Cancer Cell & Microenvironment 2016; 3: e1472. doi: 10.14800/ccm.1472; © 2016 by C. Benedikt Westphalen, et al. http://www.smartscitech.com/index.php/ccm

REVIEW

Cellular plasticity and heterogeneity in pancreatic regeneration and malignancy

C. Benedikt Westphalen1 Bernhard W. Renz2, Maximilian Reichert3,6, Anil K. Rustgi6, Timothy C. Wang4,5

1Department of Internal Medicine III, Hospital of the University of Munich, Munich, 81377, Germany 2Department of General, Visceral, and Transplantation Surgery, Hospital of the University of Munich, Munich, 81377, Germany 3Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität Munich, Munich, 81675, Germany 4Divison of Digestive and Disease, Columbia University Medical Center, New York, NY 10032, USA 5Herberg Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA 6Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA

Correspondence: Timothy C. Wang or C. Benedikt Westphalen E-mail: [email protected] or [email protected] Received: November 07, 2016 Published online: December 19, 2016

Pancreatic regeneration in response to tissue injury is a complex process. Recent progress in the understanding of this process has underscored the need for cellular plasticity as a prerequisite in the course of regeneration. A number of different pancreatic cell types have been proposed as progenitors, stem cells or differentiated cells with a high degree of plasticity. Moreover, in the setting of an oncogenic mutation, similar mechanisms appear to drive pancreatic tumorigenesis. Here, we aim to summarize the recent advances in our understanding of cellular plasticity in the setting of regeneration and tumorigenesis. Keywords: Dclk1; Prrx1; ; plasticity; pancreatic stem cells; pancreatic regeneration To cite this article: C. Benedikt Westphalen, et al. Cellular plasticity and heterogeneity in pancreatic regeneration and malignancy. Can Cell Microenviron 2016; 3: e1472. doi: 10.14800/ccm.1472. Copyright: © 2016 The Authors. Licensed under a Creative Commons Attribution 4.0 International License which allows users including authors of articles to copy and redistribute the material in any medium or format, in addition to remix, transform, and build upon the material for any purpose, even commercially, as long as the author and original source are properly cited or credited.

Introduction therapies for diabetic patients. Potential strategies for regenerating pancreatic islet cells have been extensively The possesses a remarkable capacity to respond reviewed previously and are beyond the scope of this review to injurious insults and regenerates after cessation of tissue [4]. Only in the last decade, the cellular mechanisms for injury. This ability has generated considerable interest in the regeneration within the pancreatic exocrine compartment underlying mechanisms for this regenerative response. In have gained increasing interest. Despite major scientific general, two possibilities for tissue regeneration in the adult efforts, true stem cells in the adult pancreas have not been exist: (1) the presence of bona fide or facultative stem cells found and their existence appears unlikely. Nevertheless, an or (2) differentiated cells with the ability to de- or understanding of the mechanisms underlying regeneration of transdifferentiate and then proliferate in response to injury the pancreas might provide insights into the process of [1-3]. The scientific interest in adult pancreatic stem cells has malignant transformation of the pancreas. As pancreatic largely been fueled by the interest in developing regenerative ductal adenocarcinoma is a major health burden, a deeper

Page 1 of 8

Cancer Cell & Microenvironment 2016; 3: e1472. doi: 10.14800/ccm.1472; © 2016 by C. Benedikt Westphalen, et al. http://www.smartscitech.com/index.php/ccm understanding of its core biology is imperative for future metaplasia (ADM) was subsequently uncovered [13-17]. In diagnostic and therapeutic measures. Here, we will outline response to injury, acinar cells reactivate an embryonic the recent progress in the understanding of cellular plasticity program that involves the re-expression of PDX1, Sox9 and in the process of exocrine pancreatic regeneration and Hes1, and the emergence of ductal markers such as CK19 tumorigenesis. [18-21]. The process of ADM is reversible, and after cessation of injury, regeneration of acinar cells appears to be driven Cellular Plasticity in the Pancreas from the acinar compartment.

