Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Muc16 depletion diminishes KRAS-induced tumorigenesis and metastasis by altering tumor microenvironment factors in pancreatic ductal adenocarcinoma

Abstract

MUC16, membrane-bound mucin, plays an oncogenic role in pancreatic ductal adenocarcinoma (PDAC). However, the pathological role of MUC16 in the PDAC progression, tumor microenvironment, and metastasis in cooperation with KrasG12D and Trp53R172H mutations remains unknown. Deletion of Muc16 with activating mutations KrasG12D/+ and Trp53R172H/+ in mice significantly decreased progression and prolonged overall survival in KrasG12D/+; Trp53R172H/+; Pdx-1-Cre; Muc16−/− (KPCM) and KrasG12D/+; Pdx-1-Cre; Muc16−/− (KCM), as compared to KrasG12D/+; Trp53R172H/+; Pdx-1-Cre (KPC) and KrasG12D/+; Pdx-1-Cre (KC) mice, respectively. Muc16 knockout pancreatic tumor (KPCM) displays decreased tumor microenvironment factors and significantly reduced incidence of liver and lung metastasis compared to KPC. Furthermore, in silico data analysis showed a positive correlation of MUC16 with activated stroma and metastasis-associated genes. KPCM mouse syngeneic cells had significantly lower metastatic and endothelial cell binding abilities than KPC cells. Similarly, KPCM organoids significantly decreased the growth rate compared to KPC organoids. Interestingly, RNA-seq data revealed that the cytoskeletal proteins Actg2, Myh11, and Pdlim3 were downregulated in KPCM tumors. Further knockdown of these genes showed reduced metastatic potential. Overall, our results demonstrate that Muc16 alters the tumor microenvironment factors during pancreatic cancer progression and metastasis by changing the expression of Actg2, Myh11, and Pdlim3 genes.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Establishment of Muc16 mediated pancreatic cancer mouse models.
Fig. 2: Muc16 knockout mice displayed reduced PDAC metastasis.
Fig. 3: Muc16 decreased the fibrosis and ECM content in pancreatic tumors.
Fig. 4: Muc16 in endothelial cell binding, p-selectin, colony formation, and cell migration abilities in pancreatic cancer.
Fig. 5: Transcriptome analysis, correlation of MUC16 and its gene signature in PDAC.
Fig. 6: Muc16 associated gene signatures in pancreatic cancer and their role in metastasis.
Fig. 7: Schematic diagram of Muc16 mediated tumor progression, altered tumor microenvironment, and metastasis in PDAC.

Similar content being viewed by others

Data availability

The accession number for RNA sequencing data of Muc16 knockout pancreatic tumor tissues is GSE212777, and materials associated with the current study are available from the corresponding author upon reasonable request.

References

  1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA: Cancer J Clin. 2021;71:7–33.

    PubMed  Google Scholar 

  2. Hosein AN, Brekken RA, Maitra A. Pancreatic cancer stroma: an update on therapeutic targeting strategies. Nat Rev Gastroenterol Hepatol. 2020;17:487–505.

  3. Encarnación-Rosado J, Kimmelman AC. Harnessing metabolic dependencies in pancreatic cancers. Nat Rev Gastroenterol Hepatol. 2021;18:482–92.

  4. Buscail L, Bournet B, Cordelier P. Role of oncogenic KRAS in the diagnosis, prognosis and treatment of pancreatic cancer. Nat Rev Gastroenterol Hepatol. 2020;17:153–68.

    Article  CAS  PubMed  Google Scholar 

  5. Benjamin JR, Ralph HH, Andrew JA, Richard AM, Jen JY, Chip S, et al. Integrated Genomic Characterization of Pancreatic Ductal Adenocarcinoma. Cancer Cell. 2017;32:185–203.e13.

  6. Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015;518:495–501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Tian C, Huang Y, Clauser KR, Rickelt S, Lau AN, Carr SA, et al. Suppression of pancreatic ductal adenocarcinoma growth and metastasis by fibrillar collagens produced selectively by tumor cells. Nat Commun. 2021;12:2328.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tian C, Clauser KR, Öhlund D, Rickelt S, Huang Y, Gupta M, et al. Proteomic analyses of ECM during pancreatic ductal adenocarcinoma progression reveal different contributions by tumor and stromal cells. Proc Natl Acad Sci USA. 2019;116:19609–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bafna S, Kaur S, Momi N, Batra SK. Pancreatic cancer cells resistance to gemcitabine: the role of MUC4 mucin. Br J Cancer. 2009;101:1155–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kaur S, Kumar S, Momi N, Sasson AR, Batra SK. Mucins in pancreatic cancer and its microenvironment. Nat Rev Gastroenterol Hepatol. 2013;10:607–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Caffrey T, Sagar S, Thomas D, Lewallen ME, Hollingsworth MA, Radhakrishnan P. The glycoprotein mucin-1 negatively regulates GalNAc transferase 5 expression in pancreatic cancer. FEBS Lett. 2019;593:2751–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Chen SH, Dallas MR, Balzer EM, Konstantopoulos K. Mucin 16 is a functional selectin ligand on pancreatic cancer cells. FASEB J: Off Publ Fed Am Soc Exp Biol. 2012;26:1349–59.

    Article  CAS  Google Scholar 

  13. Chen SH, Hung WC, Wang P, Paul C, Konstantopoulos K. Mesothelin binding to CA125/MUC16 promotes pancreatic cancer cell motility and invasion via MMP-7 activation. Sci Rep. 2013;3:1870.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Das S, Rachagani S, Torres-Gonzalez MP, Lakshmanan I, Majhi PD, Smith LM, et al. Carboxyl-terminal domain of MUC16 imparts tumorigenic and metastatic functions through nuclear translocation of JAK2 to pancreatic cancer cells. Oncotarget 2015;6:5772–87.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Felder M, Kapur A, Gonzalez-Bosquet J, Horibata S, Heintz J, Albrecht R, et al. MUC16 (CA125): tumor biomarker to cancer therapy, a work in progress. Mol Cancer. 2014;13:129.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Haridas D, Chakraborty S, Ponnusamy MP, Lakshmanan I, Rachagani S, Cruz E, et al. Pathobiological implications of MUC16 expression in pancreatic cancer. PLoS One. 2011;6:e26839.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Lakshmanan I, Ponnusamy MP, Das S, Chakraborty S, Haridas D, Mukhopadhyay P, et al. MUC16 induced rapid G2/M transition via interactions with JAK2 for increased proliferation and anti-apoptosis in breast cancer cells. Oncogene 2012;31:805–17.

    Article  CAS  PubMed  Google Scholar 

  18. Lakshmanan I, Salfity S, Seshacharyulu P, Rachagani S, Thomas A, Das S, et al. MUC16 regulates TSPYL5 for lung cancer cell growth and chemoresistance by suppressing p53. Clin Cancer Res. 2017;23:3906–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Das S, Batra SK. Understanding the unique attributes of MUC16 (CA125): Potential implications in targeted therapy. Cancer Res. 2015;75:4669–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Gubbels JA, Belisle J, Onda M, Rancourt C, Migneault M, Ho M, et al. Mesothelin-MUC16 binding is a high affinity, N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors. Mol Cancer. 2006;5:50.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Akita K, Tanaka M, Tanida S, Mori Y, Toda M, Nakada H. CA125/MUC16 interacts with Src family kinases, and over-expression of its C-terminal fragment in human epithelial cancer cells reduces cell-cell adhesion. Eur J Cell Biol. 2013;92:257–63.

    Article  CAS  PubMed  Google Scholar 

  22. Muniyan S, Haridas D, Chugh S, Rachagani S, Lakshmanan I, Gupta S, et al. MUC16 contributes to the metastasis of pancreatic ductal adenocarcinoma through focal adhesion mediated signaling mechanism. Genes cancer. 2016;7:110–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Belisle JA, Horibata S, Jennifer GA, Petrie S, Kapur A, Andre S, et al. Identification of Siglec-9 as the receptor for MUC16 on human NK cells, B cells, and monocytes. Mol Cancer. 2010;9:118.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Lee JW, Komar CA, Bengsch F, Graham K, Beatty GL. Genetically engineered mouse models of pancreatic cancer: The KPC Model (LSL-Kras(G12D/+);LSL-Trp53(R172H/+);Pdx-1-Cre), its variants, and their application in immuno-oncology drug discovery. Curr Protoc Pharm. 2016;73:14.39.1–14.39.20.

    Article  Google Scholar 

  25. Nimmakayala RK, Leon F, Rachagani S, Rauth S, Nallasamy P, Marimuthu S, et al. Metabolic programming of distinct cancer stem cells promotes metastasis of pancreatic ductal adenocarcinoma. Oncogene 2021;40:215–31.

    Article  CAS  PubMed  Google Scholar 

  26. Chugh S, Barkeer S, Rachagani S, Nimmakayala RK, Perumal N, Pothuraju R, et al. Disruption of C1galt1 gene promotes development and metastasis of pancreatic adenocarcinomas in mice. Gastroenterology 2018;155:1608–24.

    Article  CAS  PubMed  Google Scholar 

  27. Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005;7:469–83.

    Article  CAS  PubMed  Google Scholar 

  28. Lakshmanan I, Seshacharyulu P, Haridas D, Rachagani S, Gupta S, Joshi S, et al. Novel HER3/MUC4 oncogenic signaling aggravates the tumorigenic phenotypes of pancreatic cancer cells. Oncotarget 2015;6:21085–99.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Boj SF, Hwang CI, Baker LA, Chio II, Engle DD, Corbo V, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015;160:324–38.

    Article  CAS  PubMed  Google Scholar 

  30. Mallya K, Haridas D, Seshacharyulu P, Pothuraju R, Junker WM, Krishn SR, et al. Acinar transformed ductal cells exhibit differential mucin expression in a tamoxifen-induced pancreatic ductal adenocarcinoma mouse model. Biol Open. 2020;9:bio052878.

  31. Cheon DJ, Wang Y, Deng JM, Lu Z, Xiao L, Chen CM, et al. CA125/MUC16 is dispensable for mouse development and reproduction. PLoS One. 2009;4:e4675.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Kaushik G, Seshacharyulu P, Rauth S, Nallasamy P, Rachagani S, Nimmakayala RK, et al. Selective inhibition of stemness through EGFR/FOXA2/SOX9 axis reduces pancreatic cancer metastasis. Oncogene 2021;40:848–62.

    Article  CAS  PubMed  Google Scholar 

  33. Rauth S, Karmakar S, Batra SK, Ponnusamy MP. Recent advances in organoid development and applications in disease modeling. Biochim Biophys Acta Rev Cancer. 2021;1875:188527.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lakshmanan I, Rachagani S, Hauke R, Krishn SR, Paknikar S, Seshacharyulu P, et al. MUC5AC interactions with integrin β4 enhances the migration of lung cancer cells through FAK signaling. Oncogene 2016;35:4112–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Öhlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med. 2017;214:579–96.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Li G, Kim YJ, Mantel C, Broxmeyer HE. P-selectin enhances generation of CD14+CD16+ dendritic-like cells and inhibits macrophage maturation from human peripheral blood monocytes. J Immunol. 2003;171:669–77.

    Article  CAS  PubMed  Google Scholar 

  37. Lennon S, Oweida A, Milner D, Phan AV, Bhatia S, Van Court B, et al. Pancreatic tumor microenvironment modulation by EphB4-ephrinB2 inhibition and radiation combination. Clin Cancer Res. 2019;25:3352–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Mizrahi JD, Surana R, Valle JW, Shroff RT. Pancreatic cancer. Lancet 2020;395:2008–20.

    Article  CAS  PubMed  Google Scholar 

  39. Das S, Majhi PD, Al-Mugotir MH, Rachagani S, Sorgen P, Batra SK. Membrane proximal ectodomain cleavage of MUC16 occurs in the acidifying Golgi/post-Golgi compartments. Sci Rep. 2015;5:9759.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Marimuthu S, Lakshmanan I, Muniyan S, Gautam SK, Nimmakayala RK, Rauth S, et al. MUC16 promotes liver metastasis of pancreatic ductal adenocarcinoma by upregulating NRP2-associated cell adhesion. Mol Cancer Res. 2022;20:1208–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Nallasamy P, Nimmakayala RK, Karmakar S, Leon F, Seshacharyulu P, Lakshmanan I, et al. Pancreatic tumor microenvironment factor promotes cancer stemness via SPP1-CD44 axis. Gastroenterology 2021;161:1998–2013.e7.

    Article  CAS  PubMed  Google Scholar 

  42. Sperb N, Tsesmelis M, Wirth T. Crosstalk between tumor and stromal cells in pancreatic ductal adenocarcinoma. Int J Mol Sci. 2020;21:5486.

  43. Connor AA, Denroche RE, Jang GH, Lemire M, Zhang A, Chan-Seng-Yue M, et al. Integration of genomic and transcriptional features in pancreatic cancer reveals increased cell cycle progression in metastases. Cancer Cell. 2019;35:267–82.e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Alistar A, Morris BB, Desnoyer R, Klepin HD, Hosseinzadeh K, Clark C, et al. Safety and tolerability of the first-in-class agent CPI-613 in combination with modified FOLFIRINOX in patients with metastatic pancreatic cancer: a single-centre, open-label, dose-escalation, phase 1 trial. Lancet Oncol. 2017;18:770–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hingorani SR, Zheng L, Bullock AJ, Seery TE, Harris WP, Sigal DS, et al. HALO 202: Randomized Phase II Study of PEGPH20 Plus Nab-Paclitaxel/Gemcitabine versus Nab-Paclitaxel/Gemcitabine in patients with untreated, metastatic pancreatic ductal adenocarcinoma. J Clin Oncol. 2018;36:359–66.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Dr. Richard R. Behringer, Department of Genetics, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, for providing the Muc16 whole-body knockout (C57BL/6J) mice. We thank Corinn Grabow for all the technical support. We thank the University of Nebraska Medical Center core facilities such as Advanced Microscopy Core Facility, Genomics Core Facility, and Tissue Science Facility. This work was, in parts, supported by the support from the National Institutes of Health P01 CA217798, R01 CA263575, R01 CA256973, R01 CA273349, R01 CA247471, R01 CA254036, R01 CA206444, R01 CA210637, R01 CA228524, F99 CA234962, U01 CA200466, and U01 CA210240, and the Nebraska Department of Health and Human Services LB595, US Department of Veterans Affairs (I01 BX004676).

Author information

Authors and Affiliations

Authors

Contributions

IL, MPP, and SKB conceived and designed the experiments. IL and SM performed the experiments. IL, SM, PS, and SC assisted with in vivo experiments. Data were collected and analyzed by IL, SM, SC, PS, SR, SM, RCV, PA, SR, RKN, JAS, SKG, AS, GN, SP, NB, KM, DH, GAT, LMS, SK, AKG, MJ, MPP. The manuscript was written by IL and SM with input from MPP, MJ, and SKB and reviewed by all authors. Statistical analysis and IHC scoring were done by LMS and GAT, respectively. All authors have read and approved the final manuscript.

Corresponding authors

Correspondence to Moorthy P. Ponnusamy or Surinder K. Batra.

Ethics declarations

Competing interests

SKB is one of the co-founders of Sanguine. Diagnostics and Therapeutics, Inc. The other authors declare no potential conflicts of interest.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lakshmanan, I., Marimuthu, S., Chaudhary, S. et al. Muc16 depletion diminishes KRAS-induced tumorigenesis and metastasis by altering tumor microenvironment factors in pancreatic ductal adenocarcinoma. Oncogene 41, 5147–5159 (2022). https://doi.org/10.1038/s41388-022-02493-6

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-022-02493-6

This article is cited by

Search

Quick links