Download as pdf or txt
Download as pdf or txt
You are on page 1of 2

Cell Metabolism

Letter

Endogenous Hyperinsulinemia Contributes


to Pancreatic Cancer Development
Anni M.Y. Zhang,1 Jamie Magrill,1 Twan J.J. de Winter,1 Xiaoke Hu,1 Søs Skovsø,1 David F. Schaeffer,2 Janel L. Kopp,1,*
and James D. Johnson1,*
1Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
2Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
*Correspondence: janel.kopp@ubc.ca (J.L.K.), james.d.johnson@ubc.ca (J.D.J.)
https://doi.org/10.1016/j.cmet.2019.07.003

The incidence of cancers, including pancre- ing to 1 year of age. HFD causes hyperin- ures S1I and S1J), were found in both
atic ductal adenocarcinoma (PDAC), con- sulinemia and accelerates PanIN forma- groups. Only one mouse from each geno-
tinues to rise alongside the global obesity tion/progression in KrasG12D-expressing type developed PDAC, which expressed
and diabetes epidemics. This rise sug- mice (Chang et al., 2017). We employed the ductal marker Cytokeratin 19 (Figures
gests that factors modulated by environ- controls with a normal complement of S1K and S1M). Remarkably, Ptf1aCreER;
ment or lifestyle play a key role, but the wild-type Ins1 alleles (Ins1+/+) and experi- LSL-KrasG12D;Ins1+/ ;Ins2 / mice had
mechanisms by which obesity and type 2 mental mice with only a single functional approximately half the area covered by
diabetes promote cancer remain unclear. Ins1 allele (Ins1+/ ) from the same parental PanINs or tumor (12.7% ± 3.4%)
Obesity and type 2 diabetes are associ- sets (Figure S1A). All mice were maintained compared with Ptf1aCreER;LSL-KrasG12D;
ated with elevations in circulating insulin on an Ins2 / background to prevent insu- Ins1+/+;Ins2 / control mice (25.4% ±
and glucose, as well as systemic low- lin gene compensation (Mehran et al., 3.8%; p = 0.03) (Figure S1M). Without
grade inflammation, which have been pro- 2012). We previously found that these ge- including tumor area, experimental mice
posed to contribute to cancer initiation or netic manipulations of Ins1 and Ins2 result still had approximately half the area
progression. Primary hyperinsulinemia, in a sustained reduction in fasting insulin covered by PanINs (12.3% ± 3.1%; p =
defined as circulating insulin in excess of modest enough not to chronically impair 0.01) compared to controls (24.7% ±
what is required for glucose homeostasis glucose homeostasis (Mehran et al., 3.3%) (Figure S1N). Mice with reduced in-
and independent of insulin resistance, 2012). On the Ptf1aCreER;LSL-KrasG12D sulin also had significantly fewer high-
can be found in 30% of obese adults background, there was the expected mi- grade PanINs per section (Figures S1L
(Tricò et al., 2018). Hyperinsulinemia is nor reduction in fasting insulin and body and S1O) or per whole pancreas area.
associated with PDAC, independent of weight in mice with reduced Ins1 There was no significant difference be-
BMI (Tsujimoto et al., 2017). Nevertheless, compared to controls (Figures S1C–S1F). tween the genotypes when the number of
the cause and effect relationship between Importantly, this reduction in insulin high-grade PanINs was normalized to the
hyperinsulinemia and cancer remains to in Ptf1aCreER;LSL-KrasG12D;Ins1+/ ;Ins2 / total PanIN area. At 1 year of age, PanIN
be determined directly. mice did not affect fasting glucose in fe- plus tumor area correlated with fasting in-
Caloric restriction, fasting, and low-car- males over 1 year of age (Figure S1G). In sulin, but not glucose levels (Figures
bohydrate diets have been proposed for male mice from the same litters, we S1P–S1R), arguing against a prominent
prevention or as an adjunct to cancer ther- observed impaired fasting glucose, and in role for glycemia in PanIN formation in
apy based in part on what has come to be some cases frank diabetes (Figure S1H), our model. Although we were unable to
known as the insulin-cancer hypothesis. consistent with known sex differences follow them for 1 year, male mice with
Insulin has mitogenic effects on human in insulin sensitivity. Therefore, comparing reduced Ins1 had less PanINs (Figures
pancreatic cells via signaling implicated female Ptf1aCreER;LSL-KrasG12D;Ins1+/ ; S1S–S1U). Together, our data implicate
in tumorigenesis (Chan et al., 2014), but Ins2 / mice to female Ptf1aCreER;LSL- endogenous increases in insulin, but
there is no direct evidence that hyperinsu- KrasG12D;Ins1+/+;Ins2 / controls offered not glucose, in HFD-mediated promotion
linemia in vivo can affect the development us the unique opportunity to test the role of PanIN development and potentially
of any cancer. of insulin in pancreatic cancer initiation, pancreatic cancer.
KRAS mutations are found in 90% of in the absence of changes in fasting We investigated possible underlying
human PDAC and reliably induce preneo- glucose. mechanisms associated with insulin-
plastic pancreatic intraepithelial neoplasia To test our primary hypothesis that driven PanINs formation, starting with the
(PanIN) precursor lesions when expressed decreasing endogenous insulin production mitogenic effects of insulin (Chan et al.,
in murine pancreatic acinar cells using an would affect the initiation of KrasG12D- 2014). We were unable to measure statisti-
inducible Ptf1aCreER allele (Basturk et al., driven PDAC, we measured the percent cally significant differences in proliferation
2015; Kopp et al., 2012). To test the of total pancreatic area occupied by rate in any cell types, although variation
hypothesis that a diet-induced increase in PanINs and tumor. At 57 weeks of age, was high between mice within groups (Fig-
circulating insulin plays a causal role in abundant ductal lesions with histologic ures S1V and S1W). Our data cannot
PDAC initiation, Ptf1aCreER;LSL-KrasG12D and molecular characteristics of low-grade exclude the possibility that insulin provided
mice with reduced insulin gene dosage PanINs, including highly acidic mucin con- mitogenic and anti-apoptotic signals to
were fed a high-fat diet (HFD) from wean- tent indicated by Alcian blue staining (Fig- cells expressing oncogenic KrasG12D prior

