence microscopy (Olympus IX2-DSU).

Isolation of Rat and Mouse Primary Islets and Preparation of Primary Islet Cells.The animal experiment was reviewed and approved by the InstitutionalAnimal Care and Use Committee for Frontier Medical Sciences, Kyoto Uni-versity. For isolation of rat islets, Lewis or Wistar rats (male, aged 9–11 wk,Shimizu Laboratory Supplies) were used. Islet isolation was performedaccording to previously described methods (25). Briefly, through midlinelaparotomy, 10 mL of a type XI collagenase solution (1200 CDU/mL, C9407,Sigma-Aldrich) was infused into the common bile duct that was ligated atthe hepatic side before the inflow into the duodenum. The pancreases wereremoved and digested in a water bath set at 37 °C for 18 min. The digestedpancreases were filtered with a stainless steel sieve to separate the islets,and purified using a discontinuous gradient solution (Dextran 70, 17–0280-02, Amersham). Mouse islets were isolated from 10- to 25-wk-old maleanimals by collagenase digestion of the pancreas, followed by purificationusing a Ficoll gradient. Islets were handpicked twice. The harvested isletswere cultured in RPMI or CRML-1066 medium (11530, Gibco) supplementedwith a 1% antibiotic–antimycotic solution (15240-062, Gibco) and 10%(vol/vol) FBS (12103-78P, JRH) in an incubator set at 5% (vol/vol) CO2, 37 °C.

Primary islet cells were prepared by digesting the islets with Accutase for15 min at 37 °C. Islet cells were washed with RPMI before use in experiments.

Immunohistochemistry. Immunohistochemistry (IHC) was performed basicallyaccording to the manufacturer’s instructions. In brief, after deparaffiniza-tion, tissues sections underwent antigen retrieval by autoclaving slides for5 min in 10 mM citrate buffer (pH 6.0). For fluorescent immunohistochemicalstaining of insulin, glucagon, and Glut2, nonspecific interactions wereblocked for 30 min using a 5% (vol/vol) goat serum solution. The primaryantibodies were: guinea pig anti-insulin polyclonal antibody (Abcam) diluted1:400, mouse anti-glucagon monoclonal antibody (Sigma-Aldrich) diluted1:750 and rabbit anti-Glut2 polyclonal antibody (Alpha Diagnostic) di-luted 1:750 with Signal Enhancer HIKARI (Nacalai Tesque). These were ap-

plied to the slides and incubated overnight at 4 °C. As secondary antibodies,Alexa Fluor 488 goat anti-mouse IgG antibody (Invitrogen) diluted 1:500,Alexa Fluor 488 goat anti-rabbit IgG antibody (Invitrogen) diluted 1:500 andAlexa Fluor 546 goat anti-guinea pig IgG antibody (Invitrogen) diluted1:1,000 with PBST-BSA were applied to the slides and incubated 3 h at roomtemperature. To detect Ki67- and Chromogranin A-positive cells, sectionswere pretreated with 0.3% H2O2 for inactivation of endogenous peroxidase.The primary antibody, rat anti-Ki67 monoclonal antibody (DakoCytomation)diluted 1:200, or rabbit anti-Chromogranin A polyclonal antibody (ThermoScientific) diluted 1:200 with Signal Enhancer HIKARI were applied to theslides and incubated overnight at 4 °C. As secondary antibodies, HistofineSimple Stain MAX PO anti-rat IgG antibody (Nichirei Bioscience) or bio-tinylated anti-rabbit IgG antibody (VECTOR Laboratories) was used. We used3,3′-diaminobenzidine tetrahydrochloride (DAB; Muto Pure Chemicals) as thesubstrate chromogen. The sections were counter stained with hematoxylin.

STZ-Induced Diabetes. Eleven- to 22-wk-old wild-type and PHLDA3 knockoutmale mice were injected i.p. with 50 mg/kg streptozotocin daily for 5 con-secutive days (Sigma-Aldrich) to produce β cell injury. On days 92, 93, 99, and100, animals were killed.

Statistical Analysis. Datawere calculated and shown asmean± SD (for Figs. 4, 5and SI Appendix, Fig. S3) or as mean ± SEM (Figs. 6–8 and SI Appendix, Figs. S7,S9, and S10A). Comparisons between the samples were performed by Studentt test. Survival data were analyzed using XLStat software (version 2013.4.05;Addinsoft), and Kaplan–Meyer plots were drawn. Wilcoxon test was per-formed to assess the statistical significance of the difference betweenthe survival curves. In Fisher’s exact test, P values were obtained by usingtwo tails. Statistical significance was defined as P < 0.05.

ACKNOWLEDGMENTS. We thank T. Niwa and T. Ushijima (National CancerCenter, Japan) for advice on methylation-specific PCR, Marc Lamphier forcritical reading of the manuscript, Dr. Wanxing Cui for providing isolatedhuman islets, and Drs. T. Yoshida and H. Sakamoto and National CancerCenter Research Core Facility (supported by National Cancer Center Researchand Development Fund, 23-A-7) for the LOH analyses in this study. This studywas supported by Grants-in-Aid for Scientific Research from the Ministry ofEducation, Culture, Sports, Science and Technology of Japan (23501279 and26430133, to R.O.); New Energy and Industrial Technology DevelopmentOrganization (NEDO) (09A02012a, to R.O.); research grants from Daiichi-Sankyo Foundation of Life Science (to R.O.); the Ichiro Kanehara Foundation(R.O.); Takeda Science Foundation (R.O.); Astellas Foundation for Researchon Metabolic Disorders (R.O.); Foundation for Promotion of Cancer Research inJapan (R.O.); Extramural Collaborative Research Grant of Cancer ResearchInstitute, Kanazawa University, Japan (to R.O.); Cooperative Research Pro-gram of Institute for Frontier Medical Sciences, Kyoto University, Japan(R.O.); and grants from the National Cancer Center Research and Develop-ment Fund (23-B-9, to R.O.; and 23-A-11, to T.T.).

1. Hauso O, et al. (2008) Neuroendocrine tumor epidemiology: Contrasting Norway andNorth America. Cancer 113(10):2655–2664.

2. Yao JC, et al. (2008) One hundred years after “carcinoid”: Epidemiology of andprognostic factors for neuroendocrine tumors in 35,825 cases in the United States.J Clin Oncol 26(18):3063–3072.

3. de Wilde RF, Edil BH, Hruban RH, Maitra A (2012) Well-differentiated pancreaticneuroendocrine tumors: From genetics to therapy. Nat Rev Gastroenterol Hepatol9(4):199–208.

4. Yao JC, et al.; RAD001 in Advanced Neuroendocrine Tumors, Third Trial (RADIANT-3)Study Group (2011) Everolimus for advanced pancreatic neuroendocrine tumors.N Engl J Med 364(6):514–523.

5. Elghazi L, Bernal-Mizrachi E (2009) Akt and PTEN: Beta-cell mass and pancreas plas-ticity. Trends Endocrinol Metab 20(5):243–251.

6. Tuttle RL, et al. (2001) Regulation of pancreatic beta-cell growth and survival by theserine/threonine protein kinase Akt1/PKBalpha. Nat Med 7(10):1133–1137.

7. Jiao Y, et al. (2011) DAXX/ATRX, MEN1, and mTOR pathway genes are frequentlyaltered in pancreatic neuroendocrine tumors. Science 331(6021):1199–1203.

8. Kawase T, et al. (2009) PH domain-only protein PHLDA3 is a p53-regulated repressorof Akt. Cell 136(3):535–550.

9. Yang YM, et al. (2005) Chromosome 1q loss of heterozygosity frequently occurs in sporadicinsulinomas and is associated with tumor malignancy. Int J Cancer 117(2):234–240.

10. Chen YJ, Vortmeyer A, Zhuang Z, Huang S, Jensen RT (2003) Loss of heterozygosity ofchromosome 1q in gastrinomas: Occurrence and prognostic significance. Cancer Res63(4):817–823.

11. Corbo V, et al. (2010) MEN1 in pancreatic endocrine tumors: Analysis of gene andprotein status in 169 sporadic neoplasms reveals alterations in the vast majority ofcases. Endocr Relat Cancer 17(3):771–783.

12. Yoo NJ, Kim YR, Lee SH (2011) Expressional and mutational analysis of PHLDA3 genein common human cancers. Pathology 43(5):510–511.

13. Brenet F, et al. (2011) DNA methylation of the first exon is tightly linked to tran-scriptional silencing. PLoS ONE 6(1):e14524.

14. Lenzen S (2008) The mechanisms of alloxan- and streptozotocin-induced diabetes.Diabetologia 51(2):216–226.

15. Pannett AA, Thakker RV (1999) Multiple endocrine neoplasia type 1. Endocr RelatCancer 6(4):449–473.

16. Bernal-Mizrachi E, Wen W, Stahlhut S, Welling CM, Permutt MA (2001) Islet betacell expression of constitutively active Akt1/PKB alpha induces striking hypertrophy,hyperplasia, and hyperinsulinemia. J Clin Invest 108(11):1631–1638.

17. Vivanco I, Sawyers CL (2002) The phosphatidylinositol 3-Kinase AKT pathway inhuman cancer. Nat Rev Cancer 2(7):489–501.

18. Brady CA, et al. (2011) Distinct p53 transcriptional programs dictate acute DNA-damage responses and tumor suppression. Cell 145(4):571–583.

19. Mahapatra S, et al. (2012) Global methylation profiling for risk prediction of prostatecancer. Clin Cancer Res 18(10):2882–2895.

20. Yachida S, et al. (2011) Establishment and characterization of a new cell line, A99,from a primary small cell carcinoma of the pancreas. Pancreas 40(6):905–910.

21. Ozeki C, et al. (2011) Cancer susceptibility polymorphism of p53 at codon 72 affectsphosphorylation and degradation of p53 protein. J Biol Chem 286(20):18251–18260.

22. Yamashita S, Tsujino Y, Moriguchi K, Tatematsu M, Ushijima T (2006) Chemical genomicscreening for methylation-silenced genes in gastric cancer cell lines using 5-aza-2′-deoxycytidine treatment and oligonucleotide microarray. Cancer Sci 97(1):64–71.

23. Frank D, et al. (2002) Placental overgrowth in mice lacking the imprinted gene Ipl.Proc Natl Acad Sci USA 99(11):7490–7495.

24. Ohki R, et al. (2000) Reprimo, a new candidate mediator of the p53-mediated cellcycle arrest at the G2 phase. J Biol Chem 275(30):22627–22630.

25. Yang KC, et al. (2010) The cytoprotection of chitosan based hydrogels in xenogeneicislet transplantation: An in vivo study in streptozotocin-induced diabetic mouse.Biochem Biophys Res Commun 393(4):818–823.

Ohki et al. PNAS | Published online May 27, 2014 | E2413

MED

ICALSC

IENCE

SPN

ASPL

US