Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2015

Open Access 01-12-2015 | Research article

Extrinsic intestinal denervation modulates tumor development in the small intestine of ApcMin/+ mice

Authors: Verena Liu, Alexandra Dietrich, Michael S Kasparek, Petra Benhaqi, Marlon R Schneider, Michael Schemann, Hendrik Seeliger, Martin E Kreis

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2015

Login to get access

Abstract

Background

Innervation interacts with enteric immune responses. Chronic intestinal inflammation is associated with increased risk of colorectal cancer. We aimed to study potential extrinsic neuronal modulation of intestinal tumor development in a mouse model.

Methods

Experiments were performed with male ApcMin/+ or wild type mice (4 weeks old, body weight approximately 20 g). Subgroups with subdiaphragmatic vagotomy (apcV/wtV), sympathetic denervation of the small intestine (apcS/wtS) or sham operated controls (apcC/wtC) were investigated (n = 6-14 per group). Three months after surgical manipulation, 10 cm of terminal ileum were excised, fixed for 48 h in 4% paraformaldehyde and all tumors were counted and their area determined in mm2 (mean ± standard error of the mean (SEM)). Whole mounts of the muscularis of terminal ileum and duodenum (internal positive control) were also stained for tyrosine hydroxylase to confirm successful sympathetic denervation.

Results

Tumor count in ApcMin/+ mice was 62 ± 8 (apcC), 46 ± 11 (apcV) and 54 ± 8 (apcS) which was increased compared to wildtype controls with 4 ± 0.5 (wtC), 5 ± 0.5 (wtV) and 5 ± 0.6 (wtS; all p < 0.05). For ApcMin/+ groups, vagotomized animals showed a trend towards decreased tumor counts compared to sham operated ApcMin/+ controls while sympathetic denervation was similar to sham ApcMin/+. Area covered by tumors in ApcMin/+ mice was 55 ± 10 (apcC), 31 ± 8 (apcV) and 42 ± 8 (apcS) mm2, which was generally increased compared to wildtype controls with 7 ± 0.6 (wtC), 7 ± 0.4 (wtV) and 7 ± 0.6 (wtS) mm2 (all p < 0.05). In ApcMin/+ groups, tumor area was decreased in vagotomized animals compared to sham operated controls (p < 0.05) while sympathetically denervated mice showed a minor trend to decreased tumor area compared to controls.