Under resting conditions, there is a steady but slow Since early studies applied rather broad Cre-drivers to turnover of pancreatic endocrine and exocrine cells. After study the process of acinar regeneration, it remained unclear injury, cell division rates increase markedly and the tissue if there are subsets of acinar cells that possess more plasticity mass is restored to a certain extent but not completely, given than others. Bmi1 is a marker of reserve stem cells in the the limited regenerative capacity of the pancreas compared to intestine [22]. Using Bmi1-CreERT mice, Capecchi and say, the liver. The drivers of this cellular response in the colleagues defined a subset of long-lived acinar cells in the pancreas have been a point of considerable debate, with some adult pancreas. They proposed a role for these cells in investigators favoring the existence of dedicated pancreatic pancreatic regeneration, as they found an expansion of this stem cells, while others proposing that differentiated cells lineage after pancreatic injury. However, since labeling of drive pancreatic regeneration through an intermediate stage the cells was induced only after injurious insult to the involving dedifferentiation [1, 3, 5, 6]. pancreas, it remained unclear if the regeneration occurred from existing Bmi1+ cells, or from newly generated Bmi1+ In the search for potential pancreatic stem cells, cells secondary to upregulation of the Bmi1 transcript [23]. investigators have turned to clonogenic or sphere forming assays [7]. These experiments yielded the finding that the Dclk1 has been proposed as a marker of quiescent stem most efficient clonogenic cells seem to reside within a subset cells in the intestine and the pancreas [24-26]. Using Dclk1 of ductal cells and/or in centroacinar cells [8, 9]. However, in BAC-CreERT mice, we were able to show that Dclk1 labels contrast to studies in other organ systems, there are rare and long-lived cells in the adult pancreas [27]. While insufficient experimental data supporting the concept that quiescent under homeostatic conditions, these cells became these clonogenic cells serve as pancreatic stem cells in vivo. activated following diverse forms of pancreatic injury and While ductal cells acquire some features of embryonic contribute to organ regeneration. Loss of Dclk1+ cells progenitor cells in response to pancreatic injury [10], (through diphtheria toxin-mediated cell ablation) prevented lineage-tracing experiments using a variety of ductal pancreatic repair and caused significant mortality after Cre-markers did not show convincing evidence of cerulein-induced pancreatitis. Finally, we were able to differentiation of ductal cells into other pancreatic lineages demonstrate that Dclk1+ cells had a proliferative advantage after pancreatic injury [1]. However, these findings do not rule over the Dclk1- compartment, arguing against the hypothesis out the possible existence of a subset of ductal cells with a that all acinar cells are equal and that a stochastic model higher degree of plasticity. In fact, recent findings from our underlies the regenerative processes in the pancreas [27]. labs have provided evidence for a rare subset of Prrx1+ ductal cells with a remarkable degree of sphere forming Centroacinar cells have been proposed as yet another ability and features of progenitor cells [11]. In fact, Prrx1 source of pancreatic stem cells due to their location at the tip ductal cells expanded markedly after cerulein-induced of the ductal tree, and their transcriptional profile [2, 6, 9, 28]. pancreatitis, consistent with a role in pancreatic regeneration While these cells express embryonic markers and form after injury [11]. Of note, Prrx1+ serve as stem cells in bone pancreatic organoids with higher frequency than other cell and cartilage formation [12]. types, subsequent lineage tracing studies did not reveal a major contribution of centroacinar cells to the regenerating Acinar to Ductal Metaplasia pancreas [29, 30].

In response to pancreatic tissue damage, complex ductal In summary, cellular plasticity may be an important driver structures arise. Because of their resemblance to the ductal of exocrine pancreatic regeneration. In particular, the epithelium, investigators thought initially that these existence of specialized subsets of acinar and ductal cells structures developed from pancreatic ducts. However, with a higher degree of plasticity has emerged in recent seminal lineage tracing studies revealed that differentiated studies. Consequently, it is tempting to speculate that these acinar cells are the predominant source of these ductal cells are potential candidates for facultative progenitor cells structures and the molecular framework of acinar-to-ductal involved in pancreatic repair after injury.

Page 2 of 8

Cancer Cell & Microenvironment 2016; 3: e1472. doi: 10.14800/ccm.1472; © 2016 by C. Benedikt Westphalen, et al. http://www.smartscitech.com/index.php/ccm

