LKB1 haploinsufficiency cooperates with Kras to promote pancreatic cancer through suppression of p21-dependent growth arrest - PubMed (original) (raw)

. 2010 Aug;139(2):586-97, 597.e1-6.

doi: 10.1053/j.gastro.2010.04.055. Epub 2010 May 6.

Nigel B Jamieson, Saadia A Karim, Dimitris Athineos, Rachel A Ridgway, Colin Nixon, Colin J McKay, Ross Carter, Valerie G Brunton, Margaret C Frame, Alan Ashworth, Karin A Oien, T R Jeffry Evans, Owen J Sansom

Affiliations

LKB1 haploinsufficiency cooperates with Kras to promote pancreatic cancer through suppression of p21-dependent growth arrest

Jennifer P Morton et al. Gastroenterology. 2010 Aug.

Erratum in

Abstract

Background & aims: Patients carrying germline mutations of LKB1 have an increased risk of pancreatic cancer; however, it is unclear whether down-regulation of LKB1 is an important event in sporadic pancreatic cancer. In this study, we aimed to investigate the impact of LKB1 down-regulation for pancreatic cancer in mouse and human and to elucidate the mechanism by which Lkb1 deregulation contributes to this disease.

Methods: We first investigated the consequences of Lkb1 deficiency in a genetically modified mouse model of pancreatic cancer, both in terms of disease progression and at the molecular level. To test the relevance of our findings to human pancreatic cancer, we investigated levels of LKB1 and its potential targets in human pancreatic cancer.

Results: We definitively show that Lkb1 haploinsufficiency can cooperate with oncogenic KrasG12D to cause pancreatic ductal adenocarcinoma (PDAC) in the mouse. Mechanistically, this was associated with decreased p53/p21-dependent growth arrest. Haploinsufficiency for p21 (Cdkn1a) also synergizes with KrasG12D to drive PDAC in the mouse. We also found that levels of LKB1 expression were decreased in around 20% of human PDAC and significantly correlated with low levels of p21 and a poor prognosis. Remarkably, all tumors that had low levels of LKB1 had low levels of p21, and these tumors did not express mutant p53.

Conclusions: We have identified a novel LKB1-p21 axis that suppresses PDAC following Kras mutation in vivo. Down-regulation of LKB1 may therefore serve as an alternative to p53 mutation to drive pancreatic cancer in vivo.

Copyright (c) 2010 AGA Institute. Published by Elsevier Inc. All rights reserved.

PubMed Disclaimer

Figures

Supplementary Figure 1

Supplementary Figure 1

Western immunoblotting analysis of Lkb1 (top panel) and phospho−adenosine monophosphate−activated protein kinase (AMPK) (bottom panel) protein levels in pancreatic tumor lysates from KC, KLC, and LLC mice. Levels were normalized against β-tubulin levels.

Supplementary Figure 2

Supplementary Figure 2

Lkb1 deficiency accelerates KrasG12D-mediated pancreatic tumorigenesis through down-regulation of p21. (A) Immunohistochemical staining for p53, p21, and Ki67 in pancreatic intraepithelial neoplasia (PanIN) lesions in Pdx1-Cre Kras G12D/+ (KC) mice. (B) Immunohistochemical staining for p53, p21, and Ki67 in PanIN lesions in Pdx1-Cre Kras G12D/+ Lkb1 flox/+ (KLC) mice.

Supplementary Figure 3

Supplementary Figure 3

p21 haploinsufficiency synergizes with KrasG12D to induce pancreatic cancer and a dwarfism phenotype. (A) In vivo imaging of green fluorescent protein (GFP) fluorescence within the pancreas of a Pdx1-Cre Kras G12D/+ Cdkn1a +/− (KCp21) mouse at 6 weeks old; p21 deficiency allows outgrowth of recombined, and therefore, KrasG12D-bearing cells. (B) Loss of senescence-associated β-galactosidase staining in pancreatic intraepithelial neoplasia (PanIN) lesions from KCp21 mice, compared with that seen in PanIN lesions arising in Pdx1-Cre Kras G12D/+ (KC) mice. (C) Photograph demonstrating dwarfism phenotype in 6-week-old KCp21 mouse compared with a Pdx1-Cre CdKn1a/littermate control. (D) Representative H&E-stained sections showing reduced islet size in the pancreata of KCp21 mice, compared with Pdx1-Cre Cdkn1a +/− control mice. (E) Boxplot showing number of islets per histopathological section of pancreas from KCp21 and KC mice as indicated (P = .00008). (F) Boxplot showing quantification of islet volume per histopathological section of pancreas from KCp21 and KC mice as indicated (P = .007).

