Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

Open Access 01-12-2021 | Colorectal Cancer | Research

Neuromedin U induces an invasive phenotype in CRC cells expressing the NMUR2 receptor

Authors: Patrycja Przygodzka, Ewelina Sochacka, Kamila Soboska, Marcin Pacholczyk, Izabela Papiewska-Pająk, Tomasz Przygodzki, Przemysław Płociński, Steven Ballet, An De Prins, Joanna Boncela

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Background

Successful colorectal cancer (CRC) therapy often depends on the accurate identification of primary tumours with invasive potential. There is still a lack of identified pathological factors associated with disease recurrence that could help in making treatment decisions. Neuromedin U (NMU) is a secretory neuropeptide that was first isolated from the porcine spinal cord, and it has emerged as a novel factor involved in the tumorigenesis and/or metastasis of many types of cancers. Previously associated with processes leading to CRC cell invasiveness, NMU has the potential to be a marker of poor outcome, but it has not been extensively studied in CRC.

Methods

Data from The Cancer Genome Atlas (TCGA) were used to analyse NMU and NMU receptor (NMUR1 and NMUR2) expression in CRC tissues vs. normal tissues, and real-time PCR was used for NMU and NMU receptor expression analysis. NMU protein detection was performed by immunoblotting. Secreted NMU was immunoprecipitated from cell culture-conditioned media and analysed by immunoblotting and protein sequencing. DNA demethylation by 5-aza-CdR was used to analyse the regulation of NMUR1 and NMUR2 expression. NMU receptor activity was monitored by detecting calcium mobilisation in cells loaded with fluo-4, and ERK1/2 kinase activation was detected after treatment with NMU or receptor agonist. Cell migration and invasion were investigated using membrane filters. Integrin expression was evaluated by flow cytometry.

Results

The obtained data revealed elevated expression of NMU and NMUR2 in CRC tissue samples and variable expression in the analysed CRC cell lines. We have shown, for the first time, that NMUR2 activation induces signalling in CRC cells and that NMU increases the motility and invasiveness of NMUR2-positive CRC cells and increases prometastatic integrin receptor subunit expression.