Cellular Plasticity in Pancreatic Tumorigenesis potent cancer initiating cells in transplantation experiments [40]. Using our Dclk1 BAC-CreERT transgenic mice, we As outlined above, cellular plasticity within the exocrine investigated the role of Dclk1+ cells in the initiation of compartment plays an important role in pancreatic pancreatic cancer. Targeting mutant Kras expression regeneration. Furthermore, cellular plasticity seems to be an specifically to adult pancreatic Dclk1+ cells had no effect on underlying mechanism for malignant progression [31]. their quiescence and longevity. Similar to the behavior of Pancreatic ductal adenocarcinoma (PDAC) was originally other acinar cells, the combination of an oncogenic mutation thought to arise from cells of the ductal epithelium because in Dckl1+ cells with an injurious insult (cerulein-induced of its histological appearance [32]. However, in genetically pancreatitis) led to the emergence of PanINs from Dclk1+ engineered mouse models of PDAC, a variety of different lineage traced cells. Given their exceptional role in pancreatic cell types possess the ability to give rise to pancreatic regeneration, we investigated if these cells were precursors of pancreatic cancer [33, 34]. Acinar-to-ductal more efficient to give rise to PanINs than the bulk of the metaplasia, similar to that seen in response to pancreatic acinar compartment. The basic helix-loop-helix transcription injury, can be observed when an oncogenic Kras mutation is factor, Mist1, labels the majority of adult acinar cells. targeted to the acinar compartment [35]. In contrast to the Accordingly, cerulein-induced pancreatic leads to transient nature of injury-induced ADM, Kras-induced ADM widespread PanIN development in mice with mutant Kras persists and is also termed acinar-to-ductal reprogramming targeted to the Mist1 compartment. To estimate the degree of (ADR) [19, 36]. Importantly, expression of mutant Kras in transformation of pancreatic Dclk1+ cells we compared the adult acinar cells not only causes ADM/ADR but is sufficient efficiency of Dclk1+ and Mist1+ lineage traced cells to give to induce formation of pancreatic intraepithelial neoplasia rise to PanINs after pancreatitis. We found that Dclk1+ cells (PanIN), a precursor of pancreatic cancer [35]. were up to 12x more efficient in the induction of PanINs when compared to the Mist1+ acinar cells alone. These PanINs resemble ductal structures, and like PDAC were findings are consistent with the plasticity of Dclk1+ cells originally thought to be derived from the ductal epithelium. observed in pancreatic sphere cultures and pancreatic Lineage tracing studies, however established firmly the regeneration [27]. acinar compartment as the predominant source of murine PanINs [35]. Targeting mutant Kras to the adult acinar While there is compelling evidence that acinar cells serve compartment was shown to be sufficient to induce PanIN as the major source of murine PanINs, a number of formation. However, progression to PDAC is infrequently observations have also pointed to ductal cells as an origin for observed in these models, particularly when Kras is targeted PanIN lesions. Targeted expression of mutant Kras to CK19+ to adult pancreatic acinar cells. By contrast, Kras mutant ductal cells was sufficient to induce low frequency PanIN acinar cells readily give rise to PDAC when subjected to development [41]. Recently, Leach and co-workers were able experimental pancreatitis [37, 38]. Given the fact that to show that pancreatic cancer can also be derived from the pancreatitis reactivates an embryonic program in acinar cells ductal epithelium using Hnf1bCreERT mice, where the and increases their plasticity, it is not surprising that the specific transcription factor Hnf1b drives CreERT combination of an oncogenic mutation and an inflammatory expression. In their experiments, mutant Kras targeted to the insult converts pancreatic acinar cells into potent cancer ductal epithelium was not sufficient to initiate pancreatic initiating cells. In fact, Sander and co-workers were able to tumorigenesis. However, combined expression of mutant show that pancreatic acinar cells are >100 times more Kras and two mutant p53 alleles (R172H) led to rapid efficient in their ability to give rise to murine PanINs when development of PDAC from Hnf1b+ lineage traced cells. compared to duct cells [39]. Importantly, no other stimulus such as pancreatic injury was needed to induce malignant transformation. Of note, PanIN While these studies clearly pointed towards an important lesions were only infrequently observed when PDAC was role of acinar cells as an origin of pancreatic neoplasia, they driven from Krasmt/wt/p53mt/mt ductal cells [42]. Thus, in failed to answer the question if indeed all acinar cells were contrast to the acinar model where pancreatitis provides a alike or if some acinar cells existed that were more prone to second hit to induce pancreatic cancer, in the ductal model, give rise to PanINs. Bailey and co-workers were the first to the mutation (and loss of heterozygosity) of a tumor investigate the role of Dclk1+ cells in murine PanINs and suppressor gene (p53) suffices as the “second hit” to drive pancreatic cancer cell lines. They found that Dclk1 labeled a progression to mPDAC [43]. Taken together, these findings morphologically and functionally distinct subset of cells show that both ductal and acinar cells harbor the ability to within PanINs and pancreatic cancer cell lines. Functionally, initiate pancreatic cancer in mice. these cells displayed a high degree of sphere forming capacity and stemness. Furthermore, they were found to be Interestingly, pancreatic organoids, a long-term

Page 3 of 8

Cancer Cell & Microenvironment 2016; 3: e1472. doi: 10.14800/ccm.1472; © 2016 by C. Benedikt Westphalen, et al. http://www.smartscitech.com/index.php/ccm three-dimensional culture system of pancreatic epithelial relevance of Dclk1 expression was provided by in silico and cells, appear to be derived from pancreatic ductal epithelium in vitro studies from our group, which suggest a direct and [44]. While primary culture of acinar cells is possible as well functionally important interaction between Dclk1 and Kras and has been used to study acinar to ductal metaplasia [13, 45], [27, 48-50]. long-term primary cultures of acinar cells have not been established to date. Even if one takes into account, that acinar The Notch pathway is critically involved in cell fate cells represent the vast majority of pancreatic epithelial cells, decisions and regulates pancreatic development [51, 52]. In the experiments with forced expression of mutant Kras suggest adult, expression of Notch pathway components is restricted that a subset of acinar cells have a much higher degree of to centroacinar and terminal duct cells [29]. In response to plasticity. This higher degree of plasticity may translate into pancreatic injury, Notch signaling is reactivated and critical a higher degree of susceptibility to the deleterious effects of for pancreatic regeneration [53]. During tissue repair Notch mutant Kras and/or injury. While more resistant to Kras and Wnt/beta-catenin signaling cooperate to allow for acinar mutation and injury, ductal cells still retain the ability to give cell regeneration [53]. Furthermore, ongoing beta-catenin rise to pancreatic cancer under the appropriate conditions [42]. signaling is sufficient to block Kras-mediated acinar Our experimental data also suggest that not all acinar and transformation underscoring [19] the importance of tightly ductal cells are alike. Morphology alone is insufficient to regulated cellular identity in response to injurious stimuli. discriminate between subsets within a cellular compartment and evidence points to considerable heterogeneity within Additionally, cellular plasticity is regulated on an pancreatic lineages that remains to be explored. Considering epigenetic level. In zebrafish, loss of the pivotal DNA the remarkable heterogeneity of pancreatic ductal methyltransferase 1 (Dnmt1) leads to loss of the pancreas 84 adenocarcinoma - in terms of genetic alterations, histologic hours post fertilization. While duct and endocrine cells subtype and response to treatment - the concept of a develop normally, the acinar compartment undergoes complex, hierarchically organized pancreatic epithelium with complete apoptosis. Interestingly, loss of Dnmt1 leads to variable susceptibility towards oncogenic insults seems enhanced regeneration of pancreatic beta cells after targeted plausible. ablation. These early studies underscored the complex roles of epigenetic regulation in development and regeneration [54]. Mechanisms of Cellular Plasticity In injury, the epigenetic regulator Bmi1, a member of the polycomb repressor complex 1 (Prc1), is upregulated in Above we have aimed to outline, that cellular plasticity is acinar cells and coordinates regeneration in the exocrine an important mechanism in pancreatic regeneration and compartment. Moreover, loss of Bmi1 greatly impairs cancer development. The loss of cellular identity is exocrine regeneration after pancreatic injury. This is in part orchestrated on a genetic and epigenetic level. due to upregulation of Cdkn2a and p53-regulated Mechanistically, Sox9 seems to play a pivotal role in the proapoptotic genes [55]. Ezh2 is polycomb group protein and reprogramming of acinar cells towards dedifferentiation and part of the polycomb repressor complex 2 (Prc2). Following neoplasia. Sander and co-workers were able to show that loss pancreatic injury, Ezh2 mediates pancreatic regeneration by of the Sox9 protein prevented conversion of acinar cells into suppression of p16Ink4a. Suppression of p16Ink4a leads to PanINs; however Sox9 is not absolutely necessary for acinar enhanced proliferation of progenitor cells driving tissue to ductal metaplasia [39]. In contrast, forced overexpression of repair. Consequently, loss of Ezh2 causes failure of Sox9 in the setting of mutant Kras significantly enhanced pancreatic regeneration and accelerates pancreatic pancreatic tumorigenesis through downregulation of acinar tumorigenesis. The complex interplay of genetic and markers such as Mist1 [39]. Mist1 is a transcription factor epigenetic mechanisms mediating pancreatic plasticity and critically involved in acinar cell identity. Mist1 expression is repair have recently been excellently elsewhere [31, 56]. regulated by the transcription factor Gata6 and loss of Gata6 causes degeneration of the acinar compartment [46]. More Finally, the microenvironment plays a pivotal role in importantly, Gata6 controls multiple differentiation programs pancreatic regeneration and tumorigenesis by influencing the in the adult pancreas and counteracts Kras-driven epithelial compartment [57-59]. In response to an injurious tumorigenesis [47]. Furthermore, we were able to show that stimulus, acinar cells release proinflammatory factors such as the transcription factor Prrx1b binds to the Sox9 promoter Interleukin 6 (IL-6), tumor necrosis factor (TNF) and positively regulating Sox9 expression and thereby promoting transforming growth factor-beta (TGF-beta), which in turn cellular plasticity. Importantly, Dclk1+ cells express high activate the immune system but also otherwise quiescent levels of Sox9 under resting conditions (unpublished pancreatic stellate cells (PSC) [60]. Activated stellate cells observations), offering a potential explanation for their high secrete a variety of factors such as insulin-like growth factor, degree of plasticity and transformability. Insight into the epidermal growth factor, growth factor,

Page 4 of 8

Cancer Cell & Microenvironment 2016; 3: e1472. doi: 10.14800/ccm.1472; © 2016 by C. Benedikt Westphalen, et al. http://www.smartscitech.com/index.php/ccm

Nodal/Activin and platelet derived growth factor. These compartment are collectively mutated to induce pancreatic mediators in turn lead to increased aggressiveness of tumorigenesis. A major advance in the field would be the surrounding cancer cells [61-65]. The importance of this generation of spontaneous models that do not rely on broad crosstalk between PSC and pancreatic cancer cells is Cre-drivers to achieve malignant transformation. Interesting demonstrated by the fact, that restoration of PSC quiescence new in vivo model systems are on the rise that combine profoundly affects cancer cells in proximity [66]. Furthermore, genomic engineering and at the same time recapitulate the we were able to show that an epithelial-stromal crosstalk stochastic nature of the disease [74]. Given the fact that some mediated by the transcription factor ETV1 regulates stromal forms of chronic pancreatitis pose an increased risk for the expansion, epithelial to mesenchymal transition and development of pancreatic cancer in humans we can only metastatic spread underscoring the importance of the stromal speculate that the same mechanisms driving malignancy in compartment in regulating epithelial plasticity [67]. mice also dominate the process in humans. Further studies Immunosurveillance plays as major role to eliminate mutant and novel technologies such as organoid cultures and cells before their expansion [68]. However, ongoing activation humanized animal models will be instrumental in translating of the immune system is an important driver of early these fundamental questions from mice into humans and pancreatic tumorigenesis, in part through the might then help to design future preventive and therapeutic NFATc1-STAT3 transcription complex [69]. Furthermore, approaches for pancreatic cancer. mutant Kras causes expression of GM-CSF which in turn leads to recruitment of immunosuppressive myeloid and Conflicting interests exclusion of cytotoxic T-cells [70, 71]. The authors have declared that no conflict of interests Conclusions and Outlook exists.

It is widely acknowledged that the most efficient cellular Acknowledgements origin of PDAC precursors lies within the acinar epithelium. Only recently, it was proposed that specific subsets of acinar This work was supported by the National Pancreas cells could exist that drive pancreatic regeneration (and Foundation (MR), the KKF Program and the TUM Junior tumorigenesis) [23]. Indeed, we were able to show that Dclk1 Fellow Program of the Technical University of Munich labels such a progenitor population involved in repair and (MR), the German Cancer Aid Foundation (Max-Eder cancer initiation [27]. Future studies are needed to establish if Program, Deutsche Krebshilfe 111273 to MR), and the the “plastic pancreas” [5] contains more than just one of these AGA-Actavis Research Award in Pancreatic Disorders progenitor/stem cell populations or if multiple different pools (MR). of cell types with different properties coexist in the pancreas. Of note, coexistence of different stem cell pools has been Abbrevations demonstrated in other organs such as the skin and the intestine [72, 73]. Additionally work from our groups has CK19: Cytokeratin 19; Dclk1: Doublecortin like kinase clearly demonstrated that a distinct subset of Prrx1+ ductal protein 1; ADM: acinar to ductal metaplasia; (m)PanIN: cells exists and that these are functionally different from the murine pancreatic intraepithelial neoplasia; PDAC: bulk of the ductal epithelium [11]. It is tempting to speculate pancreatic ductal adenocarcinoma; Sox9: SRY-box 9; Prxx1: that cancers arising in the setting of mutant Kras/p53 targeted Paired related homeobox 1; Hnf1b: HNF1 homeobox B; to the duct compartment (see above) originate from this IPMN: intraductal papillary mucinous neoplasia; ADR: specific population. acinar-to ductal reprogramming.

At this point it is important to acknowledge that all of the References evidence for potential origins of pancreatic cancer that is 1. Kopp JL, Grompe M, and Sander M, Stem cells versus plasticity presented here is based on genetically engineered mouse in liver and pancreas regeneration. Nat Cell Biol 2016; 3:238-245. models in which forced expression of oncogenes to different 2. Kong B, Michalski CW, Erkan M, Friess H, and Kleeff J, From compartments is the common approach to answer one of the tissue turnover to the cell of origin for pancreatic cancer. Nat Rev fundamental questions in pancreatic cancer research. While Gastroenterol Hepatol 2011; 8:467-472. genetically engineered mouse models closely resemble the 3. Puri S, Folias AE, and Hebrok M, Plasticity and dedifferentiation histology and biology of human pancreatic adenocarcinoma within the pancreas: development, homeostasis, and disease. Cell we do not know if the underlying mechanisms of sporadic stem cell 2015; 1:18-31. human pancreatic cancer mirror those in mice in which the 4. Sekine K and Taniguchi H, Basics and applications of stem cells whole pancreatic anlage or the majority of a given in the pancreas. J Hepatobiliary Pancreat Sci 2012; 6:594-599.

Page 5 of 8

Cancer Cell & Microenvironment 2016; 3: e1472. doi: 10.14800/ccm.1472; © 2016 by C. Benedikt Westphalen, et al. http://www.smartscitech.com/index.php/ccm