Conclusions

Extrinsic innervation of the small bowel is likely to modulate tumor development in ApcMin/+ mice. Interrupted vagal innervation, but not sympathetic denervation, seems to inhibit tumor growth.
Literature
1.
go back to reference Berthoud HR, Neuhuber WL. Functional and chemical anatomy of the afferent vagal system. Auton Neurosci. 2000;85:1–17.CrossRefPubMed Berthoud HR, Neuhuber WL. Functional and chemical anatomy of the afferent vagal system. Auton Neurosci. 2000;85:1–17.CrossRefPubMed
2.
go back to reference Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405:458–62.CrossRefPubMed Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405:458–62.CrossRefPubMed
3.
go back to reference Sun P, Zhou K, Wang S, Li P, Chen S, Lin G, et al. Involvement of MAPK/NF-κB signaling in the activation of the cholinergic anti-inflammatory pathway in experimental colitis by chronic vagus nerve stimulation. PLoS One. 2013;8:e69424.CrossRefPubMedCentralPubMed Sun P, Zhou K, Wang S, Li P, Chen S, Lin G, et al. Involvement of MAPK/NF-κB signaling in the activation of the cholinergic anti-inflammatory pathway in experimental colitis by chronic vagus nerve stimulation. PLoS One. 2013;8:e69424.CrossRefPubMedCentralPubMed
4.
go back to reference The FO, Boeckxstaens GE, Snoek SA, Cash JL, Bennink R, Larosa GJ, et al. Activation of the cholinergic anti-inflammatory pathway ameliorates postoperative ileus in mice. Gastroenterology. 2007;133:1219–28.CrossRefPubMed The FO, Boeckxstaens GE, Snoek SA, Cash JL, Bennink R, Larosa GJ, et al. Activation of the cholinergic anti-inflammatory pathway ameliorates postoperative ileus in mice. Gastroenterology. 2007;133:1219–28.CrossRefPubMed
5.
go back to reference Momi N, Kaur S, Krishn SR, Batra SK. Discovering the route from inflammation to pancreatic cancer. Minerva Gastroenterol Dietol. 2012;58:283–97.PubMedCentralPubMed Momi N, Kaur S, Krishn SR, Batra SK. Discovering the route from inflammation to pancreatic cancer. Minerva Gastroenterol Dietol. 2012;58:283–97.PubMedCentralPubMed
6.
go back to reference Itzkowitz SH, Yio X. Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol. 2004;287:G7–17.CrossRefPubMed Itzkowitz SH, Yio X. Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol. 2004;287:G7–17.CrossRefPubMed
7.
go back to reference Moss SF, Blaser MJ. Mechanisms of disease: inflammation and the origins of cancer. Nat Clin Pract Oncol. 2005;2:90–7. quiz 1 p following 113.CrossRefPubMed Moss SF, Blaser MJ. Mechanisms of disease: inflammation and the origins of cancer. Nat Clin Pract Oncol. 2005;2:90–7. quiz 1 p following 113.CrossRefPubMed
8.
9.
go back to reference Frucht H, Jensen RT, Dexter D, Yang WL, Xiao Y. Human colon cancer cell proliferation mediated by the M3 muscarinic cholinergic receptor. Clin Cancer Res. 1999;5:2532–9.PubMed Frucht H, Jensen RT, Dexter D, Yang WL, Xiao Y. Human colon cancer cell proliferation mediated by the M3 muscarinic cholinergic receptor. Clin Cancer Res. 1999;5:2532–9.PubMed
10.
go back to reference Moser AR, Pitot HC, Dove WF. A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse. Science. 1990;247:322–4.CrossRefPubMed Moser AR, Pitot HC, Dove WF. A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse. Science. 1990;247:322–4.CrossRefPubMed
11.
go back to reference McCart AE, Vickaryous NK, Silver A. Apc mice: models, modifiers and mutants. Pathol Res Pract. 2008;204:479–90.CrossRefPubMed McCart AE, Vickaryous NK, Silver A. Apc mice: models, modifiers and mutants. Pathol Res Pract. 2008;204:479–90.CrossRefPubMed
12.
go back to reference Prechtl JC, Powley TL. Organization and distribution of the rat subdiaphragmatic vagus and associated paraganglia. J Comp Neurol. 1985;235:182–95.CrossRefPubMed Prechtl JC, Powley TL. Organization and distribution of the rat subdiaphragmatic vagus and associated paraganglia. J Comp Neurol. 1985;235:182–95.CrossRefPubMed
13.
go back to reference Berthoud HR, Carlson NR, Powley TL. Topography of efferent vagal innervation of the rat gastrointestinal tract. Am J Physiol. 1991;260(1 Pt 2):R200–7.PubMed Berthoud HR, Carlson NR, Powley TL. Topography of efferent vagal innervation of the rat gastrointestinal tract. Am J Physiol. 1991;260(1 Pt 2):R200–7.PubMed
14.
go back to reference Resende RR, Adhikari A. Cholinergic receptor pathways involved in apoptosis, cell proliferation and neuronal differentiation. Cell Commun Signal. 2009;7:20.CrossRefPubMedCentralPubMed Resende RR, Adhikari A. Cholinergic receptor pathways involved in apoptosis, cell proliferation and neuronal differentiation. Cell Commun Signal. 2009;7:20.CrossRefPubMedCentralPubMed
