Next Article in Journal
Genome-Wide DNA Methylation and LncRNA-Associated DNA Methylation in Metformin-Treated and -Untreated Diabetes
Next Article in Special Issue
Cell-Free DNA Methylation as Blood-Based Biomarkers for Pancreatic Adenocarcinoma—A Literature Update
Previous Article in Journal
Functions of Polycomb Proteins on Active Targets
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

New Aspects of the Epigenetics of Pancreatic Carcinogenesis

by
Murat Toruner
1,2,
Martin E. Fernandez-Zapico
1 and
Christopher L. Pin
3,*
1
Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
2
Department of Gastroenterology, Ankara University School of Medicine, School of Medicine, 06100 Ankara, Turkey
3
Departments of Paediatrics, Oncology, and Physiology and Pharmacology, University of Western Ontario, Children’s Health Research Institute, London, ON N6C 2V5, Canada
*
Author to whom correspondence should be addressed.
Submission received: 20 August 2020 / Accepted: 25 August 2020 / Published: 31 August 2020
(This article belongs to the Special Issue Epigenetics of Pancreatic Cancer 2.0)

Abstract

:
Pancreatic cancer remains among the deadliest forms of cancer with a 5 year survival rate less than 10%. With increasing numbers being observed, there is an urgent need to elucidate the pathogenesis of pancreatic cancer. While both contribute to disease progression, neither genetic nor environmental factors completely explain susceptibility or pathogenesis. Defining the links between genetic and environmental events represents an opportunity to understand the pathogenesis of pancreatic cancer. Epigenetics, the study of mitotically heritable changes in genome function without a change in nucleotide sequence, is an emerging field of research in pancreatic cancer. The main epigenetic mechanisms include DNA methylation, histone modifications and RNA interference, all of which are altered by changes to the environment. Epigenetic mechanisms are being investigated to clarify the underlying pathogenesis of pancreatic cancer including an increasing number of studies examining the role as possible diagnostic and prognostic biomarkers. These mechanisms also provide targets for promising new therapeutic approaches for this devastating malignancy.

Pancreatic cancer is among the most aggressive gastrointestinal cancers, difficult to treat either by surgical intervention or conventional chemotherapy. Therefore, there is clearly a need to clarify the underlying mechanisms and to develop novel therapies based on these mechanisms. In 2000, a model for pancreatic cancer was presented based solely on genetic alterations [1]. In this model, pancreatic cancer arises from multiple genetic alterations, starting with mutations that promote the constitutive activation of KRAS (Kirsten Rat Sarcoma). However, it is now clear that mechanisms based purely on genetic mutations do not reveal the whole story. For example, pancreatic cancers with identical genetic alterations do not respond in a similar way to chemotherapy or show similar disease course.
Following the complete sequencing of the human genome in 2001 [2,3], our understanding of disease pathogenesis significantly improved. Pathogenesis for most neoplastic or non-neoplastic disorders cannot be solely explained by genetics or environmental factors. They both contribute to susceptibility, initiation and progression of most diseases. In pancreatic cancer, a significant gap in understanding the underlying processes regulating disease pathogenesis still exists. In this regard, epigenetic mechanisms have emerged as key mechanisms that could address this gap. The term epigenetics was originally described in the 1950s by Conrad Waddington to delineate changes in phenotype with no change in genotype [4,5]. In the current view, epigenetics is the study of mitotically heritable changes in the genome function without a change in the nucleotide sequence [6]. Recently, epigenetic mechanisms have emerged as possible explanations for the differential individual behavior of pancreatic cancers. This Special Issue focuses on the role of epigenetics in pancreatic cancer which is projected to be the second cause of cancer death by 2030.
The main epigenetic mechanisms can be grouped into three main categories—DNA methylation, modifications to core histone proteins comprising the nucleosome and RNA interference. By definition, epigenetic modifications are heritable and reversible through cell division [7]. In development, epigenetic modifications promote multipotent cells to differentiate into more defined cell types, with new phenotypes transferred to daughter cells [8]. DNA methylation is established by DNA methyltransferases (DNMTs), which include the maintenance DNMT1 and the de novo methyltransferases DNMT3a/b and DNMT3L [9]. Conversely, DNA demethylation can occur through both passive demethylation through inhibition of methylation and active enzymatic-based demethylation.
Histone modifications occur mainly in the tail regions of the histones, and include acetylation, methylation, phosphorylation, and ubiquitination amongst other modifications. Post-transcriptional modifications to histone structures are usually kept in balance [10]. When the balance is distorted in either way, it leads to gene overexpression or gene repression. Finally, RNA interference refers to noncoding RNAs such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), which modulate gene expression in a sequence specific fashion [11,12].
In this issue, recent advances in epigenetic mechanisms that control pancreatic carcinogenesis and the potential for targeting these mechanisms for diagnostic or therapeutic purposes will be discussed. This issue also includes reviews on the role of the nuclear structure and nutriepigenomics in pancreatic cancer. In each article, authors provide insight regarding potential therapeutic opportunities in targeting epigenetic mechanisms.
There are two main precursor lesions resulting in pancreatic ductal adenocarcinoma (PDAC) formation—pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasm (IPMN). From a genetic mutation perspective, more than 90% of pancreatic cancer patients have gain of function KRAS mutations, and tumor suppressor genes such as SMAD4 and p53, are frequently inactivated. However, no clear correlation has been established between the genetic subtypes of pancreatic cancer and these lesion types, nor the transcriptional subtypes more recently identified including squamous, pancreatic progenitor, exocrine-like (ADEX) and immunogenic PDACs. This lack of correlation highlights possible underlying epigenetic mechanisms. Hank and Liss review these basic epigenetics mechanisms in pancreatic cancer. They summarize basic epigenetic mechanisms that pertain to pancreatic cancer, including histone acetylation and deacetylation models, histone methylation and bromodomain and extraterminal (BET) regulation. Recent advances in chromatin regulation and the potential role of such regulation in PDAC pathogenesis are highlighted.
Cellular function is affected by chromatin control through several mechanisms, including structural mechanisms. Disruption of the nuclear structure can have significant consequences that contribute to oncogenesis. Preston and Faustino describe how the nuclear envelope plays a key role in controlling chromatin processes that are crucial for regulating PDAC pathogenesis. They spotlight three specific components of the nuclear envelope including the LINC (linker of nucleoskeleton and cytoskeleton) complex, which structurally couples cytoskeleton to chromatin and facilitates gene regulation, the nuclear lamina, which provides structural integrity to nucleus, and the nuclear pore complex, which mediates bidirectional nucleocytoplasmic transport. In addition, they describe how components of the nuclear pore complex—nucleoporins—are used as diagnostic markers in cancer.
While mortality rates of cancer are generally declining, pancreatic cancer is a notable exception, likely due to late diagnosis and resistance to conventional treatments. Epigenetic mechanisms are likely to contribute to these factors in pancreatic cancer. Hamdan and Johnsen review epigenetic targets as potential treatment options for pancreatic cancer. They highlight inhibitors for BET proteins and histone deacetylases (HDAC) that exert direct or synergistic effects with other chemotherapeutic agents. They emphasize how mediating their effects on distal regulatory elements, BET and HDAC inhibitors may be effective in PDAC treatment.
In our current understanding of PDAC progression, there are two proposed models. The first involves clonal evolution and suggests tumors are biologically homogenous, derived from the same clone. The second model includes stem cells in which heterogeneity exists across tumor cells providing a defined hierarchical structure within the tumor. Recent studies suggest both models contribute to tumor heterogeneity. Zagorac, Garcia-Bermejo and Sainz describe the epigenetic landscape of cancer stem cells. They provide a summary of the effects that DNA methylation, histone modifications and miRNAs have on cancer stem cells. Moreover, they outline possible epigenetic therapies that target these mechanisms in cancer stem cells.
There is a significant body of research on the association between nutrition and pancreatic cancer. The study of nutrients and their effects on human health through epigenetic mechanisms is termed Nutriepigenomics. However, how specific diets increase or decrease cancer risk is still unclear, as well as the effects that diet has on underlying epigenetic mechanisms. Specific to PDAC, epidemiological studies show folate levels, fruit/vegetable consumption, red meat consumption and saturated fat can alter pancreatic carcinogenesis. Epigenetic mechanisms linked to diet include folate changes that induce donation of methyl groups, and diallyl disulfide and sulforaphone inhibition of histone deacetylase [13,14], which stimulate apoptosis and inhibit cell proliferation.
Understanding epigenetic dysregulation mechanisms will contribute to our efforts in early diagnostic biomarker development, define predictive and prognostic biomarkers and identify new therapeutic approaches in pancreatic cancer. Thompson and Bednar review the clinical aspects of epigenetic changes in PDAC. They describe how genome wide assessment of changes in methylation status gave rise to next generation diagnostic assays. Cohen et al. describe a parallel approach for the testing of protein markers and circulating free DNA (cfDNA) with high specificity [15]. Epigenetic changes, including ARID1A, MLL1, MLL2, and MLL3 mutations or altered DNA methyltransferase 1 expression, alter survival in patients with pancreatic cancer [16,17]. Thompson and Bednar also describe the relationship between epigenetic mechanisms and pancreatic cancer diagnosis, prognosis and survival.
In total, this issue makes a strong case for future studies and a better understanding of the epigenetic mechanisms that regulate pancreatic cancer initiation and progression. Through continued work in these areas, better prognostic and diagnostic markers will be identified, patient-specific treatment options will be supported and novel combinations of epigenetic and nonepigenetic drugs will be investigated.

Author Contributions

All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Canadian Institutes of Health Research (MOP#PJT166029), the Cancer Research Society of Canada and the Rob Lutterman Foundation for Pancreatic Cancer Research to C.L.P. National Cancer Institute (CA136526) and Mayo Clinic Cancer Center to M.E.F.-Z.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hruban, R.H.; Goggins, M.; Parsons, J.; Kern, S.E. Progression model for pancreatic cancer. Clin. Cancer Res. 2000, 6, 2969–2972. [Google Scholar] [PubMed]
  2. Lander, E.S.; Linton, L.M.; Birren, B.; Nusbaum, C.; Zody, M.C.; Baldwin, J.; Devon, K.; Dewar, K.; Doyle, M.; FitzHugh, W.; et al. Initial sequencing and analysis of the human genome. Nature 2001, 409, 860–921. [Google Scholar] [PubMed] [Green Version]
  3. McPherson, J.D.; Marra, M.; Hillier, L.D.; Waterston, R.H.; Chinwalla, A.; Wallis, J.; Sekhon, M.; Wylie, K.; Mardis, E.R.; Wilson, R.K.; et al. A physical map of the human genome. Nature 2001, 409, 934–941. [Google Scholar] [PubMed] [Green Version]
  4. Waddington, C.H. Canalization of development and genetic assimilation of acquired characters. Nature 1959, 183, 1654–1655. [Google Scholar] [CrossRef] [PubMed]
  5. Waddington, C.H. The epigenotype 1942. Int. J. Epidemiol. 2012, 41, 10–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Bird, A. Perceptions of epigenetics. Nature 2007, 447, 396–398. [Google Scholar] [CrossRef] [PubMed]
  7. Cooney, R.; Baker, J.; Brain, O.; Danis, B.; Pichulik, T.; Allan, P.; Ferguson, D.J.; Campbell, B.J.; Jewell, D.; Simmons, A. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nat. Med. 2010, 16, 90–97. [Google Scholar] [CrossRef] [PubMed]
  8. Brooks, W.H.; Le Dantec, C.; Pers, J.O.; Youinou, P.; Renaudineau, Y. Epigenetics and autoimmunity. J. Autoimmun. 2010, 34, J207–J219. [Google Scholar] [CrossRef] [PubMed]
  9. Reik, W.; Dean, W.; Walter, J. Epigenetic reprogramming in mammalian development. Science 2001, 293, 1089–1093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Kouzarides, T. Chromatin modifications and their function. Cell 2007, 128, 693–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef] [PubMed]
  12. He, L.; Hannon, G.J. MicroRNAs: Small RNAs with a big role in gene regulation. Nat. Rev. Genet. 2004, 5, 522–531. [Google Scholar] [CrossRef]
  13. Davis, C.D.; Ross, S.A. Dietary components impact histone modifications and cancer risk. Nutr. Rev. 2007, 65, 88–94. [Google Scholar] [CrossRef]
  14. Garfinkel, M.D.; Ruden, D.M. Chromatin effects in nutrition, cancer, and obesity. Nutrition 2004, 20, 56–62. [Google Scholar] [CrossRef] [PubMed]
  15. Cohen, J.D.; Javed, A.A.; Thoburn, C.; Wong, F.; Tie, J.; Gibbs, P.; Schmidt, C.M.; Yip-Schneider, M.T.; Allen, P.J.; Schattner, M.; et al. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers. Proc. Natl. Acad. Sci. USA 2017, 114, 10202–10207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Sausen, M.; Phallen, J.; Adleff, V.; Jones, S.; Leary, R.J.; Barrett, M.T.; Anagnostou, V.; Parpart-Li, S.; Murphy, D.; Li, Q.K.; et al. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nat. Commun. 2015, 6, 7686. [Google Scholar] [CrossRef] [PubMed]
  17. Wang, W.; Gao, J.; Man, X.H.; Li, Z.S.; Gong, Y.F. Significance of DNA methyltransferase-1 and histone deacetylase-1 in pancreatic cancer. Oncol. Rep. 2009, 21, 1439–1447. [Google Scholar] [PubMed] [Green Version]

Share and Cite

MDPI and ACS Style

Toruner, M.; Fernandez-Zapico, M.E.; Pin, C.L. New Aspects of the Epigenetics of Pancreatic Carcinogenesis. Epigenomes 2020, 4, 18. https://0-doi-org.brum.beds.ac.uk/10.3390/epigenomes4030018

AMA Style

Toruner M, Fernandez-Zapico ME, Pin CL. New Aspects of the Epigenetics of Pancreatic Carcinogenesis. Epigenomes. 2020; 4(3):18. https://0-doi-org.brum.beds.ac.uk/10.3390/epigenomes4030018

Chicago/Turabian Style

Toruner, Murat, Martin E. Fernandez-Zapico, and Christopher L. Pin. 2020. "New Aspects of the Epigenetics of Pancreatic Carcinogenesis" Epigenomes 4, no. 3: 18. https://0-doi-org.brum.beds.ac.uk/10.3390/epigenomes4030018

Article Metrics

Back to TopTop