Cell Metabolism 30, September 3, 2019 ª 2019 Elsevier Inc. 403


Cell Metabolism

Letter

to the time points we studied. PDAC is by a grant from the Cancer Research Society Dawson, D.W., Hines, O.J., et al. (2017).
(Canada), an Ideas Grant from the Pancreas Incidence of pancreatic cancer is dramatically
characterized by an intense desmoplastic increased by a high fat, high calorie diet in
Centre BC (Canada), and an Innovation Grant (IN-
reaction, and the increased inflammation NOV16-1) from the Canadian Cancer Society KrasG12D mice. PLoS One 12, e0184455.
associated with PanIN formation could Research Institute (Canada) to J.D.J. and J.L.K.
A.M.Y.Z. was supported by Canada Graduate Kopp, J.L., von Figura, G., Mayes, E., Liu, F.F.,
drive the desmoplasia. Studies have sug- Dubois, C.L., Morris, J.P., 4th, Pan, F.C.,
Scholarships – Master’s program, a Four-Year
gested that resulting cancer-associated Fellowship from the University of British
Akiyama, H., Wright, C.V., Jensen, K., et al.
(2012). Identification of Sox9-dependent acinar-
stroma could support tumor survival and Columbia, and Frederick Banting and Charles to-ductal reprogramming as the principal mecha-
growth (Sousa et al., 2016). Insulin has Best Canada Graduate Scholarships. nism for initiation of pancreatic ductal adenocarci-
also been shown to enhance the prolifera- noma. Cancer Cell 22, 737–750.
AUTHOR CONTRIBUTIONS
tion of pancreatic stellate cells and aug- Lee, A.Y.L., Dubois, C.L., Sarai, K., Zarei, S.,
ments their production of extracellular A.M.Y.Z. designed and managed the study; ac-
Schaeffer, D.F., Sander, M., and Kopp, J.L.
matrix proteins through PI3K (Yang et al., (2018). Cell of origin affects tumour development
quired, analyzed, and interpreted all data unless
and phenotype in pancreatic ductal adenocarci-
2016). By staining the collagen with Sirius otherwise noted; and wrote the manuscript. J.M. noma. Gut. Published online January 23, 2018.
acquired, analyzed, and interpreted data (CK19, https://doi.org/10.1136/gutjnl-2017-314426.
Red, we found that mice with reduced in-
Ki67, and Alcian blue staining). T.J.J.d.W. ac-
sulin had significantly less collagen depo- quired, analyzed, and interpreted data (male PanIN Mehran, A.E., Templeman, N.M., Brigidi, G.S., Lim,
sition than controls (20.1% ± 4.3% and staining). X.H. managed the mouse colony and G.E., Chu, K.Y., Hu, X., Botezelli, J.D., Asadi, A.,
41.3% ± 5.1%, respectively; p = 0.007) performed mouse husbandry. S.S. provided expert Hoffman, B.G., Kieffer, T.J., et al. (2012).
advice on breeding and study design. D.F.S. pro- Hyperinsulinemia drives diet-induced obesity inde-
(Figures S1X and S1Y). This result is vided expert advice on pancreatic cancer pathol- pendently of brain insulin production. Cell Metab.
consistent with the idea that hyperinsuline- ogy and study design. J.L.K. designed, analyzed, 16, 723–737.
mia promotes PanIN development in part and interpreted experiments; edited the manu-
script; obtained funding; and supervised the study. Pan, F.C., Bankaitis, E.D., Boyer, D., Xu, X., Van de
by contributing to the fibrogenesis associ- J.D.J. analyzed and interpreted data, edited the Casteele, M., Magnuson, M.A., Heimberg, H., and
ated with PanINs, or that increased fibrosis Wright, C.V. (2013). Spatiotemporal patterns of
manuscript, obtained funding, supervised the
multipotentiality in Ptf1a-expressing cells during
is caused by the greater number of study, and conceived the study concept and
pancreas organogenesis and injury-induced facul-
design. tative restoration. Development 140, 751–764.
PanINs.
Our study is the first to separate the
SUPPORTING CITATIONS Sousa, C.M., Biancur, D.E., Wang, X., Halbrook,
causal role of hyperinsulinemia from hy- C.J., Sherman, M.H., Zhang, L., Kremer, D.,