15.
go back to reference Paduch R, Kandefer-Szerszeń M. Transforming growth factor-beta1 (TGF-beta1) and acetylcholine (ACh) alter nitric oxide (NO) and interleukin-1beta (IL-1beta) secretion in human colon adenocarcinoma cells. In Vitro Cell Dev Biol Anim. 2009;45:543–50.CrossRefPubMed Paduch R, Kandefer-Szerszeń M. Transforming growth factor-beta1 (TGF-beta1) and acetylcholine (ACh) alter nitric oxide (NO) and interleukin-1beta (IL-1beta) secretion in human colon adenocarcinoma cells. In Vitro Cell Dev Biol Anim. 2009;45:543–50.CrossRefPubMed
16.
go back to reference Novotny A, Ryberg K, Heiman Ullmark J, Nilsson L, Khorram-Manesh A, Nordgren S, et al. Is acetylcholine a signaling molecule for human colon cancer progression? Scand J Gastroenterol. 2011;46:446–55.CrossRefPubMed Novotny A, Ryberg K, Heiman Ullmark J, Nilsson L, Khorram-Manesh A, Nordgren S, et al. Is acetylcholine a signaling molecule for human colon cancer progression? Scand J Gastroenterol. 2011;46:446–55.CrossRefPubMed
18.
go back to reference Von Rosenvinge EC, Raufman J-P. Muscarinic receptor signaling in colon cancer. Cancers. 2011;3:971–81.CrossRef Von Rosenvinge EC, Raufman J-P. Muscarinic receptor signaling in colon cancer. Cancers. 2011;3:971–81.CrossRef
19.
go back to reference Egleton RD, Brown KC, Dasgupta P. Nicotinic acetylcholine receptors in cancer: multiple roles in proliferation and inhibition of apoptosis. Trends Pharmacol Sci. 2008;29:151–8.CrossRefPubMed Egleton RD, Brown KC, Dasgupta P. Nicotinic acetylcholine receptors in cancer: multiple roles in proliferation and inhibition of apoptosis. Trends Pharmacol Sci. 2008;29:151–8.CrossRefPubMed
20.
go back to reference Watkins LR, Wiertelak EP, Goehler LE, Smith KP, Martin D, Maier SF. Characterization of cytokine-induced hyperalgesia. Brain Res. 1994;654:15–26.CrossRefPubMed Watkins LR, Wiertelak EP, Goehler LE, Smith KP, Martin D, Maier SF. Characterization of cytokine-induced hyperalgesia. Brain Res. 1994;654:15–26.CrossRefPubMed
21.
go back to reference Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S, et al. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature. 2003;421:384–8.CrossRefPubMed Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S, et al. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature. 2003;421:384–8.CrossRefPubMed
22.
go back to reference Czura CJ, Friedman SG, Tracey KJ. Neural inhibition of inflammation: the cholinergic anti-inflammatory pathway. J Endotoxin Res. 2003;9:409–13.CrossRefPubMed Czura CJ, Friedman SG, Tracey KJ. Neural inhibition of inflammation: the cholinergic anti-inflammatory pathway. J Endotoxin Res. 2003;9:409–13.CrossRefPubMed
23.
go back to reference Huston JM. The vagus nerve and the inflammatory reflex: wandering on a new treatment paradigm for systemic inflammation and sepsis. Surg Infect. 2012;13:187–93.CrossRef Huston JM. The vagus nerve and the inflammatory reflex: wandering on a new treatment paradigm for systemic inflammation and sepsis. Surg Infect. 2012;13:187–93.CrossRef
24.
go back to reference Van Westerloo DJ, Giebelen IAJ, Florquin S, Daalhuisen J, Bruno MJ, de Vos AF, et al. The cholinergic anti-inflammatory pathway regulates the host response during septic peritonitis. J Infect Dis. 2005;191:2138–48.CrossRefPubMed Van Westerloo DJ, Giebelen IAJ, Florquin S, Daalhuisen J, Bruno MJ, de Vos AF, et al. The cholinergic anti-inflammatory pathway regulates the host response during septic peritonitis. J Infect Dis. 2005;191:2138–48.CrossRefPubMed
25.
go back to reference Yang W-L, Frucht H. Cholinergic receptor up-regulates COX-2 expression and prostaglandin E2 production in colon cancer cells. Carcinogenesis. 2000;21:1789–93.CrossRefPubMed Yang W-L, Frucht H. Cholinergic receptor up-regulates COX-2 expression and prostaglandin E2 production in colon cancer cells. Carcinogenesis. 2000;21:1789–93.CrossRefPubMed
26.
go back to reference Cheng K, Samimi R, Xie G, Shant J, Drachenberg C, Wade M, et al. Acetylcholine release by human colon cancer cells mediates autocrine stimulation of cell proliferation. Am J Physiol Gastrointest Liver Physiol. 2008;295:G591–7.CrossRefPubMedCentralPubMed Cheng K, Samimi R, Xie G, Shant J, Drachenberg C, Wade M, et al. Acetylcholine release by human colon cancer cells mediates autocrine stimulation of cell proliferation. Am J Physiol Gastrointest Liver Physiol. 2008;295:G591–7.CrossRefPubMedCentralPubMed
27.