Conclusions

Our results show the ability of CRC cells to respond to NMU via activation of the NMUR2 receptor, which ultimately leads to a shift in the CRC phenotype towards a more invasive phenotype.
Appendix
Available only for authorised users
Literature
1.
go back to reference Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Pineros M, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019;144(8):1941–53.CrossRefPubMed Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Pineros M, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019;144(8):1941–53.CrossRefPubMed
2.
3.
go back to reference Guinney J, Dienstmann R, Wang X, de Reynies A, Schlicker A, Soneson C, et al. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21(11):1350–6.PubMedPubMedCentralCrossRef Guinney J, Dienstmann R, Wang X, de Reynies A, Schlicker A, Soneson C, et al. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21(11):1350–6.PubMedPubMedCentralCrossRef
4.
go back to reference Wang W, Kandimalla R, Huang H, Zhu L, Li Y, Gao F, et al. Molecular subtyping of colorectal cancer: Recent progress, new challenges and emerging opportunities. Semin Cancer Biol. 2019;55:37–52.PubMedCrossRef Wang W, Kandimalla R, Huang H, Zhu L, Li Y, Gao F, et al. Molecular subtyping of colorectal cancer: Recent progress, new challenges and emerging opportunities. Semin Cancer Biol. 2019;55:37–52.PubMedCrossRef
5.
go back to reference Przygodzka P, Papiewska-Pajak I, Bogusz H, Kryczka J, Sobierajska K, Kowalska MA, et al. Neuromedin U is upregulated by Snail at early stages of EMT in HT29 colon cancer cells. Biochim Biophys Acta. 2016;1860(11 Pt A):2445–53.PubMedCrossRef Przygodzka P, Papiewska-Pajak I, Bogusz H, Kryczka J, Sobierajska K, Kowalska MA, et al. Neuromedin U is upregulated by Snail at early stages of EMT in HT29 colon cancer cells. Biochim Biophys Acta. 2016;1860(11 Pt A):2445–53.PubMedCrossRef
6.
go back to reference Przygodzka P, Soboska K, Sochacka E, Boncela J. Neuromedin U. A Small Peptide in the Big World of Cancer. Basel: Cancers; 2019;11(9):1312. Przygodzka P, Soboska K, Sochacka E, Boncela J. Neuromedin U. A Small Peptide in the Big World of Cancer. Basel: Cancers; 2019;11(9):1312.
7.
go back to reference Li Q, Han L, Ruan S, Shen S, Cao Q, Cai X, et al. The prognostic value of neuromedin U in patients with hepatocellular carcinoma. BMC Cancer. 2020;20(1):95.PubMedPubMedCentralCrossRef Li Q, Han L, Ruan S, Shen S, Cao Q, Cai X, et al. The prognostic value of neuromedin U in patients with hepatocellular carcinoma. BMC Cancer. 2020;20(1):95.PubMedPubMedCentralCrossRef
8.
go back to reference Mori K, Miyazato M, Ida T, Murakami N, Serino R, Ueta Y, et al. Identification of neuromedin S and its possible role in the mammalian circadian oscillator system. EMBO J. 2005;24(2):325–35.PubMedPubMedCentralCrossRef Mori K, Miyazato M, Ida T, Murakami N, Serino R, Ueta Y, et al. Identification of neuromedin S and its possible role in the mammalian circadian oscillator system. EMBO J. 2005;24(2):325–35.PubMedPubMedCentralCrossRef
9.
go back to reference Mitchell JD, Maguire JJ, Davenport AP. Emerging pharmacology and physiology of neuromedin U and the structurally related peptide neuromedin S. Br J Pharmacol. 2009;158(1):87–103.PubMedPubMedCentralCrossRef Mitchell JD, Maguire JJ, Davenport AP. Emerging pharmacology and physiology of neuromedin U and the structurally related peptide neuromedin S. Br J Pharmacol. 2009;158(1):87–103.PubMedPubMedCentralCrossRef
10.
go back to reference Yates AD, Achuthan P, Akanni W, Allen J, Allen J, Alvarez-Jarreta J, et al. Ensembl 2020. Nucleic Acids Res. 2020;48(D1):D682-D8. Yates AD, Achuthan P, Akanni W, Allen J, Allen J, Alvarez-Jarreta J, et al. Ensembl 2020. Nucleic Acids Res. 2020;48(D1):D682-D8.
11.
go back to reference Brighton PJ, Szekeres PG, Willars GB. Neuromedin U and its receptors: structure, function, and physiological roles. Pharmacol Rev. 2004;56(2):231–48.PubMedCrossRef Brighton PJ, Szekeres PG, Willars GB. Neuromedin U and its receptors: structure, function, and physiological roles. Pharmacol Rev. 2004;56(2):231–48.PubMedCrossRef
12.
go back to reference De Prins A, Van Eeckhaut A, Smolders I, Tourwe D, Ballet S. Neuromedin U and Structural Analogs: An Overview of their Structure, Function and Selectivity. Curr Med Chem. 2020;27(39):6744–68.PubMedCrossRef De Prins A, Van Eeckhaut A, Smolders I, Tourwe D, Ballet S. Neuromedin U and Structural Analogs: An Overview of their Structure, Function and Selectivity. Curr Med Chem. 2020;27(39):6744–68.PubMedCrossRef
13.
go back to reference Peier AM, Desai K, Hubert J, Du X, Yang L, Qian Y, et al. Effects of peripherally administered neuromedin U on energy and glucose homeostasis. Endocrinology. 2011;152(7):2644–54.PubMedCrossRef Peier AM, Desai K, Hubert J, Du X, Yang L, Qian Y, et al. Effects of peripherally administered neuromedin U on energy and glucose homeostasis. Endocrinology. 2011;152(7):2644–54.PubMedCrossRef
14.
go back to reference Teranishi H, Hanada R. Neuromedin U. a Key Molecule in Metabolic Disorders. Int J Mol Sci. 2021;22(8):4238. Teranishi H, Hanada R. Neuromedin U. a Key Molecule in Metabolic Disorders. Int J Mol Sci. 2021;22(8):4238.
15.
go back to reference Lin TY, Huang WL, Lee WY, Luo CW. Identifying a Neuromedin U Receptor 2 Splice Variant and Determining Its Roles in the Regulation of Signaling and Tumorigenesis In Vitro. PLoS One. 2015;10(8):e0136836.PubMedPubMedCentralCrossRef Lin TY, Huang WL, Lee WY, Luo CW. Identifying a Neuromedin U Receptor 2 Splice Variant and Determining Its Roles in the Regulation of Signaling and Tumorigenesis In Vitro. PLoS One. 2015;10(8):e0136836.PubMedPubMedCentralCrossRef
16.
go back to reference Minamino N, Kangawa K, Matsuo H. Neuromedin. U-8 and U-25: novel uterus stimulating and hypertensive peptides identified in porcine spinal cord. Biochem Biophys Res Commun. 1985;130(3):1078–85.PubMedCrossRef Minamino N, Kangawa K, Matsuo H. Neuromedin. U-8 and U-25: novel uterus stimulating and hypertensive peptides identified in porcine spinal cord. Biochem Biophys Res Commun. 1985;130(3):1078–85.PubMedCrossRef
17.
go back to reference De Prins A, Martin C, Van Wanseele Y, Skov LJ, Tomboly C, Tourwe D, et al. Development of potent and proteolytically stable human neuromedin U receptor agonists. Eur J Med Chem. 2018;144:887–97.PubMedCrossRef De Prins A, Martin C, Van Wanseele Y, Skov LJ, Tomboly C, Tourwe D, et al. Development of potent and proteolytically stable human neuromedin U receptor agonists. Eur J Med Chem. 2018;144:887–97.PubMedCrossRef
18.
go back to reference Thery C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 2006;Chap. 3:Unit 3 22. Thery C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 2006;Chap. 3:Unit 3 22.
19.
go back to reference Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, et al. Fiji: an open-source platform for biological-image analysis. Nat Methods. 2012;9(7):676–82.PubMedCrossRef Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, et al. Fiji: an open-source platform for biological-image analysis. Nat Methods. 2012;9(7):676–82.PubMedCrossRef
20.
go back to reference Tsanou E, Peschos D, Batistatou A, Charalabopoulos A, Charalabopoulos K. The E-cadherin adhesion molecule and colorectal cancer. A global literature approach. Anticancer Res. 2008;28(6A):3815–26.PubMed Tsanou E, Peschos D, Batistatou A, Charalabopoulos A, Charalabopoulos K. The E-cadherin adhesion molecule and colorectal cancer. A global literature approach. Anticancer Res. 2008;28(6A):3815–26.PubMed
21.
go back to reference Said AH, Raufman JP, Xie G. The role of matrix metalloproteinases in colorectal cancer. Cancers (Basel). 2014;6(1):366–75.CrossRef Said AH, Raufman JP, Xie G. The role of matrix metalloproteinases in colorectal cancer. Cancers (Basel). 2014;6(1):366–75.CrossRef
22.
go back to reference Schlicker A, Beran G, Chresta CM, McWalter G, Pritchard A, Weston S, et al. Subtypes of primary colorectal tumors correlate with response to targeted treatment in colorectal cell lines. BMC Med Genomics. 2012;5:66.PubMedPubMedCentralCrossRef Schlicker A, Beran G, Chresta CM, McWalter G, Pritchard A, Weston S, et al. Subtypes of primary colorectal tumors correlate with response to targeted treatment in colorectal cell lines. BMC Med Genomics. 2012;5:66.PubMedPubMedCentralCrossRef
23.
go back to reference Berg KCG, Eide PW, Eilertsen IA, Johannessen B, Bruun J, Danielsen SA, et al. Multi-omics of 34 colorectal cancer cell lines - a resource for biomedical studies. Mol Cancer. 2017;16(1):116.PubMedPubMedCentralCrossRef Berg KCG, Eide PW, Eilertsen IA, Johannessen B, Bruun J, Danielsen SA, et al. Multi-omics of 34 colorectal cancer cell lines - a resource for biomedical studies. Mol Cancer. 2017;16(1):116.PubMedPubMedCentralCrossRef
24.
go back to reference Zheng X, Xu K, Zhou B, Chen T, Huang Y, Li Q, et al. A circulating extracellular vesicles-based novel screening tool for colorectal cancer revealed by shotgun and data-independent acquisition mass spectrometry. J Extracell Vesicles. 2020;9(1):1750202.PubMedPubMedCentralCrossRef Zheng X, Xu K, Zhou B, Chen T, Huang Y, Li Q, et al. A circulating extracellular vesicles-based novel screening tool for colorectal cancer revealed by shotgun and data-independent acquisition mass spectrometry. J Extracell Vesicles. 2020;9(1):1750202.PubMedPubMedCentralCrossRef
25.
go back to reference Kamimae S, Yamamoto E, Kai M, Niinuma T, Yamano HO, Nojima M, et al. Epigenetic silencing of NTSR1 is associated with lateral and noninvasive growth of colorectal tumors. Oncotarget. 2015;6(30):29975–90.PubMedPubMedCentralCrossRef Kamimae S, Yamamoto E, Kai M, Niinuma T, Yamano HO, Nojima M, et al. Epigenetic silencing of NTSR1 is associated with lateral and noninvasive growth of colorectal tumors. Oncotarget. 2015;6(30):29975–90.PubMedPubMedCentralCrossRef
26.
go back to reference Takahashi K, Furukawa C, Takano A, Ishikawa N, Kato T, Hayama S, et al. The neuromedin U-growth hormone secretagogue receptor 1b/neurotensin receptor 1 oncogenic signaling pathway as a therapeutic target for lung cancer. Cancer Res. 2006;66(19):9408–19.PubMedCrossRef Takahashi K, Furukawa C, Takano A, Ishikawa N, Kato T, Hayama S, et al. The neuromedin U-growth hormone secretagogue receptor 1b/neurotensin receptor 1 oncogenic signaling pathway as a therapeutic target for lung cancer. Cancer Res. 2006;66(19):9408–19.PubMedCrossRef
27.
go back to reference Jin W. Regulation of Src Family Kinases during Colorectal Cancer Development and Its Clinical Implications. Basel: Cancers; 2020;12(5):1339. Jin W. Regulation of Src Family Kinases during Colorectal Cancer Development and Its Clinical Implications. Basel: Cancers; 2020;12(5):1339.
28.
go back to reference Wang L, Chen C, Li F, Hua QQ, Chen S, Xiao B, et al. Overexpression of neuromedin U is correlated with regional metastasis of head and neck squamous cell carcinoma. Mol Med Rep. 2016;14(2):1075–82.PubMedPubMedCentralCrossRef Wang L, Chen C, Li F, Hua QQ, Chen S, Xiao B, et al. Overexpression of neuromedin U is correlated with regional metastasis of head and neck squamous cell carcinoma. Mol Med Rep. 2016;14(2):1075–82.PubMedPubMedCentralCrossRef
29.
go back to reference Lin TY, Wu FJ, Chang CL, Li Z, Luo CW. NMU signaling promotes endometrial cancer cell progression by modulating adhesion signaling. Oncotarget. 2016;7(9):10228–42.PubMedPubMedCentralCrossRef Lin TY, Wu FJ, Chang CL, Li Z, Luo CW. NMU signaling promotes endometrial cancer cell progression by modulating adhesion signaling. Oncotarget. 2016;7(9):10228–42.PubMedPubMedCentralCrossRef
30.
go back to reference Garczyk S, Klotz N, Szczepanski S, Denecke B, Antonopoulos W, von Stillfried S, et al. Oncogenic features of neuromedin U in breast cancer are associated with NMUR2 expression involving crosstalk with members of the WNT signaling pathway. Oncotarget. 2017;8(22):36246–65.PubMedPubMedCentralCrossRef Garczyk S, Klotz N, Szczepanski S, Denecke B, Antonopoulos W, von Stillfried S, et al. Oncogenic features of neuromedin U in breast cancer are associated with NMUR2 expression involving crosstalk with members of the WNT signaling pathway. Oncotarget. 2017;8(22):36246–65.PubMedPubMedCentralCrossRef
31.
go back to reference Kaushik AC, Mehmood A, Wei DQ, Dai X. Systems Biology Integration and Screening of Reliable Prognostic Markers to Create Synergies in the Control of Lung Cancer Patients. Front Mol Biosci. 2020;7:47.PubMedPubMedCentralCrossRef Kaushik AC, Mehmood A, Wei DQ, Dai X. Systems Biology Integration and Screening of Reliable Prognostic Markers to Create Synergies in the Control of Lung Cancer Patients. Front Mol Biosci. 2020;7:47.PubMedPubMedCentralCrossRef