Supplementary Figure 4

Supplementary Figure 4

(A) Kaplan–Meier survival curve showing tumor-free survival of littermate Pdx1-Cre Kras G12D/+ Lkb1 fl/+ mice (KLC, broken blue line) and Pdx1-Cre Kras G12D/+ Lkb1 fl/+ Trp53 R172H/+ mice (KLPC, solid red line). (B) Kaplan–Meier survival curve showing tumor-free survival of littermate Pdx1-Cre Kras G12D/+ Lkb1 fl/+ mice (KLC, broken blue line) and Pdx1-Cre Kras G12D/+ Lkb1 fl/+ p21 +/− mice (KLCp21, solid red line).

Supplementary Figure 5

Supplementary Figure 5

p21 expression in human pancreatic ductal adenocarcinoma. (A) p21 immunostaining of normal pancreatic ductal tissue on human tissue microarray (TMA). (B–E) Representative immunostaining of p21 in pancreatic ductal adenocarcinoma on human TMA: (B) absent, (C) weak, (D) moderate, and (E) strong staining.

Figure 1

Figure 1

Lkb1 heterozygosity combined with KrasG12D is sufficient to induce pancreatic cancer. (A) Kaplan–Meier survival curve showing tumor-free survival of Pdx1-Cre Lkb1 flox/+ (LC, green dashed line), Pdx1-Cre Kras G12D/+ (KC, red solid line), and Pdx1-Cre Kras G12D/+ Lkb1 flox/+ (KLC, blue dashed line) mice. (B) Boxplot showing number of pancreatic intraepithelial neoplasia (PanINs) per histopathological section of pancreas from wild-type, KC, and KLC mice (n = 6 mice per genotype) as indicated (P = .007). (C) Boxplot showing number of PanINs of grades 1−3 per histopathological section of pancreas from wild-type, KC, and KLC mice (n = 6 mice per genotype) as indicated. (D) H&E-stained sections of a normal duct, a PanIN lesion, and an Alcian blue−stained section of a PanIN lesion arising in the pancreas of a 6-week-old KLC mouse, as indicated. (E) H&E-stained sections of pancreatic ductal adenocarcinoma (PDAC) arising in KLC mice, with some tumors exhibiting a cystic component and others exhibiting increased lymphocytic involvement.

Figure 2

Figure 2

Homozygous loss of Lkb1 is sufficient to initiate pancreatic tumorigenesis. (A) Kaplan–Meier survival curve showing tumor-free survival of Pdx1-Cre Lkb1 fl/+ mice (LC, green dashed line), Pdx1-Cre Lkb1 fl/fl mice (LLC, blue dashed line), Pdx1-Cre Kras G12D/+ mice (KC, red solid line), and Pdx1-Cre Kras G12D/+ Lkb1 fl/+ mice (KLC, blue solid line). (B) Gross pathology of a cystic pancreatic tumor arising in an LLC mouse. (C, D) H&E-stained sections of cystic pancreatic tumors arising in LLC mice. (E) Alcian blue−stained section of a cystic pancreatic tumor arising in an LLC mouse.

Figure 3

Figure 3

Lkb1 haploinsufficiency synergizes with KrasG12D to induce pancreatic cancer. (A) Immunohistochemical analysis of Lkb1 levels in normal duct, pancreatic intraepithelial neoplasia (PanIN), and pancreatic ductal adenocarcinoma (PDAC) arising in Pdx1-Cre Kras G12D/+ (KC) mice, as indicated. (B) Immunohistochemical analysis of Lkb1 levels in normal duct, PanIN, and PDAC arising in Pdx1-Cre Kras G12D/+ Lkb1 flox/+ (KLC) mice. (C) Immunohistochemical analysis of Lkb1 levels in cystadenoma arising in a Pdx1-Cre Lkb1 flox/flox (LLC) mouse. (D) Detection of the Lkb1 transcript by reverse-transcription polymerase chain reaction in tissue microdissected from lesions in frozen sections harvested from KC, KLC, and LLC mice. β-actin serves as control for RNA quantity and integrity. (E) Quantification of Western immunoblotting analysis of Lkb1 (top panel) and phospho−adenosine monophosphate−activated protein kinase (AMPK; bottom panel) protein levels in pancreatic tumor lysates from KC, KLC, and LLC mice. Levels were normalized against β-tubulin levels.