go back to reference Ukegawa J-I, Takeuchi Y, Kusayanagi S, Mitamura K. Growth-promoting effect of muscarinic acetylcholine receptors in colon cancer cells. J Cancer Res Clin Oncol. 2003;129:272–8.PubMed Ukegawa J-I, Takeuchi Y, Kusayanagi S, Mitamura K. Growth-promoting effect of muscarinic acetylcholine receptors in colon cancer cells. J Cancer Res Clin Oncol. 2003;129:272–8.PubMed
28.
go back to reference Cheng K, Zimniak P, Raufman J-P. Transactivation of the epidermal growth factor receptor mediates cholinergic agonist-induced proliferation of H508 human colon cancer cells. Cancer Res. 2003;63:6744–50.PubMed Cheng K, Zimniak P, Raufman J-P. Transactivation of the epidermal growth factor receptor mediates cholinergic agonist-induced proliferation of H508 human colon cancer cells. Cancer Res. 2003;63:6744–50.PubMed
29.
go back to reference Cargnello M, Roux PP. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev. 2011;75:50–83.CrossRefPubMedCentralPubMed Cargnello M, Roux PP. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev. 2011;75:50–83.CrossRefPubMedCentralPubMed
30.
go back to reference Park Y-S, Cho NJ. EGFR and PKC are involved in the activation of ERK1/2 and p90 RSK and the subsequent proliferation of SNU-407 colon cancer cells by muscarinic acetylcholine receptors. Mol Cell Biochem. 2012;370:191–8.CrossRefPubMed Park Y-S, Cho NJ. EGFR and PKC are involved in the activation of ERK1/2 and p90 RSK and the subsequent proliferation of SNU-407 colon cancer cells by muscarinic acetylcholine receptors. Mol Cell Biochem. 2012;370:191–8.CrossRefPubMed
31.
go back to reference Raufman J-P, Cheng K, Saxena N, Chahdi A, Belo A, Khurana S, et al. Muscarinic receptor agonists stimulate matrix metalloproteinase 1-dependent invasion of human colon cancer cells. Biochem Biophys Res Commun. 2011;415:319–24.CrossRefPubMedCentralPubMed Raufman J-P, Cheng K, Saxena N, Chahdi A, Belo A, Khurana S, et al. Muscarinic receptor agonists stimulate matrix metalloproteinase 1-dependent invasion of human colon cancer cells. Biochem Biophys Res Commun. 2011;415:319–24.CrossRefPubMedCentralPubMed
32.
go back to reference Raufman J-P, Samimi R, Shah N, Khurana S, Shant J, Drachenberg C, et al. Genetic ablation of M3 muscarinic receptors attenuates murine colon epithelial cell proliferation and neoplasia. Cancer Res. 2008;68:3573–8.CrossRefPubMedCentralPubMed Raufman J-P, Samimi R, Shah N, Khurana S, Shant J, Drachenberg C, et al. Genetic ablation of M3 muscarinic receptors attenuates murine colon epithelial cell proliferation and neoplasia. Cancer Res. 2008;68:3573–8.CrossRefPubMedCentralPubMed
33.
go back to reference Raufman J-P, Shant J, Xie G, Cheng K, Gao X-M, Shiu B, et al. Muscarinic receptor subtype-3 gene ablation and scopolamine butylbromide treatment attenuate small intestinal neoplasia in Apcmin/+ mice. Carcinogenesis. 2011;32:1396–402.CrossRefPubMedCentralPubMed Raufman J-P, Shant J, Xie G, Cheng K, Gao X-M, Shiu B, et al. Muscarinic receptor subtype-3 gene ablation and scopolamine butylbromide treatment attenuate small intestinal neoplasia in Apcmin/+ mice. Carcinogenesis. 2011;32:1396–402.CrossRefPubMedCentralPubMed
34.
go back to reference Zhao C-M, Hayakawa Y, Kodama Y, Muthupalani S, Westphalen CB, Andersen GT, et al. Denervation suppresses gastric tumorigenesis. Sci Transl Med. 2014;6:250. ra115.CrossRef Zhao C-M, Hayakawa Y, Kodama Y, Muthupalani S, Westphalen CB, Andersen GT, et al. Denervation suppresses gastric tumorigenesis. Sci Transl Med. 2014;6:250. ra115.CrossRef
35.
go back to reference Pettersson A, Nilsson L, Nylund G, Khorram-Manesh A, Nordgren S, Delbro DS. Is acetylcholine an autocrine/paracrine growth factor via the nicotinic alpha7-receptor subtype in the human colon cancer cell line HT-29? Eur J Pharmacol. 2009;609:27–33.CrossRefPubMed Pettersson A, Nilsson L, Nylund G, Khorram-Manesh A, Nordgren S, Delbro DS. Is acetylcholine an autocrine/paracrine growth factor via the nicotinic alpha7-receptor subtype in the human colon cancer cell line HT-29? Eur J Pharmacol. 2009;609:27–33.CrossRefPubMed
Metadata
Title
Extrinsic intestinal denervation modulates tumor development in the small intestine of ApcMin/+ mice
Authors
Verena Liu
Alexandra Dietrich
Michael S Kasparek
Petra Benhaqi
Marlon R Schneider
Michael Schemann
Hendrik Seeliger
Martin E Kreis
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2015
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-015-0159-0

Other articles of this Issue 1/2015

Journal of Experimental & Clinical Cancer Research 1/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine