Men1 maintains exocrine pancreas homeostasis in response to inflammation and oncogenic stress

Amanda R. Wasylishena, Chang Suna,b, Gilda P. Chaua, Yuan Qic, Xiaoping Suc, Michael P. Kimd, Jeannelyn S. Estrellae, and Guillermina Lozanoa,b,1

aDepartment of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030; bGenetics and Epigenetics Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030; cDepartment of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030; dDepartment of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030; and eDepartment of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030

Contributed by Guillermina Lozano, January 30, 2020 (sent for review November 14, 2019; reviewed by Zobeida Cruz-Monserrate and Tomoo Iwakuma) A more comprehensive understanding of the molecular mechanisms (Pdx1-Cre) (13) collectively results in islet cell hyperplasia and underlying pancreatic diseases, including pancreatitis and cancer, is PanNET development, with the majority of tumors being essential to improve clinical management. MEN1 has established roles functional insulinomas. in epigenetic regulation and tumor suppression in the endocrine Mechanistically, MEN1 is part of a large macromolecular Set- pancreas; however, intriguing recent data suggest MEN1 may also domain histone methyltransferase complex, containing MLL2, function in the exocrine pancreas. Using physiologically relevant ASH2L, RBBP5, and WDR5 (14). This complex mediates the genetic mouse models, we provide direct evidence that Men1 is methylation of histone 3 on lysine 4, and directly activates the ex- essential for exocrine pancreas homeostasis in response to inflam- pression of important cyclin-dependent kinase inhibitors CDKN1B mation and oncogenic stress. Men1 loss causes increased injury and (p27KIP1)andCDKN2C (p18INK4c) (15). Combined knockout of impaired regeneration following acute caerulein-induced pancreatitis, Cdkn1b and Cdkn2c in mice indeed results in a tumor spectrum leading to more severe damage, loss of the normal acinar compart- similar to MEN1 syndrome with tumors developing in the pituitary, ment, and increased cytokeratin 19-positive metaplasias and immune adrenal, thyroid, parathyroid, and pancreas, suggesting that loss of cell infiltration. We further demonstrate the Men1 is stabi- regulation of these is functionally contributing to tumori- lized in response to insult, and loss of Men1 is associated with the genesis upon loss of Men1 (16). The other well-established tran- overexpression of proinflammatory Jund target genes, suggesting scriptional targets of MEN1/MLL are the clustered homeobox that loss of Men1-mediated repression of Jund activity is, at least in genes, Hoxa9, Hoxc6,andHoxc8 (14, 17, 18), which are func- part, responsible for the impaired response. Finally, we demonstrate tionally important in the hematopoietic system. In this context that Men1 loss significantly accelerates mutant Kras-dependent onco- MEN1 is oncogenic and is an essential cofactor in leukemias with genesis. Combined, this work establishes Men1 as an important me- translocations that produce MLL fusion (19). The diator of pancreas homeostasis in vivo. MEN1:MLL interaction is currently being pursued as a thera- peutic target in these cancers. In addition to this epigenetic acute pancreatitis | Men1 | Jund | MLL function, MEN1 interacts with other transcription factors to modulate their activity, although the functional significance of ancreas diseases represent a significant medical burden. The these interactions is not well characterized in vivo. Of note, MEN1 Pincidence of acute pancreatitis is increasing and is attributed specifically associates with and inhibits the transcriptional activity to growing rates of obesity and gallstones, and is a significant cause of hospitalizations (1, 2). Chronic pancreatitis is associated Significance with pancreatic ductal adenocarcinoma, which is one of the most deadly human cancers with a 5-y survival rate of only 9% (3). A Understanding the molecular mechanisms that maintain pan- more comprehensive view of the molecular mechanisms that creas homeostasis in response to inflammatory stress is an maintain normal pancreas homeostasis is fundamental to un- urgent area of study due to the strong associations between derstanding dysfunction and disease pathogenesis. pancreatitis and cancer and diabetes. Through the use of MEN1 (multiple endocrine neoplasia 1) is a well-established physiologically relevant mouse models, we identify an essen- tumor suppressor in the endocrine pancreas. Rare germline tial role for Men1 in the normal tissue response to both in- mutations, generally nonsense or frameshift alterations, define flammatory and oncogenic stress. Loss of Men1 induces a the autosomal-dominant cancer predisposition syndrome, mul- proinflammatory expression program, associated with tiple endocrine neoplasia type 1 (MEN1) (4, 5). This syndrome is deregulation of Jund transcriptional activity. These results characterized by the presence of multiple tumors in the endo- provide insights into a regulatory axis that maintains pancreas crine pancreas, parathyroid glands, and anterior pituitary, with homeostasis in vivo. additional tumor types occurring in some patients. Consistent with these germline observations, somatic MEN1 mutations have Author contributions: A.R.W., M.P.K., and G.L. designed research; A.R.W., C.S., G.P.C., and also been identified in sporadic human tumors, including pan- J.S.E. performed research; A.R.W., Y.Q., X.S., M.P.K., J.S.E., and G.L. analyzed data; and A.R.W. and G.L. wrote the paper. creatic neuroendocrine tumors (PanNETs) and parathyroid tu- – Reviewers: Z.C.-M., The Ohio State University Wexner Medical Center; and T.I., University mors (6 8). Mouse models have further demonstrated that Men1 of Kansas Medical Center. loss is sufficient to promote tumorigenesis. While homozygous The authors declare no competing interest. deletion of Men1 results in embryo lethality at embryonic day (E) Published under the PNAS license. 11.5 to 12.5, germline heterozygous mice faithfully mimic the Data deposition: RNA-seq data reported in this paper have been deposited in the Gene human syndrome (9). Mice develop hyperplasias, and insulin- Expression Omnibus (GEO) database, https://www.ncbi.nlm.nih.gov/ (accession no. producing islet cell tumors as well as parathyroid adenomas as GSE145175). early as 9 mo of age, and tumors involving the thyroid, adrenal 1To whom correspondence may be addressed. Email: [email protected]. cortex, and pituitary by 16 mo of age. Conditional homozygous This article contains supporting information online at https://www.pnas.org/lookup/suppl/ deletion of Men1 from the β-cells (RIP-Cre) (10), α-cells (Glu- doi:10.1073/pnas.1920017117/-/DCSupplemental. Cre) (11, 12) or generally from all epithelial cells in the pancreas First published March 10, 2020.

6622–6629 | PNAS | March 24, 2020 | vol. 117 | no. 12 www.pnas.org/cgi/doi/10.1073/pnas.1920017117 Downloaded by guest on September 28, 2021 of JUND, a member of the AP1 family (20). This association is pancreatitis, Men1 is also important for the physiological response specific to JUND and not other AP1 family members, including to oncogenic stress, as Men1 loss cooperates with mutant Kras to JUN, JUNB, and FOS. accelerate exocrine pancreas tumorigenesis. While MEN1 clearly plays an essential tumor suppressor role in the endocrine pancreas, there are intriguing data suggesting that Results MEN1 may also play a functional role in the exocrine pancreas. A Men1 Deficiency Impairs Tissue Recovery following Acute Pancreatitis. recent case report identified a concurrent PanNET and a pancre- The pancreas is a remarkably regenerative organ. An intact re- atic ductal adenocarcinoma (PDAC) in one patient with MEN1 sponse to and recovery from stress or insult are essential for syndrome (21). Moreover, decreased expression and promoter normal tissue homeostasis of the pancreas, and prolonged injury methylation of MEN1 have been reported in PDAC (22). We were or damage have been linked to diabetes and tumorigenesis (3, 23– therefore interested to directly evaluate the role of Men1 in the 25). To evaluate the function of Men1 in this context, we used the exocrine pancreas. previously established conditional model of Men1 deficiency Using a physiologically relevant mouse model to genetically driven by the Pdx1-Cre transgene (10, 13) (Men1fl/flPdx1-CreTg, ablate Men1 expression throughout the pancreas, we demonstrate abbreviated as MC). Consistent with previous reports, there were that Men1 plays an essential role in the normal homeostatic re- no remarkable changes to the exocrine pancreas (SI Appendix, Fig. sponse to acute pancreatitis. We further demonstrate the Men1 S1), despite robust reduction in Men1 protein levels (Fig. 1A). We protein is stabilized in response to stress and the altered response next challenged the pancreas with the established model of is not associated with the known MLL-dependent functions of caerulein-induced acute inflammation (25, 26). Briefly, caerulein Men1, but rather to its role as a negative regulator of the Jund is a cholecystokinin analog that stimulates the local release of transcription factor. Finally, we demonstrate that in addition to digestive enzymes and results in edema, infiltration of immune

A WT MC B C 0.8 E 40 90 kDa caerulein p=0.0005 Men1 8x 8x 0.6 *** 30

160 kDa GENETICS 0.4 20 Vcl 1 2 7 days D 0.2 10 Pancreas weight % metaplastic area % metaplastic

WT MC (% total mouse weight) 0.0 0 WT MC WT MC

FGp=0.0062 p=0.0033 50 ** 100 ** 40 80 H&E

30 60

20 40 CD45+ cells CD45+ 10 20 CK19+ metaplasias CK19+ (Average # per 20x field) (Average (Average # per 20x field) (Average 0 0 WT MC WT MC

H 2.0 2.0 IHC: CK19

1.5 1.5

1.0 1.0 p=0.0355 Expression p=0.0064 * 0.5 ** 0.5 Ptf1a Expression Ptf1a Cpa1

IHC: CD45 0.0 0.0 WT MC WT MC

Fig. 1. Men1 is essential for the normal pancreas response to acute pancreatitis. (A) Western blot analysis comparing wild-type (WT) to Men1-deficient (MC) pancreas. Men1 expression is compared to vinculin (Vcl) as a loading control. (B) Schematic representation of experimental induction of acute pancreatitis. (C) Pancreas weight 5 d following acute pancreatitis. (D) Pancreas histology 5 d following acute inflammation. Immunohistochemical staining for cytokertatin 19 (CK19) and CD45. Images are at 20×. (Scale bar, 100 μm.) (E) Quantification of percent metaplastic area as a percent of total pancreas area. (F and G) Quantification of immunohistochemical analysis for CK19 and CD45. (H) qRT-PCR analysis of acinar , Cpa1 and Ptf1a. Data are all presented as mean ± SD from individual mice, with female mice indicated in red and males in blue, with a Student’s t test for all statistical analysis.

Wasylishen et al. PNAS | March 24, 2020 | vol. 117 | no. 12 | 6623 Downloaded by guest on September 28, 2021 cells and an almost complete loss of identifiable acinar cells. The A C 2.5 organ recovers quickly and acinar cell morphology and mass are Caerulein 2.0 completely restored within 1 wk following insult, demonstrating PBS 24h 72h the robustness of this regenerative system. 1.5 We treated mice with eight hourly injections of caerulein, administered on 2 consecutive days, based on established pro- Men1 1.0 tocols (27, 28) (Fig. 1B). Five days following the final treatment, 0.5 Men1 expression mice were euthanized to evaluate pancreas histology and re- Vcl 0.0 covery from acute caerulien-induced pancreatitis (29). The PBS 24h 72h pancreas weight of Men1-deficient mice was significantly re- 1.0 1.6 1.1 Caerulein duced compared to wild-type (WT) controls (Fig. 1C), sug- B WT 24h gesting increased cell death in response to caerulein-induced MC (neg) PBS Caerulein damage. Histologically, the Men1-deficient pancreas demon- strated an impaired regenerative response to caerulein-induced injury, with limited normal acinar architecture at this time point, replaced by cytokeratin 19-positive ductal structures (metaplastic lesions, Fig. 1 D–F), and accompanied by signifi- cantly increased inflammatory infiltrate, highlighted by CD45 IHC: Men1 immunohistochemistry (Fig. 1 D and G). The percent of total Fig. 2. Men1 protein is stabilized in response to acute pancreatitis. (A) pancreatic area containing metaplastic lesions was robustly in- Western blot analysis of Men1 protein expression comparing PBS-treated creased in Men1-deficient pancreas, with the WT controls control to caerulein-treated pancreas, 24 and 72 h following the final exhibiting only few areas of metaplasia (Fig. 1E). Moreover, treatment. Protein expression is compared to vinculin (Vcl). Quantification of there was also a significant decrease in the mRNA expression of average Men1/Vlc signal is indicated below bands. (B) Immunohistochemical the acinar markers Cpa1 and Ptf1a (Fig. 1H). We also evaluated analysis for Men1 protein expression. Untreated MC pancreas is used as a pancreas histology 4 wk following the final treatment, and ob- negative control (Left). Staining was conducted on tissue samples 24 h fol- serve persistent damage in the MC pancreas, with a significant lowing the final treatment with PBS control or caerulein. Images are at 100× μ proportion of persistent metaplastic area (SI Appendix, Fig. S2). magnification. (Scale bar, 10 m.) (C) qRT-PCR analysis of relative Men1 mRNA 24 and 72 h following caerulein treatment. Data represent mean ± SD Combined, these data demonstrate that Men1 is essential for the from individual mice. normal pancreas response to caerulein-induced acute pancrea- titis. Notably, consistent results were obtained from both female and male mice (Fig. 1). suggesting an increase in Men1 protein synthesis or protein Given the robust effects of Men1 loss on pancreas recovery stability rather than gene transcription. following acute pancreatitis 5 d following treatment, we next investigated the timing of the tissue response. Wild-type and MC Men1/MLL Target Genes Do Not Mediate the Pancreas Response to mice were treated with the same acute dosing of caerulein over Acute Inflammation. The major function that has been ascribed to 2 d, and subsequently euthanized at 24- and 72-h time points Men1 as an endocrine tumor suppressor is as an epigenetic after the final injection. Representative histological images are regulator of histone methylation through the MLL complex. presented in SI Appendix, Fig. S1A. In the wild-type pancreas, Specifically MEN1 is important in the transcriptional activation the 24-h time point revealed the expected histological changes, of the cell cycle inhibitors CDKN1B (p27KIP1) and CDKN2C with immune cell infiltration and ductal transdifferentiation of (p18INK4c). To investigate this function in the response to acute the acinar compartment. By 72 h, the pancreas regained normal inflammation, we treated WT and MC mice with caerulein or histology. More pronounced acinar to ductal metaplasia was PBS control and harvested pancreas 24 h following the final evident in the MC pancreas at 24 h, and tissue injury persisted at treatment (Fig. 3A). To assay the molecular response at this time the 72-h time point. Serum amylase and lipase levels can be used point, we first evaluated the mRNA expression of the acinar gene as confirmatory markers of pancreatitis, so we also evaluated Cpa1, and ductal gene Sox9. Caerulein treatment led to a signifi- levels in mice euthanized 17 to 18 h following the final caerulein cant down-regulation of Cpa1 in both genotypes compared to treatment. There is a significant increase in serum amylase levels PBS-treated WT controls (****P = 0.0001, ANOVA with Dun- in caerulein-treated MC mice compared to the phosphate buff- nett’s multiple comparisons test), consistent with a loss of acinar ered saline (PBS)-treated controls (SI Appendix,Fig.S1B)vali- cell state (Fig. 3B). At this time point we also observed a sig- dating the results of our histological and molecular analyses. In nificant increase in Sox9 expression, but only in the MC pan- wild-type mice, at this time point, there is no significant difference creas, indicative of ductal metaplasia (Fig. 3B). We next in serum amylase between the two treatment groups (SI Appendix, evaluated the mRNA expression of Men1/MLL target genes Fig. S1B). Lipase levels were also increased upon caerulein Cdkn1b or Cdkn2c. If epigenetic transcriptional activator func- treatment but the data were not statistically significant (SI Ap- tion is important in this context, we anticipated that the ex- pendix,Fig.S1C). pression of these targets would be lost in the Men1-deficinent Men1 Protein Is Stabilized following Caerulein Treatment. We next pancreas following caerulein treatment. Neither of these genes evaluated the effect of caerulein treatment and pancreatitis on is down-regulated in this context (Fig. 3C). Cdkn2c actually the expression of Men1 in wild-type mice. Western blot analysis showsasignificantincreaseinexpressioncomparedtoPBS- revealed an increase in Men1 protein 24 h following caerulein treated controls. Notably, in PBS-treated mice both Cdkn1b treatment compared to controls (Fig. 2A). Men1 protein sub- and Cdkn2c showed reduced, but not statistically significant, sequently returned to near baseline at 72 h, consistent with tissue expression in MC mice compared to wild-type controls. Expression recovery at this time point. Immunohistochemical analysis also of Hoxc6 and Hoxc8, additional Men1/MLL target genes, was revealed an increase in nuclear and cytoplasmic Men1 staining assayed but not detected. These data indicate that the established 24 h following caerulein treatment (Fig. 2B). Men1-deficient target genes of the Men1/MLL histone methyltransferase complex pancreas was used to validate the specificity of the staining. are not important in mediating the pancreas response to acute We found no significant difference in Men1 mRNA (Fig. 2C), inflammation.

6624 | www.pnas.org/cgi/doi/10.1073/pnas.1920017117 Wasylishen et al. Downloaded by guest on September 28, 2021 p=0.0351 1.5 6 A B * h42,SBP h42,nielureaC

WT MC WT MC 1.0 4 xpression expression 0.5 p=0.0001 2

H&E **** Cpa1 e Cpa1 **** Sox9

0.0 0 WT MC WT MC WT MC WT MC PBS Caerulein PBS Caerulein C D E WT MC Upstream Regulator 4 4 p=0.0108 SMO * TWIST1 3 3 JUN SMARCA4 2 2 CG expression expression F2 IFN alpha/beta 1 1 2 EP300 Cdkn2c Cdkn1b IRF3 0 0 0 GLI1 Z-score WT MC WT MC WT MC WT MC 402 DEGs, p <0.05 adj −2 0246 PBS Caerulein PBS Caerulein z-score -log10p-value F G 800 p=0.0082 20 p=0.0462 p=0.0005 150 150 ** p=0.0001

* *** GENETICS 15 600 **** 100 100 10 400 expression expression 50 50 5 200 Ccl7 expression Mmp7 C3ar1 Cxcl16 expression

0 0 0 0 WT MC WT MC WT MC WT MC WT MC WT MC WT MC WT MC PBS Caerulein PBS Caerulein PBS Caerulein PBS Caerulein H I 4% Input IP: Jund endocrine pancreas WT MC WT MC MLL (H3K4me3) + + - - + + - - Caerulein, 24h Men1 Cdkn1b 75 kDa Men1 Cdkn2c 63 kDa exocrine pancreas 48 kDa Jund Jund 35 kDa Men1 135 kDa C3ar1 Vcl 100 kDa Mmp7...

Fig. 3. Molecular response to acute pancreatitis 24 h following treatment. (A) Representative images of pancreas histology 24 h following the final treatment. Images are at 20×. (Scale bar, 100 μm.) (B) qRT-PCR analysis of relative expression of Cpa1 (acinar) and Sox9 (ductal) gene. (C) qRT-PCR analysis of relative expression of Men1/MLL target genes, Cdkn1b and Cdkn2c.(D) Heatmap depicting differentially expressed genes in MC pancreas compared to WT control following chronic pancreatitis. (E) Ingenuity Upstream Regulator Analysis of RNA-sequencing data from chronic caerulein-inducted pancreatitis. The top 10 most significant predicted activators are presented. Z-score indicated with yellow triangle. (F) qRT-PCR analysis of genes identified as JUN targets in the analysis in D 24 h following acute caerulein-induced pancreatitis. (G) qRT-PCR validation of independent Jund target genes, Ccl7 and Cxcl16, 24 h following acute caerulein-induced pancreatitis. (H) Coimmunoprecipitation of Men1 with Jund. (I) Model of important Men1 functions in the endocrine and exocrine pancreas, respectively. All data are presented as mean ± SD from individual mice, relative to PBS-treated WT controls. Statistical analysis of gene expression was conducted with ANOVA with Dunnett’s multiple comparisons test to compare experimental samples to PBS-treated WT controls.