5. Ziv O, Glaser B, and Dor Y, The plastic pancreas. Dev Cell 2013; Jensen J, Recapitulation of elements of embryonic development in 1:3-7. adult mouse pancreatic regeneration. Gastroenterology 2005; 3:728-741. 6. Yanger K and Stanger BZ, Facultative stem cells in liver and pancreas: fact and fancy. Dev Dyn 2011; 3:521-529. 22. Sangiorgi E and Capecchi MR, Bmi1 is expressed in vivo in intestinal stem cells. Nat Genet 2008; 7:915-920. 7. Wang YJ, Bailey JM, Rovira M, and Leach SD, Sphere-forming assays for assessment of benign and malignant pancreatic stem 23. Sangiorgi E and Capecchi MR, Bmi1 lineage tracing identifies a cells. Methods Mol Biol 2013; 281-290. self-renewing pancreatic acinar cell subpopulation capable of maintaining pancreatic organ homeostasis. Proc Natl Acad Sci U S 8. Huch M, Bonfanti P, Boj SF, Sato T, Loomans CJ, van de A 2009; 17:7101-7106. Wetering M, et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J 24. May R, Sureban SM, Lightfoot SA, Hoskins AB, Brackett DJ, 2013; 20:2708-2721. Postier RG, et al. Identification of a novel putative pancreatic stem/progenitor cell marker DCAMKL-1 in normal mouse 9. Rovira M, Scott SG, Liss AS, Jensen J, Thayer SP, and Leach SD, pancreas. Am J Physiol Gastrointest Liver Physiol 2010; Isolation and characterization of centroacinar/terminal ductal 2:G303-310. progenitor cells in adult mouse pancreas. Proc Natl Acad Sci U S A 2010; 1:75-80. 25. May R, Sureban SM, Hoang N, Riehl TE, Lightfoot SA, Ramanujam R, et al. Doublecortin and CaM kinase-like-1 and 10. Reichert M and Rustgi AK, Pancreatic ductal cells in leucine-rich-repeat-containing G-protein-coupled receptor mark development, regeneration, and neoplasia. J Clin Invest 2011; quiescent and cycling intestinal stem cells, respectively. Stem 12:4572-4578. Cells 2009; 10:2571-2579. 11. Reichert M, Takano S, von Burstin J, Kim SB, Lee JS, 26. May R, Riehl TE, Hunt C, Sureban SM, Anant S, and Houchen Ihida-Stansbury K, et al. The Prrx1 homeodomain transcription CW, Identification of a novel putative gastrointestinal stem cell factor plays a central role in pancreatic regeneration and and adenoma stem cell marker, doublecortin and CaM carcinogenesis. Genes Dev 2013; 3:288-300. kinase-like-1, following radiation injury and in adenomatous 12. Greenbaum A, Hsu YM, Day RB, Schuettpelz LG, Christopher polyposis coli/multiple intestinal neoplasia mice. Stem Cells 2008; MJ, Borgerding JN, et al. CXCL12 in early mesenchymal 3:630-637. progenitors is required for haematopoietic stem-cell maintenance. 27. Westphalen CB, Takemoto Y, Tanaka T, Macchini M, Jiang Z, Nature 2013; 7440:227-230. Renz BW, et al. Dclk1 Defines Quiescent Pancreatic Progenitors 13. Means AL, Meszoely IM, Suzuki K, Miyamoto Y, Rustgi AK, that Promote Injury-Induced Regeneration and Tumorigenesis. Coffey RJ, Jr., et al. Pancreatic epithelial plasticity mediated by Cell stem cell 2016; 4:441-455. acinar cell transdifferentiation and generation of nestin-positive 28. Stanger BZ, Stiles B, Lauwers GY, Bardeesy N, Mendoza M, intermediates. Development 2005; 16:3767-3776. Wang Y, et al. Pten constrains centroacinar cell expansion and malignant transformation in the pancreas. Cancer Cell 2005; 14. Shi G, DiRenzo D, Qu C, Barney D, Miley D, and Konieczny SF, 3:185-195. Maintenance of acinar cell organization is critical to preventing Kras-induced acinar-ductal metaplasia. Oncogene 2013; 29. Kopinke D, Brailsford M, Pan FC, Magnuson MA, Wright CV, 15:1950-1958. and Murtaugh LC, Ongoing Notch signaling maintains phenotypic fidelity in the adult exocrine pancreas. Dev Biol 2012; 1:57-64. 15. Prevot PP, Simion A, Grimont A, Colletti M, Khalaileh A, Van den Steen G, et al. Role of the ductal transcription factors HNF6 30. Kopinke D, Brailsford M, Shea JE, Leavitt R, Scaife CL, and and Sox9 in pancreatic acinar-to-ductal metaplasia. Gut 2012; Murtaugh LC, Lineage tracing reveals the dynamic contribution of 12:1723-1732. Hes1+ cells to the developing and adult pancreas. Development 2011; 3:431-441. 16. Scotti ML, Smith KE, Butler AM, Calcagno SR, Crawford HC, Leitges M, et al. Protein kinase C iota regulates pancreatic 31. Roy N and Hebrok M, Regulation of Cellular Identity in Cancer. acinar-to-ductal metaplasia. PLoS One 2012; 2:e30509. Dev Cell 2015; 6:674-684. 17. Ebine K, Chow CR, DeCant BT, Hattaway HZ, Grippo PJ, Kumar 32. Murtaugh LC and Leach SD, A case of mistaken identity? K, et al. Slug inhibits pancreatic cancer initiation by blocking Nonductal origins of pancreatic "ductal" cancers. Cancer Cell Kras-induced acinar-ductal metaplasia. Sci Rep 2016; 29133. 2007; 3:211-213. 18. Strobel O, Dor Y, Alsina J, Stirman A, Lauwers G, Trainor A, et 33. Gidekel Friedlander SY, Chu GC, Snyder EL, Girnius N, Dibelius al. In vivo lineage tracing defines the role of acinar-to-ductal G, Crowley D, et al. Context-dependent transformation of adult transdifferentiation in inflammatory ductal metaplasia. pancreatic cells by oncogenic K-Ras. Cancer Cell 2009; Gastroenterology 2007; 6:1999-2009. 5:379-389. 19. Morris JPt, Cano DA, Sekine S, Wang SC, and Hebrok M, 34. Westphalen CB and Olive KP, Genetically engineered mouse Beta-catenin blocks Kras-dependent reprogramming of acini into models of pancreatic cancer. Cancer J 2012; 6:502-510. pancreatic cancer precursor lesions in mice. J Clin Invest 2010; 35. Habbe N, Shi G, Meguid RA, Fendrich V, Esni F, Chen H, et al. 2:508-520. Spontaneous induction of murine pancreatic intraepithelial 20. Fendrich V, Esni F, Garay MV, Feldmann G, Habbe N, Jensen JN, neoplasia (mPanIN) by acinar cell targeting of oncogenic Kras in et al. Hedgehog signaling is required for effective regeneration of adult mice. Proc Natl Acad Sci U S A 2008; 48:18913-18918. exocrine pancreas. Gastroenterology 2008; 2:621-631. 36. Reichert M, Blume K, Kleger A, Hartmann D, and von Figura G, 21. Jensen JN, Cameron E, Garay MV, Starkey TW, Gianani R, and Developmental Pathways Direct Pancreatic Cancer Initiation from

Page 6 of 8

Cancer Cell & Microenvironment 2016; 3: e1472. doi: 10.14800/ccm.1472; © 2016 by C. Benedikt Westphalen, et al. http://www.smartscitech.com/index.php/ccm

Its Cellular Origin. Stem cells international 2016; 9298535. 21:4585-4595. 37. Guerra C, Collado M, Navas C, Schuhmacher AJ, 52. Vooijs M, Liu Z, and Kopan R, Notch: architect, landscaper, and Hernandez-Porras I, Canamero M, et al. Pancreatitis-induced guardian of the intestine. Gastroenterology 2011; 2:448-459. inflammation contributes to pancreatic cancer by inhibiting 53. Siveke JT, Lubeseder-Martellato C, Lee M, Mazur PK, Nakhai H, oncogene-induced senescence. Cancer Cell 2011; 6:728-739. Radtke F, et al. Notch signaling is required for exocrine 38. Guerra C, Schuhmacher AJ, Canamero M, Grippo PJ, Verdaguer regeneration after acute pancreatitis. Gastroenterology 2008; L, Perez-Gallego L, et al. Chronic pancreatitis is essential for 2:544-555. induction of pancreatic ductal adenocarcinoma by K-Ras 54. Anderson RM, Bosch JA, Goll MG, Hesselson D, Dong PD, Shin oncogenes in adult mice. Cancer Cell 2007; 3:291-302. D, et al. Loss of Dnmt1 catalytic activity reveals multiple roles for 39. Kopp JL, von Figura G, Mayes E, Liu FF, Dubois CL, Morris JPt, DNA methylation during pancreas development and regeneration. et al. Identification of Sox9-dependent acinar-to-ductal Dev Biol 2009; 1:213-223. reprogramming as the principal mechanism for initiation of 55. Fukuda A, Morris JPt, and Hebrok M, Bmi1 is required for pancreatic ductal adenocarcinoma. Cancer Cell 2012; 6:737-750. regeneration of the exocrine pancreas in mice. Gastroenterology 40. Bailey JM, Alsina J, Rasheed ZA, McAllister FM, Fu YY, Plentz 2012; 3:821-831 e821-822. R, et al. DCLK1 marks a morphologically distinct subpopulation 56. Quilichini E and Haumaitre C, Implication of epigenetics in of cells with stem cell properties in preinvasive pancreatic cancer. pancreas development and disease. Best Pract Res Clin Endocrinol Gastroenterology 2014; 1:245-256. Metab 2015; 6:883-898. 41. Ray KC, Bell KM, Yan J, Gu G, Chung CH, Washington MK, et 57. Feig C, Gopinathan A, Neesse A, Chan DS, Cook N, and Tuveson al. Epithelial tissues have varying degrees of susceptibility to DA, The pancreas cancer microenvironment. Clin Cancer Res Kras(G12D)-initiated tumorigenesis in a mouse model. PLoS One 2012; 16:4266-4276. 2011; 2:e16786. 58. Stromnes IM, DelGiorno KE, Greenberg PD, and Hingorani SR, 42. Bailey JM, Hendley AM, Lafaro KJ, Pruski MA, Jones NC, Alsina Stromal reengineering to treat pancreas cancer. Carcinogenesis J, et al. p53 mutations cooperate with oncogenic Kras to promote 2014; 7:1451-1460. adenocarcinoma from pancreatic ductal cells. Oncogene 2016; 32:4282-4288. 59. Waghray M, Yalamanchili M, di Magliano MP, and Simeone DM, Deciphering the role of stroma in pancreatic cancer. Curr Opin 43. Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Gastroenterol 2013; 5:537-543. Hruban RH, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic 60. Neesse A, Algul H, Tuveson DA, and Gress TM, Stromal biology ductal adenocarcinoma in mice. Cancer Cell 2005; 5:469-483. and therapy in pancreatic cancer: a changing paradigm. Gut 2015; 9:1476-1484. 44. Boj SF, Hwang CI, Baker LA, Chio, II, Engle DD, Corbo V, et al. Organoid models of human and mouse ductal pancreatic cancer. 61. Kikuta K, Masamune A, Watanabe T, Ariga H, Itoh H, Hamada S, et al. Pancreatic stellate cells promote epithelial-mesenchymal Cell 2015; 1-2:324-338. transition in pancreatic cancer cells. Biochem Biophys Res 45. Miyamoto Y, Maitra A, Ghosh B, Zechner U, Argani P, Commun 2010; 3-4:380-384. Iacobuzio-Donahue CA, et al. Notch mediates TGF alpha-induced 62. Lonardo E, Frias-Aldeguer J, Hermann PC, and Heeschen C, changes in epithelial differentiation during pancreatic Pancreatic stellate cells form a niche for cancer stem cells and tumorigenesis. Cancer Cell 2003; 6:565-576. promote their self-renewal and invasiveness. Cell cycle 2012; 46. Martinelli P, Canamero M, del Pozo N, Madriles F, Zapata A, and 7:1282-1290. Real FX, Gata6 is required for complete acinar differentiation and 63. Apte MV, Wilson JS, Lugea A, and Pandol SJ, A starring role for maintenance of the exocrine pancreas in adult mice. Gut 2013; stellate cells in the pancreatic cancer microenvironment. 10:1481-1488. Gastroenterology 2013; 6:1210-1219. 47. Martinelli P, Madriles F, Canamero M, Pau EC, Pozo ND, Guerra 64. Vonlaufen A, Phillips PA, Xu Z, Goldstein D, Pirola RC, Wilson C, et al. The acinar regulator Gata6 suppresses KrasG12V-driven JS, et al. Pancreatic stellate cells and pancreatic cancer cells: an pancreatic tumorigenesis in mice. Gut 2016; 3:476-486. unholy alliance. Cancer Res 2008; 19:7707-7710. 48. di Magliano MP and Logsdon CD, Roles for KRAS in pancreatic 65. Vonlaufen A, Joshi S, Qu C, Phillips PA, Xu Z, Parker NR, et al. tumor development and progression. Gastroenterology 2013; Pancreatic stellate cells: partners in crime with pancreatic cancer 6:1220-1229. cells. Cancer Res 2008; 7:2085-2093. 49. Ying H, Kimmelman AC, Lyssiotis CA, Hua S, Chu GC, 66. Froeling FE, Feig C, Chelala C, Dobson R, Mein CE, Tuveson Fletcher-Sananikone E, et al. Oncogenic Kras maintains DA, et al. Retinoic acid-induced quiescence pancreatic tumors through regulation of anabolic glucose reduces paracrine Wnt-beta-catenin signaling to slow tumor metabolism. Cell 2012; 3:656-670. progression. Gastroenterology 2011; 4:1486-1497, 1497 50. Collins MA, Bednar F, Zhang Y, Brisset JC, Galban S, Galban CJ, e1481-1414. et al. Oncogenic Kras is required for both the initiation and 67. Heeg S, Das KK, Reichert M, Bakir B, Takano S, Caspers J, et al. maintenance of pancreatic cancer in mice. J Clin Invest 2012; ETS-Transcription Factor ETV1 Regulates Stromal Expansion and 2:639-653. Metastasis in Pancreatic Cancer. Gastroenterology 2016; 51. Perdigoto CN, Schweisguth F, and Bardin AJ, Distinct levels of 3:540-553 e514. Notch activity for commitment and terminal differentiation of 68. Clark CE, Beatty GL, and Vonderheide RH, Immunosurveillance stem cells in the adult fly intestine. Development 2011; of pancreatic adenocarcinoma: insights from genetically

Page 7 of 8

Cancer Cell & Microenvironment 2016; 3: e1472. doi: 10.14800/ccm.1472; © 2016 by C. Benedikt Westphalen, et al. http://www.smartscitech.com/index.php/ccm

engineered mouse models of cancer. Cancer Lett 2009; 1:1-7. factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 2012; 6:822-835. 69. Baumgart S, Chen NM, Siveke JT, Konig A, Zhang JS, Singh SK, et al. Inflammation-induced NFATc1-STAT3 transcription 72. Jaks V, Kasper M, and Toftgard R, The hair follicle-a stem cell complex promotes pancreatic cancer initiation by KrasG12D. zoo. Exp Cell Res 2010; 8:1422-1428. Cancer Discov 2014; 6:688-701. 73. Stange DE and Clevers H, Concise review: the yin and yang of 70. Pylayeva-Gupta Y, Lee KE, Hajdu CH, Miller G, and Bar-Sagi D, intestinal (cancer) stem cells and their progenitors. Stem Cells Oncogenic Kras-induced GM-CSF production promotes the 2013; 11:2287-2295. development of pancreatic neoplasia. Cancer Cell 2012; 6:836-847. 74. Maresch R, Mueller S, Veltkamp C, Ollinger R, Friedrich M, Heid I, et al. Multiplexed pancreatic genome engineering and cancer 71. Bayne LJ, Beatty GL, Jhala N, Clark CE, Rhim AD, Stanger BZ, induction by transfection-based CRISPR/Cas9 delivery in mice. et al. Tumor-derived granulocyte-macrophage colony-stimulating Nature communications 2016; 10770.

Page 8 of 8