Gastroenterology

Gastroenterology

Volume 159, Issue 5, November 2020, Pages 1866-1881.e8
Gastroenterology

Original Research
Basic and Translational—Pancreas
Tuft Cells Inhibit Pancreatic Tumorigenesis in Mice by Producing Prostaglandin D2

https://doi.org/10.1053/j.gastro.2020.07.037Get rights and content

Background & Aims

Development of pancreatic ductal adenocarcinoma (PDA) involves acinar to ductal metaplasia and genesis of tuft cells. It has been a challenge to study these rare cells because of the lack of animal models. We investigated the role of tuft cells in pancreatic tumorigenesis.

Methods

We performed studies with LSL-KrasG12D/+;Ptf1aCre/+ mice (KC; develop pancreatic tumors), KC mice crossed with mice with pancreatic disruption of Pou2f3 (KPouC mice; do not develop tuft cells), or mice with pancreatic disruption of the hematopoietic prostaglandin D synthase gene (Hpgds, KHC mice) and wild-type mice. Mice were allowed to age or were given caerulein to induce pancreatitis; pancreata were collected and analyzed by histology, immunohistochemistry, RNA sequencing, ultrastructural microscopy, and metabolic profiling. We performed laser-capture dissection and RNA-sequencing analysis of pancreatic tissues from 26 patients with pancreatic intraepithelial neoplasia (PanIN), 19 patients with intraductal papillary mucinous neoplasms (IPMNs), and 197 patients with PDA.

Results

Pancreata from KC mice had increased formation of tuft cells and higher levels of prostaglandin D2 than wild-type mice. Pancreas-specific deletion of POU2F3 in KC mice (KPouC mice) resulted in a loss of tuft cells and accelerated tumorigenesis. KPouC mice had increased fibrosis and activation of immune cells after administration of caerulein. Pancreata from KPouC and KHC mice had significantly lower levels of prostaglandin D2, compared with KC mice, and significantly increased numbers of PanINs and PDAs. KPouC and KHC mice had increased pancreatic injury after administration of caerulein, significantly less normal tissue, more extracellular matrix deposition, and higher PanIN grade than KC mice. Human PanIN and intraductal papillary mucinous neoplasm had gene expression signatures associated with tuft cells and increased expression of Hpgds messenger RNA compared with PDA.

Conclusions

In mice with KRAS-induced pancreatic tumorigenesis, loss of tuft cells accelerates tumorigenesis and increases the severity of caerulein-induced pancreatic injury, via decreased production of prostaglandin D2. These data are consistent with the hypothesis that tuft cells are a metaplasia-induced tumor attenuating cell type.

Section snippets

Mice

Mice were housed in accordance with National Institutes of Health guidelines in American Association for Accreditation of Laboratory Animal Care-accredited facilities at the Salk Institute for Biological Studies. The Salk Institute Institutional Animal Care and Use Committee IACUC approved all animal studies. LSL-KrasG12D/+;Ptf1aCre/+ (KC) and Hpgdsfl/fl mice have been previously described.16,17 FLARE25 (Il25F25/F25) mice were generously provided by the Locksley (University of California–San

Pou2f3 Ablation Accelerates Tumorigenesis

Transcription factor Pou2f3 is the master regulator of tuft cell formation in several organs, and its expression has recently been identified in PanIN.6,18,19 We therefore used pancreas-specific Pou2f3 deletion as a genetic strategy to investigate tuft cell contribution to tumor progression. The specificity of this strategy is supported by the fact that 99% of tuft cells in the LSL-KrasG12D;Ptf1aCre/+ (KC) mouse model of pancreatic tumorigenesis, identified by coexpression of tuft cell markers

Discussion

Despite much conjecture over a protumorigenic role for tuft cells in pancreatic tumorigenesis, we have found that eliminating tuft cells through Pou2f3 ablation accelerates tumor formation, indicating a protective role for this cell type. We propose that tuft cells, rather than seeding cancer, instill homeostasis, in part by generating and secreting lipid eicosanoid mediators. Consistent with this, our ultrastructural analyses identified the previously undescribed presence of lipid droplets in

Acknowledgments

The authors thank Eugene Ke, Alexis Roth, Annie Odelson, Yesol Go, and Gidsela Luna for technical expertise; Haiyong Han and Daniel Von Hoff for human samples; and Jakob von Moltke, Howard Crawford, Meggie Hoffman, and members of the Wahl laboratory for helpful discussions.

Kathleen E. DelGiorno’s current affiliation is the Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee. Chi-Yeh Chung’s and Rajshekhar R. Giraddi’s current affiliation is Pfizer Inc, San

References (40)

  • A. Sato

    Tuft cells

    Anat Sci Int

    (2007)
  • O. Jarvi et al.

    On the cellular structures of the epithelial invasions in the glandular stomach of mice caused by intramural application of 20-methylcholantren

    Acta Pathol Microbiol Scand Suppl

    (1956)
  • F. Gerbe et al.

    Intestinal epithelial tuft cells initiate type 2 mucosal immunity to helminth parasites

    Nature

    (2016)
  • M.R. Howitt et al.

    Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the gut

    Science

    (2016)
  • J. von Moltke et al.

    Tuft-cell-derived IL-25 regulates an intestinal ILC2-epithelial response circuit

    Nature

    (2016)
  • W. Lei et al.

    Activation of intestinal tuft cell-expressed Sucnr1 triggers type 2 immunity in the mouse small intestine

    Proc Natl Acad Sci U S A

    (2018)
  • M.S. Nadjsombati et al.

    Detection of succinate by intestinal tuft cells triggers a type 2 innate immune circuit

    Immunity

    (2018)
  • C. Schneider et al.

    A metabolite-triggered tuft cell-ILC2 circuit drives small intestinal remodeling

    Cell

    (2018)
  • C.N. Miller et al.

    Thymic tuft cells promote an IL-4-enriched medulla and shape thymocyte development

    Nature

    (2018)
  • C. Bornstein et al.

    Single-cell mapping of the thymic stroma identifies IL-25-producing tuft epithelial cells

    Nature

    (2018)
  • Cited by (0)

    Conflicts of interest The authors disclose no conflicts.

    Funding Salk Core Facilities are supported, in part, by the National Institutes of Health (NIH)/National Cancer Institute (NCI) CCSG: P30 014195. The Salk Next Generation Sequencing Core is additionally supported by the Chapman Foundation and the Helmsley Charitable Trust. The Salk Waitt Advanced Biophotonics Core Facility is additionally supported by the Waitt Foundation. The Flow Cytometry Core Facility of the Salk Institute is additionally funded by Shared Instrumentation Grant S10-OD023 (Aria Fusion cell sorter). The Salk IGC is also supported by The Helmsley Charitable Trust, and Maxim Nikolaievich Shokhirev is supported by R01 GM102491-07 and 1RF1AG064049-01. Yoshihiro Urade is supported by a JSPS KAKENHI grant (16H01881). Ichiro Matsumoto and Makoto Ohmoto are supported by NIH R01 DC015491 and the Monell Chemical Senses Center. Kenneth P. Olive and H. Carlo Maurer are supported by NIH/NCI CCSG P30CA013696, NIH/NCI 1U54CA209997, and the Columbia/NYP Pancreas Center. Pankaj K. Singh’s laboratory is supported by the following NIH/NCI awards R01CA163649, R01CA210439, R01CA216853, P50CA127297, P01CA217798, and P30 CA036727. Geoffrey M. Wahl’s laboratory is supported by NIH/NCI CCSG P30 014195, NIH R35 CA197687, The Leona M. and Harry B. Helmsley Charitable Trust (2012-PG-MED002), the Freeberg Foundation, the Copley Foundation, and The William H. Isacoff MD Research Foundation for Gastrointestinal Cancer. Kathleen E. DelGiorno was supported by a T32 training grant (5T32CA9370-34), the Salk Pioneer Award, the Salk Women & Science Special Award, and a Hirshberg Foundation Seed Grant.

    View full text