RNA-Sequence Analysis Identifies Jun as an Upstream Regulator of induce inflammation, but mice were treated with four injections Differentially Expressed Genes in a Model of Chronic Inflammation. per day, 3 days a week for 3 wk to model chronic injury (30). We took advantage of a different study in the laboratory to ex- Mice were euthanized 5 d following the final treatment and the plore the transcriptional changes associated with Men1 loss in pancreas of chronically treated MC mice showed a similar phe- the context of chronic pancreatitis. Caerulein was again used to notype to the acute treatment, with an exacerbated injury and

Wasylishen et al. PNAS | March 24, 2020 | vol. 117 | no. 12 | 6625 Downloaded by guest on September 28, 2021 failure to recover compared to wild type (SI Appendix, Fig. S3A). that impair the homeostatic response to pancreatitis also co- RNA-sequence (RNA-seq) and differential gene expression operate with mutant Kras, the most frequently mutated gene in – analysis identified 402 differentially expressed genes (padj < 0.05) human PDAC, to accelerate disease progression (14, 37 39), in the Men1-deficient pancreas compared to wild-type controls: suggest that loss of Men1 might also cooperate and promote 252 (62.7%) genes were up-regulated and 150 (37.3%) genes neoplastic progression in the exocrine pancreas. fl were down-regulated (Fig. 3D and Dataset S1). We next used To directly test this hypothesis, we crossed the Men1 allele to LSL-G12D/+ Tg Ingenuity Pathway Analysis (31) to conduct an upstream regulator Kras Pdx1-Cre mice (KC), and established a small cohort LSL-G12D/+ fl/fl analysis of the differentially expressed genes (Fig. 3E). Notable in of mice with homozygous Men1 loss (Kras Men1 Pdx1- Tg fl the predicted activators was JUN, with one of the most significant Cre , KMC) and control mice heterozygous for the Men1 allele LSL-G12D/+ fl/+ Tg + P values (2.95E-05) and among the highest activation Z-scores (Kras Men1 Pdx1-Cre , KM C). Mice were born at the (2.764). Moreover, previous data have identified MEN1 as a expected Mendelian ratio; however, the majority of mice with ho- specific inhibitor of JUND transcriptional activity, in addition to mozygous Men1 deletion became moribund within 2 mo of age and = its role as part of the MLL histone methyltransferase complex exhibited significantly reduced survival (P 0.0001), with a median (20). Our analysis identified 10 genes with a measurement di- survival of only 45 d compared to 174 d for heterozygous controls rection consistent with activation of JUN (SI Appendix,Fig.S3B). (Fig. 4B). Moribund animals had hunched posture and were sig- Of interest, several of these are inflammation-associated genes. nificantly smaller than littermate controls of the same age (Fig. 4C). Palpation of the sacroiliac bones suggested the mice were under Overexpression of Jun transcription factors also mediates a + proinflammatory response and impaired recovery following pan- conditioned or emaciated (40). The KM C control mice reached creatitis in the context of Nr5a2 heterozygosity (32). humane endpoints for a number of reasons which were not directly We next validated the expression of two of these genes in our related to pancreas pathology, including papillomas and rectal prolapse (SI Appendix,TableS1). On necropsy, the KMC pancreas acute pancreatitis assay at the 24-h time point. Both C3ar1 and + Mmp7 were significantly induced only in MC pancreas compared was firm and notably distinct from KM C mice euthanized at the to PBS-treated WT controls (Fig. 3F). We extended this analysis same time point. We next compared pancreas histology. In all five to two additional direct Jund targets that had previously been KMC mice evaluated, the entire exocrine pancreas was replaced validated by chromatin immunoprecipitation (ChIP) and asso- with ductal proliferation, with little or no acini remaining (SI Ap- ciated with an altered inflammatory response in the pancreas pendix,Fig.S5A and B), suggesting that morbidity is likely a result (32). Both Ccl7 and Cxcl16 also demonstrate significant in- of the general loss of exocrine pancreas function. The majority of duction only in the MC pancreas compared to control (Fig. 3G). the pancreatic lesions were PanIN-1s and PanIN-2s, with dilated Notably, expression of these genes is not induced by Men1 loss ducts partially or completely lined by mucinous epithelium and alone in PBS-treated controls, suggesting that caerulein treat- containing goblet cells, highlighted by Alcian blue staining for mu- ment is required for their induction. cins (Fig. 4D). The cells showed enlarged hyperchromatic, pencilate Previous data suggest Men1 directly binds and inhibits the nuclei with focal pseudostratification. In a few dilated ducts, there transcriptional activity of Jund (20). As expected, we did not see was evidence of further disease progression to PanIN-3, lined by robust changes in total Jund protein expression 24 h following large pleomorphic cells with vesicular nuclei, prominent nucleoli, caerulein treatment in MC pancreas compared to wild-type con- and exhibiting loss of polarity. PanIN lesions were associated with abundant collagen deposition assayed through picrosirius red trols (SI Appendix,Fig.S4). We did, however, note a marked + difference in the ratio of the two Jund bands in caerulean-treated staining (Fig. 4D). The pancreas from age-matched KM Cmice pancreas compared to PBS-treated controls (SI Appendix,Fig.S4, was generally histologically normal in all four mice evaluated, with marked by arrows). Control pancreas show a roughly equal dis- the exception of a single mouse that exhibited chronic pancreatitis tribution of the two Jund bands, whereas, the slower migrating in one region of the pancreas (Fig. 4D and SI Appendix,Fig.S5C). band is the predominant species in caerulein-treated mice re- We quantified the extent of disease in all mice by determining the percent of pancreas lobules that where normal, or contained gardless of genotype. These data suggest that Jund is post- + translationally regulated, and likely activated, following caerulein metaplasias, or PanINs. The pancreas from KM C mice contained treatment in vivo. We next evaluated the association between an average of 90.2% normal lobules, focal areas of metaplasia in Men1 and Jund in wild-type mice 24 h following caerulein treat- 6.5%oflobules,andisolatedPanINsin3.2%(Fig.4E). Pancreas ment. Men1 coimmunoprecipitates with Jund (Fig. 3H), demon- sections from KMC mice, however, had no normal lobules: 18.8% strating that these two proteins interact in the context of contained only metaplastic lesions and 81.2% contained PanINs pancreatitis. Combined, these data indicate that Men1 loss pro- (Fig. 4E). We further evaluated the molecular response to Men1 loss in motes the expression of proinflammatory Jund target genes fol- + lowing caerulein-induced pancreatitis (Fig. 3I). the pancreas of KMC mice to age-matched KM C controls. As anticipated from tissue histology, there was a significant decrease Men1 Loss Accelerates Mutant Kras Driven Oncogenesis. We next in the expression of the acinar cell gene Cpa1, and a significant evaluated the differentially expressed genes from our RNA-seq increase in the expression of the ductal gene Sox9 (Fig. 4F). This data using Enrichr, an integrative web-based software appli- was accompanied by robust or significant increases in the ex- cation to rank enrichment terms (33, 34). From Enrichr, the pression of C3ar1, Mmp7, Ccl7, and Cxcl16, the same proin- disease perturbations from Gene Expression Omnibus (GEO) flammatory Jund target genes we evaluated in response to acute analysis revealed significant enrichment of genes up-regulated in caerulein-induced pancreatitis (Fig. 4G). Combined these data two PDAC datasets: GSE61412, which compared gene expression demonstrate that Men1 loss, and the subsequent deregulation of + in invasive mouse PDACs from Ink4a/Arffl/flKrasLSL-G12D/ Pdx1- Jund, can cooperate with oncogenic Kras to accelerate tumorigenesis. Tg Cre mice compared to Cre-negative control pancreas (35), and Discussion GSE15471 which evaluated gene expression between matched tumor and normal samples from human PDAC patients (36). Through the use of genetic mouse models, we have identified a Thus, the gene expression changes caused by Men1 deficiency in role for Men1 in the regulation of the inflammatory response of our model of chronic pancreatitis show significant overlap with the the pancreas in vivo. Men1 deficiency in the pancreas leads to gene expression changes in pancreatic ductal adenocarcinoma robust defects in the homeostatic response following caerulein- compared to normal tissue, in both mouse and human datasets induced pancreatitis and cooperates with mutant Kras to accel- (Fig. 4A). These associations, combined with data from other erate oncogenesis. Understanding the molecular mechanisms mouse models that clearly demonstrate that genetic perturbations that regulate and maintain pancreas homeostasis is an urgent

6626 | www.pnas.org/cgi/doi/10.1073/pnas.1920017117 Wasylishen et al. Downloaded by guest on September 28, 2021 area of research due to the strong associations between pan- Notably, none of the samples were classified as having high creatitis and cancer. While recurrent MEN1 mutations have not MEN1 staining. Future work to understand the mechanisms that been identified in human PDAC, there are many other mecha- regulate MEN1 protein levels and function in the pancreas and nisms that can alter a pathway or gene. One study examined in PDAC will be instructive. Taken together, our results in MEN1 protein expression by immunofluorescence in normal combination with published and publicly available data strongly exocrine tissue compared to PDAC and identified reduced suggest that MEN1 may have tumor suppressor activity in the MEN1 expression in tumor tissue compared to controls (22). exocrine pancreas. Moreover, using methylation-specific PCR they identified We also explored the mechanisms through which Men1 main- methylation of multiple CpGs in the MEN1 gene promoter in tains pancreas homeostasis in response to caerulean-induced some cases, suggesting that altered epigenetic regulation may stress. Studies in mouse models have suggested that the primary contribute to the reduced MEN1 protein expression in PDAC. tumor suppressor function for Men1 in the endocrine pancreas is Data from the Human Protein Atlas (www.proteinatlas.org) also through epigenetic regulation of gene expression as part of the support these observations (41). Twelve PDAC samples were MLL histone methyltransferase complex. Specifically, combined stained with an antibody against MEN1, and in 8/12 (66.7%) loss of the cell cycle inhibitors Cdkn1b and Cdkn2c promotes samples, MEN1 protein was not detected, while staining was endocrine tumor development in the endocrine pancreas (16). classified as low in 3/12 (25%) and medium in 1/12 (8.3%). Men1 loss, however, did not lead to reduced expression of these

A B C Disease Perturbations (GEO, up) + 30 100 KM C (n=11) p=0.0018 pancreatic ductal adenocarcinoma (mouse, GSE61412) KMC (n=7) ** pancreatic ductal adenocarcinoma (human, GSE15471) ulcerative colitis (human, GSE11223) 20 neurofibromatosis (mouse, GSE29343) Spinal Muscular Atrophy (mouse, GSE10599) 50 Barrett's esophagus(human, GSE34619) Barrett's esophagus (human, GSE34619) Mass (g) 10

schizophrenia (human, GSE25673) Percent survival morbid obesity (human, GSE48964) ***p=0.0001 muscular dystrophy (human, GSE42806)

0 GENETICS 0 02460 100 200 300 + KM CKMC -log10 Adjusted p-value Time (d) D H&E Alcian blue Picrosirius red E PanIN

C 100 Metaplasia + Normal KM

50 Percent of lobules

0 KM+CKMC KMC

G F p=0.0192 p=0.0577 p=0.1954p=0.0072 p=0.0236 50 60 6000 ** * * 1.0 10 40 3000 10 40 30 30 2000 expression 20 expression expression expression expression 0.5 5 expression 5 20

10 Ccl7 Cpa1 p<0.0001 Sox9 Mmp7 10 C3ar1 Cxcl16 **** 0.0 0 0 0 0 0 KM+C KMC KM+C KMC KM+C KMC KM+CKMC KM+CKMC KM+CKMC

Fig. 4. Men1 loss accelerates mutant Kras-driven oncogenesis. (A) Enricher analysis of disease perturbations (GEO, up). (B) Kaplan–Meier survival analysis. ***P = 0.0001, Log-rank (Mantel–Cox) test. (C) Weight of moribund KMC mice compared to age-matched controls. Male mice are indicated in blue and female in red. (D) Representative images of 6- to 7-wk-old pancreas samples stained with H&E, alcian blue, and picrosirius red. Images taken at 10× magnification. (Scale bar, 100 μm.) (E) Quantification of lesions present in pancreas sections represented as a percentage of total lobules, with mean ± SD from four in- dependent mice presented. (F) qRT-PCR analysis of Cpa1 (acinar) and Sox9 (ductal) gene expression. (G) qRT-PCR analysis of Jund target genes. Weight and expression data are all presented as mean ± SD from individual mice with a Student’s t test for statistical analysis.

Wasylishen et al. PNAS | March 24, 2020 | vol. 117 | no. 12 | 6627 Downloaded by guest on September 28, 2021 genes in response to caerulein treatment. Transcriptome analysis in and obtained from The Jackson Laboratory (10, 44). Mice were maintained a chronic model of caerulein-induced pancreatitis identified JUN on a mixed background (C57BL/6, FVB, 129), Cre-negative littermates were as a candidate upstream regulator of the differentially expressed used as functionally wild-type controls, and both male and female mice were genes in the Men1-deficient pancreas. Previous studies have used in our analyses. Genotyping primer sequences are provided in SI Ap- demonstrated that Men1 is also a direct negative regulator of Jund pendix, Table S2. transcriptional activity (20). Indeed, Jund target genes are up- regulated in our assays, suggesting that increased Jun activity is Caerulein-Induced Acute Pancreatitis. Acute pancreatitis was induced based on established protocols. Caerulein (MedChemExpress) was reconstituted in contributing to the altered pancreas response. A recent study di- sterile PBS. Adult mice (6 to 10 wk old) were injected (individual dose: 75 μg/kg), rectly tested the functional impact of Jun family proteins in a given as eight hourly i.p. injections on 2 consecutive days during the light cycle. similar model of impaired pancreas regeneration. Mice hetero- Mice were s.c. administered sustained release buprenorphine as an analgesic zygous for the orphan nuclear receptor Nr5a2 basally express el- prior to the first caerulein injection. evated levels of proinflammatory genes. This thereby sensitizes the pancreas to damage, impairs regeneration, and promotes PDAC Histology and Immunohistochemistry. Tissues harvested from mice were fixed development in cooperation with mutant Kras (32, 39). Mecha- in 10% neutral buffered formalin and paraffin embedded. Four-micrometer nistically, this was linked to up-regulation of AP1 family members sections were stained with hematoxylin and eosin (H&E) and examined by + − Jun, Junb,andJund in Nr5a2 / pancreas, which increased promoter light microscopy. Tissue processing, paraffin embedding, sectioning, and binding of Jun family proteins and induced the expression of in- H&E staining were performed by the MD Anderson Department of Veteri- flammatory genes. Genetically, pancreas deletion of Jun rescued the nary Medicine and Surgery Histology Laboratory. Immunohistochemistry + − phenotype of Nr5a2 / mice and restored the normal tissue re- was performed using standard methods with citrate buffer for antigen re- sponse to and recovery from caerulein-induced pancreatitis. While trieval and stained with antibodies against cytokeratin 19 (ab133496, this study genetically manipulated Jun, AP1 family members share Abcam), CD45 (550539, BD Pharmingen), and Men1 (ab3605, Abcam). Visu- many common targets and these data support the proposed alization was performed using ABC and DAB kits (Vector Laboratories) and mechanisms of unrestricted Jund activity contributing to the phe- slides were counterstained with hematoxylin. notypes in our Men1-deficient model. Image Quantification. Acinar to ductal metaplasias and PanIN lesions were Our data also have potential clinical implications. We dem- quantified from whole pancreas sections scanned on an Aperio Scan Scope XT onstrate that the homozygous loss of Men1 increases the severity (Leica Biosystems) and manually annotated using Aperio ImageScope soft- of and impairs the recovery from an experimental model of ware (Leica Biosystems). For acute inflammation experiments, the percent of pancreatitis. While rare, MEN1 syndrome patients inherit one total pancreas area occupied by metaplasia was calculated for each sample. mutant MEN1 allele and have an over 80% lifetime risk of de- Cytokeratin 19-positive metaplasias and CD45-positive cells were manually veloping pancreatic neuroendocrine tumors. As such, these pa- quantified from four random and nonoverlapping 20× fields and averaged tients undergo active surveillance for tumors. Once identified, it for each mouse. For quantification of metaplasias and PanIN lesions in the is challenging to predict which pancreas lesions are likely to Kras cohort, individual pancreas lobules were counted and evaluated as progress and metastasize from imaging alone. Tissue sampling normal, containing metaplasia, or containing PanIN. The number of lobules + through fine needle aspirate or core biopsy provides the oppor- in KM C pancreas was estimated based on the number of individual ductules tunity to obtain cells and tissue to evaluate features that might present. Lobules were demarcated by surrounding collagen in KMC mice. correlate with diseases status or progression, such as proliferation (Ki67). Approximately 1 to 3% of all patients receiving pancreas Western Blotting. Tissue samples were flash frozen, pulverized, and lysed in biopsies by endoscopic ultrasound and fine needle aspiration de- sodium dodecyl sulfate (SDS) lysis buffer (1% SDS, 6.5 mM Tris·HCl pH = 6.8, β velop pancreatitis, and our data suggest that this number may be 25% glycerol, 10% -mercaptoethanol). Protein extracts were separated by increased in the MEN1 patient population. As recommendations SDS/polyacrylamide gel electrophoresis (PAGE), transferred to nitrocellulose for screening continue to be modified, the potential risk of pan- membranes, and probed with antibodies against Men1 (ab3605, Abcam), Jund (ab181615, Abcam), and Vinculin (V1931, Sigma). Proteins were visu- creatitis as identified through this work should be considered. alized using Li-COR secondary antibodies and scanned on a Li-COR Odyssey Secondly, while MEN1 is an established tumor suppressor in en- Imager. Signal was quantified using ImageJ software (NIH). docrine tumors, recent work in MLL translocated leukemia sug- gests that MEN1 may also have tumor-promoting functions (19) Coimmunoprecipitation. Flash frozen and pulverized pancreas was homoge- and inhibitors of MEN1 have demonstrated therapeutic efficacy in nized and then agitated for 2 h at 4 °C in lysis buffer (50 mM Tris pH 7.4, preclinical models of this disease (42). MLL and JUND share the 150 mM NaCl, 2 mM ethylenediaminetetraacetic acid (EDTA), 1% Nonidet P- same MEN1 binding pocket (43); therefore, as potential small 40 with protease inhibitor and phosphatase inhibitor). The samples were molecules of the MLL:MEN1 interaction advance for clinical use, then centrifuged at 12,000 rpm for 20 min. The supernatant was taken for our data suggest that it may be important to consider increased coimmunoprecipitation. One milligram of total protein lysate was incubated susceptibility to pancreatitis in the clinical evaluation. with 3 μg Jund antibody (ab181615, Abcam) overnight at 4 °C. The lysate was Combined, our data identify Men1 as an essential mediator of further incubated with 15 μL wet protein A-agarose beads (SC-2001, Santa the normal pancreas response to pancreatitis, and encourage Cruz Biotechnology) for 4 h at 4 °C. The beads were collected and washed future studies to understand the potential of Men1 to function as three times with lysis buffer and protein was eluted with 1× SDS buffer. a tumor suppressor in pancreatic ductal adenocarcinomas. JunD and Men1 were detected by Western blot. The antibody for JunD (ab181615, Abcam) is diluted 1 in 1,000. The antibody for Men1 (A300- Materials and Methods 105A, Bethyl) is diluted 1 in 10,000. Light chain-specific secondary anti- body (211-032-171, Jackson ImmunoReseach) was used and proteins were Mice. All mouse experiments were performed in compliance with the National Institutes of Health guidelines for animal research, and approved by the MD detected by chemiluminescence. Loading of inputs was assayed by immuno- Anderson Cancer Center Institutional Animal Care and Use Committee. Mice blotting for Vcl (V1931, Sigma), using a Li-COR secondary antibody and scan- were housed in individually ventilated cages, on individually ventilated cage ned on a Li-COR Odyssey Imager. racks, with up to five animals per cage after weaning. Direct cage bedding consisted of corn cob, Enviro-dri, Enrich-n-nest, and nestlets. The facility uses a RNA Isolation and Quantitative PCR. Flash frozen tissue was pulverized and 14-h light (7:00 AM to 9:00 PM) and 10-h dark (9:00 PM to 7:00 AM) cycle. Mice homogenized in TRIzol RNA isolation reagent (Invitrogen). RNA was then were fed a standard diet sterilized by irradiation (PicoLab Rodent Diet 5053, purified using an RNeasy Mini Kit (Qiagen). RNA was then used as a template Purina) and were provided reverse osmosis chlorinated or acidified water for first strand synthesis using the iScript Reverse Transcription Supermix (Bio- ad libitum. Rad). Quantitative RT-PCR was carried out using 2× SYBR Green qPCR master Men1 conditional knockout (stock no. 005109), Pdx1-CreTg (stock no. mix on a CFX384 Realtime System (Bio-Rad). Primer sequences are provided 014647), and KRasLSL-G12D (stock no. 008179) mice were previously described in SI Appendix, Table S2.

6628 | www.pnas.org/cgi/doi/10.1073/pnas.1920017117 Wasylishen et al. Downloaded by guest on September 28, 2021 Differential Gene Expression Analysis from RNA-Seq Data. STAR (version 2.6.0b) considered statistically significant. Comparisons between two groups were wasusedtoalignreadstotheGRCm38.p6 reference genome (45). Samtools made with a Student’s t test, and comparisons among multiple groups were (version 1.8) was used to sort, convert between formats, and calculate mapping made with ANOVA with Dunnett’s multiple comparisons test to compare statistics (46). FastQC (0.11.5) was used to check for FASTQ read qualities. Gene experimental samples to control treated wild-type controls. Comparison of annotation was carried out using GENCODE M19 (Ensembl 94) annotation, survival curves was done using a Log-rank (Mantel–Cox) test. which was downloaded from the GENCODE project (47). Aligned reads were summarized at the gene level using HTseq-count (48). Differential gene ex- Data Availability Statement. RNA-seq data were deposited in NCBI’s Gene pression was determined using R (3.5.1) and Bioconductor package DESeq2 (49). Expression Omnibus and are accessible through GEO Series accession no. Read count was first prefiltered to remove extremely low expressed genes. GSE145175. DESeq2 was then used to carry out read count filtering, normalization, disper- sion estimation, and identification of differential expression. DESeq2 modeled ACKNOWLEDGMENTS. We would like to thank Drs. Florencia McAllister and the counts using a negative binomial distribution, followed by the Walk test. The Anirban Maitra and members of the G.L. laboratory for helpful discussions. – final adjusted P value was obtained using the Benjamini Hochberg method. This research was supported by grants from the Neuroendocrine Tumor Research Foundation and NIH (NIH/National Cancer Institute R21 CA208463) Statistics. All data are presented as mean ± SD. All statistical analyses were to G.L. and a fellowship from the Canadian Institutes of Health Research (to performed using GraphPad Prism 6 software, and P value of <0.05 was A.R.W.). A.R.W. is the recipient of a Dodie P. Hawn Award in Genetics.

1. ChE. Forsmark, S. S. Vege, C. M. Wilcox, Acute pancreatitis. N. Engl. J. Med. 376, 598– 25. C. Guerra et al., Chronic pancreatitis is essential for induction of pancreatic ductal 599 (2017). adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell 11, 291–302 (2007). 2. P. J. Lee, G. I. Papachristou, New insights into acute pancreatitis. Nat. Rev. Gastro- 26. J. J. Hyun, H. S. Lee, Experimental models of pancreatitis. Clin. Endosc. 47, 212–216 enterol. Hepatol. 16, 479–496 (2019). (2014). 3. S. Hausmann, B. Kong, C. Michalski, M. Erkan, H. Friess, The role of inflammation in 27. J. L. Kopp et al., Identification of Sox9-dependent acinar-to-ductal reprogramming as . Adv. Exp. Med. Biol. 816, 129–151 (2014). the principal mechanism for initiation of pancreatic ductal adenocarcinoma. Cancer 4. F. Giusti, F. Marini, M. L. Brandi, “Multiple endocrine neoplasia type 1” in Cell 22, 737–750 (2012). GeneReviews, M. P. Adam et al., Eds. (University of Washington, Seattle, WA, 1993). 28. J. P. Morris, 4th, D. A. Cano, S. Sekine, S. C. Wang, M. Hebrok, Beta-catenin blocks 5. R. V. Thakker, Multiple endocrine neoplasia type 1 (MEN1) and type 4 (MEN4). Mol. Kras-dependent reprogramming of acini into pancreatic cancer precursor lesions in Cell. Endocrinol. 386,2–15 (2014). mice. J. Clin. Invest. 120, 508–520 (2010). 6. Y. Jiao et al., DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in 29. National Research Council, Guide for the Care and Use of Laboratory Animals (Na- pancreatic neuroendocrine tumors. Science 331, 1199–1203 (2011). tional Academies Press, Washington, DC, ed. 8, 2011). 7. A. Scarpa et al.; Australian Pancreatic Cancer Genome Initiative, Whole-genome 30. F. McAllister et al., Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 sig- – landscape of pancreatic neuroendocrine tumours. Nature 543,65–71 (2017). naling axis in preinvasive pancreatic neoplasia. Cancer Cell 25, 621 637 (2014). 8. M. K. Cromer et al., Identification of somatic mutations in parathyroid tumors using 31. A. Krämer, J. Green, J. Pollard, Jr., S. Tugendreich, Causal analysis approaches in in-

– GENETICS whole-exome sequencing. J. Clin. Endocrinol. Metab. 97, E1774–E1781 (2012). genuity pathway analysis. Bioinformatics 30, 523 530 (2014). 9. J. S. Crabtree et al., A mouse model of multiple endocrine neoplasia, type 1, develops 32. I. Cobo et al., Transcriptional regulation by NR5A2 links differentiation and in- – multiple endocrine tumors. Proc. Natl. Acad. Sci. U.S.A. 98, 1118–1123 (2001). flammation in the pancreas. Nature 554, 533 537 (2018). 10. J. S. Crabtree et al., Of mice and MEN1: Insulinomas in a conditional mouse knockout. 33. E. Y. Chen et al., Enrichr: Interactive and collaborative HTML5 gene list enrichment Mol. Cell. Biol. 23, 6075–6085 (2003). analysis tool. BMC Bioinformatics 14, 128 (2013). 11. J. Lu et al., Alpha cell-specific Men1 ablation triggers the transdifferentiation of 34. M. V. Kuleshov et al., Enrichr: A comprehensive gene set enrichment analysis web – glucagon-expressing cells and insulinoma development. Gastroenterology 138, 1954– server 2016 update. Nucleic Acids Res. 44, W90 W907 (2016). 35. F. Guillaumond et al., Cholesterol uptake disruption, in association with chemother- 1965 (2010). apy, is a promising combined metabolic therapy for pancreatic adenocarcinoma. Proc. 12. H. C. Shen et al., Multiple endocrine neoplasia type 1 deletion in pancreatic alpha- Natl. Acad. Sci. U.S.A. 112, 2473–2478 (2015). cells leads to development of insulinomas in mice. Endocrinology 151, 4024–4030 36. L. Badea, V. Herlea, S. O. Dima, T. Dumitrascu, I. Popescu, Combined gene expression (2010). analysis of whole-tissue and microdissected pancreatic ductal adenocarcinoma iden- 13. H. C. Shen et al., Recapitulation of pancreatic neuroendocrine tumors in human tifies genes specifically overexpressed in tumor epithelia. Hepatogastroenterology 55, multiple endocrine neoplasia type I syndrome via Pdx1-directed inactivation of Men1. 2016–2027 (2008). Cancer Res. 69, 1858–1866 (2009). 37. J. Mallen-St Clair et al., EZH2 couples pancreatic regeneration to neoplastic pro- 14. C. M. Hughes et al., Menin associates with a trithorax family histone methyl- gression. Genes Dev. 26, 439–444 (2012). transferase complex and with the hoxc8 locus. Mol. Cell 13, 587–597 (2004). 38. N. M. Krah et al., The acinar differentiation determinant PTF1A inhibits initiation of 15. T. A. Milne et al., Menin and MLL cooperatively regulate expression of cyclin- pancreatic ductal adenocarcinoma. eLife 4, e07125 (2015). dependent kinase inhibitors. Proc. Natl. Acad. Sci. U.S.A. 102, 749–754 (2005). 39. M. Flandez et al., Nr5a2 heterozygosity sensitises to, and cooperates with, in- 16. D. S. Franklin, V. L. Godfrey, D. A. O’Brien, C. Deng, Y. Xiong, Functional collaboration flammation in KRas(G12V)-driven pancreatic tumourigenesis. Gut 63, 647–655 (2014). between different cyclin-dependent kinase inhibitors suppresses tumor growth with 40. T. Burkholder, C. Foltz, E. Karlsson, C. G. Linton, J. M. Smith, Health evaluation of – distinct tissue specificity. Mol. Cell. Biol. 20, 6147 6158 (2000). experimental laboratory mice. Curr. Protoc. Mouse Biol. 2, 145–165 (2012). 17. A. Yokoyama et al., Leukemia proto-oncoprotein MLL forms a SET1-like histone 41. M. Uhlen et al., A pathology atlas of the human cancer transcriptome. Science 357, methyltransferase complex with menin to regulate Hox gene expression. Mol. Cell. eaan2507 (2017). – Biol. 24, 5639 5649 (2004). 42. D. Borkin et al., Pharmacologic inhibition of the Menin-MLL interaction blocks pro- 18. T. A. Milne et al., MLL targets SET domain methyltransferase activity to Hox gene gression of MLL leukemia in vivo. Cancer Cell 27, 589–602 (2015). – promoters. Mol. Cell 10, 1107 1117 (2002). 43. J. Huang et al., The same pocket in menin binds both MLL and JUND but has opposite 19. A. Yokoyama et al., The menin tumor suppressor protein is an essential oncogenic effects on transcription. Nature 482, 542–546 (2012). cofactor for MLL-associated leukemogenesis. Cell 123, 207–218 (2005). 44. S. R. Hingorani et al., Preinvasive and invasive ductal pancreatic cancer and its early 20. S. K. Agarwal et al., Menin interacts with the AP1 transcription factor JunD and re- detection in the mouse. Cancer Cell 4, 437–450 (2003). presses JunD-activated transcription. Cell 96, 143–152 (1999). 45. A. Dobin et al., STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 29,15–21 21. A. Karpathakis et al., Pancreatic adenocarcinoma in a patient with multiple endocrine (2013). neoplasia 1 syndrome. Pancreas 42, 725–726 (2013). 46. H. Li et al.; 1000 Genome Project Data Processing Subgroup, The sequence alignment/ 22. I. Cavallari et al., Decreased expression and promoter methylation of the menin tu- map format and SAMtools. Bioinformatics 25, 2078–2079 (2009). mor suppressor in pancreatic ductal adenocarcinoma. Genes Cancer 48, 47. J. Harrow et al., GENCODE: The reference annotation for the ENCODE 383–396 (2009). project. Genome Res. 22, 1760–1774 (2012). 23. J. Kleeff et al., Chronic pancreatitis. Nat. Rev. Dis. Primers 3, 17060 (2017). 48. S. Anders, P. T. Pyl, W. Huber, HTSeq–A Python framework to work with high- 24. C. Carrière, A. L. Young, J. R. Gunn, D. S. Longnecker, M. Korc, Acute pancreatitis throughput sequencing data. Bioinformatics 31, 166–169 (2015). markedly accelerates pancreatic cancer progression in mice expressing oncogenic 49. M. I. Love, W. Huber, S. Anders, Moderated estimation of fold change and dispersion Kras. Biochem. Biophys. Res. Commun. 382, 561–565 (2009). for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

Wasylishen et al. PNAS | March 24, 2020 | vol. 117 | no. 12 | 6629 Downloaded by guest on September 28, 2021