perglycemia and directly test the insulin- The following references appear in the Supple- Hwang, R.F., Witkiewicz, A.K., Ying, H., et al.
(2016). Pancreatic stellate cells support tumour
cancer hypothesis in vivo. Limitations of mental Information: Alejandro et al. (2011); Basturk
metabolism through autophagic alanine secretion.
et al. (2015); Chan et al. (2014); Chang et al. (2017);
our study include the analysis of PanIN Nature 536, 479–483.
Kopp et al. (2012); Lee et al. (2018); Mehran et al.
at a single time point. Future studies (2012); Pan et al. (2013); Sousa et al. (2016); Templeman, N.M., Flibotte, S., Chik, J.H.L., Sinha,
should provide a more detailed time Templeman et al. (2017); Tricò et al. (2018); Tsuji- S., Lim, G.E., Foster, L.J., Nislow, C., and Johnson,
course of the early initiation and address moto et al. (2017); Tuveson et al. (2004); Yang J.D. (2017). Reduced circulating insulin enhances
et al. (2016). insulin sensitivity in old mice and extends lifespan.
the role of insulin in progression to Cell Rep. 20, 451–463.
PDAC. Notwithstanding, our findings are
REFERENCES Tricò, D., Natali, A., Arslanian, S., Mari, A., and
consistent with our recent study demon-
Ferrannini, E. (2018). Identification, pathophysi-
strating that a reduction of fasting insulin, Alejandro, E.U., Lim, G.E., Mehran, A.E., Hu, X., ology, and clinical implications of primary insulin
so mild that it did not significantly increase Taghizadeh, F., Pelipeychenko, D., Baccarini, M., hypersecretion in nondiabetic adults and adoles-
and Johnson, J.D. (2011). Pancreatic b-cell Raf-1 cents. JCI Insight 3, https://doi.org/10.1172/jci.
fasting glucose, was sufficient to signifi- insight.124912.
is required for glucose tolerance, insulin secre-
cantly extend mean and maximum life- tion, and insulin 2 transcription. FASEB J. 25,
span of mice (Templeman et al., 2017). 3884–3895. Tsujimoto, T., Kajio, H., and Sugiyama, T. (2017).
Association between hyperinsulinemia and
Insulin-lowering interventions such as ex- Basturk, O., Hong, S.M., Wood, L.D., Adsay, N.V., increased risk of cancer death in nonobese and
ercise, diet, and metformin warrant further Albores-Saavedra, J., Biankin, A.V., Brosens, L.A., obese people: a population-based observational
investigation as strategies to prevent Fukushima, N., Goggins, M., Hruban, R.H., et al.; study. Int. J. Cancer 141, 102–111.
Baltimore Consensus Meeting (2015). A revised
cancer or limit its progression. classification system and recommendations from Tuveson, D.A., Shaw, A.T., Willis, N.A., Silver, D.P.,
the Baltimore Consensus Meeting for Neoplastic Jackson, E.L., Chang, S., Mercer, K.L., Grochow,
Precursor Lesions in the Pancreas. Am. J. Surg. R., Hock, H., Crowley, D., et al. (2004).
SUPPLEMENTAL INFORMATION
Pathol. 39, 1730–1741. Endogenous oncogenic K-ras(G12D) stimulates
proliferation and widespread neoplastic and devel-
Supplemental Information can be found online at Chan, M.T., Lim, G.E., Skovsø, S., Yang, Y.H., opmental defects. Cancer Cell 5, 375–387.
https://doi.org/10.1016/j.cmet.2019.07.003. Albrecht, T., Alejandro, E.U., Hoesli, C.A., Piret,
J.M., Warnock, G.L., and Johnson, J.D. (2014). Yang, J., Waldron, R.T., Su, H.Y., Moro, A., Chang,
ACKNOWLEDGMENTS Effects of insulin on human pancreatic cancer pro- H.H., Eibl, G., Ferreri, K., Kandeel, F.R., Lugea, A.,
gression modeled in vitro. BMC Cancer 14, 814. Li, L., and Pandol, S.J. (2016). Insulin promotes
proliferation and fibrosing responses in activated
The authors thank members of the Johnson and Chang, H.H., Moro, A., Takakura, K., Su, H.Y., Mo, pancreatic stellate cells. Am. J. Physiol.
Kopp labs for discussions. This study was funded A., Nakanishi, M., Waldron, R.T., French, S.W., Gastrointest. Liver Physiol. 311, G675–G687.

404 Cell Metabolism 30, September 3, 2019

You might also like