Figure 4

Figure 4

Lkb1 deficiency accelerates KrasG12D-mediated pancreatic tumorigenesis through down-regulation of p21. (A) Immunohistochemical staining for p53, p21, and Ki67, and senescence-associated β-gal staining in pancreatic intraepithelial neoplasia (PanIN) lesions in Pdx1-Cre Kras G12D/+ (KC) mice. (B) Immunohistochemical staining for p53, p21, and Ki67, and senescence-associated β-gal staining in PanIN lesions in Pdx1-Cre Kras G12D/+ Lkb1 flox/+ (KLC) mice (Supplementary Figure 2 for high-magnification images). (C) Boxplot showing quantification of p21 staining in PanINs in KC mice compared with KLC mice (n = 6) as indicated (P < .002). (D) Boxplot showing quantification of p53 staining in PanINs in KC mice compared with KLC mice (n = 6) as indicated (P < .004).

Figure 5

Figure 5

(A) Immunohistochemical staining for MCM2, p16, IgfBP7, and p19ARF in pancreatic intraepithelial neoplasia (PanIN) lesions in Pdx1-Cre Kras G12D/+ (KC) mice. (B) Immunohistochemical staining for MCM2, p16, IgfBP7, and p19ARF in PanIN lesions in Pdx1-Cre Kras G12D/+ Lkb1 flox/+ (KLC) mice.

Figure 6

Figure 6

(A) Boxplot showing number of pancreatic intraepithelial neoplasias (PanINs) per histopathological section of pancreas from wild-type, KC, KLC, and KCp21 mice (n = 6) as indicated. (B) Kaplan–Meier survival curve showing disease-free survival of Pdx1-Cre p21 +/− mice (p21, broken red line, n = 13), and Pdx1-Cre Kras G12D/+ p21 +/− mice (KCp21, solid blue line, n = 13). (C) H&E-stained section of a pancreatic ductal adenocarcinoma (PDAC) from a KCp21 mouse. (D) Immunohistochemical staining for p53 in PDAC and PanIN arising in KCp21 mice. (E) Senescence-associated β-gal staining in PanIN lesions from KC and KCp21 mice. (F) Immunohistochemical staining for the senescence markers p16, IgfBP7, and MCM2 in PanIN lesions from KCp21 mice.

Figure 7

Figure 7

Decreased Lkb1 expression in human pancreatic ductal adenocarcinoma (PDAC) correlates with low p21 expression and reduced survival. (A) Lkb1 immunostaining of duct and PDAC on human tissue microarray (TMA). (B) Low-grade tumors (n = 81) exhibited a higher level of Lkb1 expression (median histoscore, 128) vs high-grade tumors (n = 33) (median histoscore, 100) (P = .01). Stage T2 tumors (n = 13) had a higher level of Lkb1 expression (median histoscore, 150) vs stage T3 tumors (n = 111) (median histoscore, 105) (P = .02). (C) Correlation of Lkb1 protein with p21 protein expression in 106 cases of PDAC (Spearman's rho correlation coefficient 0.34; P < .001). (D) Kaplan–Meier analyses showing cases with low Lkb1 expression (n = 20) have poorer outcomes compared to those with high expression (n = 86; P = .006), and that cases with p21 low expression (n = 78) have poorer outcomes compared to those with high expression (n = 28; P = .035). (E) Kaplan–Meier analysis illustrates that Lkb1hi/p21hi patients have a more favorable outcome compared to Lkb1hi/p21lo and Lkb1lo /p21lo cases. (F) Boxplot of p53 histoscore in Lkb1lo/p21lo tumors (blue bar, n = 20) compared with Lkb1hi/p21lo tumors (red bar, n = 58) (P = .05).

References

    1. Jemal A., Siegel R., Ward E. Cancer statistics, 2007. CA Cancer J Clin. 2007;57:43–66. - PubMed
    1. Sener S.F., Fremgen A., Menck H.R., Winchester D.P. Pancreatic cancer: a report of treatment and survival trends for 100,313 patients diagnosed from 1985–1995, using the National Cancer Database. J Am Coll Surg. 1999;189:1–7. - PubMed
    1. Hruban R.H., Goggins M., Parsons J., Kern S.E. Progression model for pancreatic cancer. Clin Cancer Res. 2000;6:2969–2972. - PubMed
    1. Almoguera C., Shibata D., Forrester K. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell. 1988;53:549–554. - PubMed
    1. Hemminki A., Markie D., Tomlinson I. A serine/threonine kinase gene defective in Peutz-Jeghers syndrome. Nature. 1998;391:184–187. - PubMed

Publication types

MeSH terms

Substances

Grants and funding

LinkOut - more resources