32.
go back to reference Ketterer K, Kong B, Frank D, Giese NA, Bauer A, Hoheisel J, et al. Neuromedin U is overexpressed in pancreatic cancer and increases invasiveness via the hepatocyte growth factor c-Met pathway. Cancer Lett. 2009;277(1):72–81.PubMedCrossRef Ketterer K, Kong B, Frank D, Giese NA, Bauer A, Hoheisel J, et al. Neuromedin U is overexpressed in pancreatic cancer and increases invasiveness via the hepatocyte growth factor c-Met pathway. Cancer Lett. 2009;277(1):72–81.PubMedCrossRef
33.
go back to reference Martinez VG, O’Driscoll L. Neuromedin U. a multifunctional neuropeptide with pleiotropic roles. Clin Chem. 2015;61(3):471–82.PubMedCrossRef Martinez VG, O’Driscoll L. Neuromedin U. a multifunctional neuropeptide with pleiotropic roles. Clin Chem. 2015;61(3):471–82.PubMedCrossRef
34.
go back to reference Karsenty G. Translational Endocrinology of Bone: Reproduction, Metabolism, and the Central Nervous System, 1st eds. Neuromedin U, Karsenty G, editors: Ed. Academic Press; 2012:27–38. Karsenty G. Translational Endocrinology of Bone: Reproduction, Metabolism, and the Central Nervous System, 1st eds. Neuromedin U, Karsenty G, editors: Ed. Academic Press; 2012:27–38.
35.
go back to reference Ji Q, Zhou L, Sui H, Yang L, Wu X, Song Q, et al. Primary tumors release ITGBL1-rich extracellular vesicles to promote distal metastatic tumor growth through fibroblast-niche formation. Nat Commun. 2020;11(1):1211.PubMedPubMedCentralCrossRef Ji Q, Zhou L, Sui H, Yang L, Wu X, Song Q, et al. Primary tumors release ITGBL1-rich extracellular vesicles to promote distal metastatic tumor growth through fibroblast-niche formation. Nat Commun. 2020;11(1):1211.PubMedPubMedCentralCrossRef
36.
go back to reference Martinez VG, O’Neill S, Salimu J, Breslin S, Clayton A, Crown J, et al. Resistance to HER2-targeted anti-cancer drugs is associated with immune evasion in cancer cells and their derived extracellular vesicles. Oncoimmunology. 2017;6(12):e1362530.PubMedPubMedCentralCrossRef Martinez VG, O’Neill S, Salimu J, Breslin S, Clayton A, Crown J, et al. Resistance to HER2-targeted anti-cancer drugs is associated with immune evasion in cancer cells and their derived extracellular vesicles. Oncoimmunology. 2017;6(12):e1362530.PubMedPubMedCentralCrossRef
37.
go back to reference Ehrlich M. DNA methylation in cancer: too much, but also too little. Oncogene. 2002;21(35):5400–13.PubMedCrossRef Ehrlich M. DNA methylation in cancer: too much, but also too little. Oncogene. 2002;21(35):5400–13.PubMedCrossRef
38.
go back to reference Herszenyi L, Barabas L, Hritz I, Istvan G, Tulassay Z. Impact of proteolytic enzymes in colorectal cancer development and progression. World J Gastroenterol. 2014;20(37):13246–57.PubMedPubMedCentralCrossRef Herszenyi L, Barabas L, Hritz I, Istvan G, Tulassay Z. Impact of proteolytic enzymes in colorectal cancer development and progression. World J Gastroenterol. 2014;20(37):13246–57.PubMedPubMedCentralCrossRef
39.
go back to reference Hashimoto T, Kurosawa K, Sakura N. Structure-activity relationships of neuromedin U. II. Highly potent analogs substituted or modified at the N-terminus of neuromedin U-8. Chem Pharm Bull (Tokyo). 1995;43(7):1154–7.CrossRef Hashimoto T, Kurosawa K, Sakura N. Structure-activity relationships of neuromedin U. II. Highly potent analogs substituted or modified at the N-terminus of neuromedin U-8. Chem Pharm Bull (Tokyo). 1995;43(7):1154–7.CrossRef
40.
go back to reference Rajagopal S, Shenoy SK. GPCR desensitization: Acute and prolonged phases. Cell Signal. 2018;41:9–16.PubMedCrossRef Rajagopal S, Shenoy SK. GPCR desensitization: Acute and prolonged phases. Cell Signal. 2018;41:9–16.PubMedCrossRef
41.
go back to reference Yang X, Wang CC, Lee WYW, Trovik J, Chung TKH, Kwong J. Long non-coding RNA HAND2-AS1 inhibits invasion and metastasis in endometrioid endometrial carcinoma through inactivating neuromedin U. Cancer Lett. 2018;413:23–34.PubMedCrossRef Yang X, Wang CC, Lee WYW, Trovik J, Chung TKH, Kwong J. Long non-coding RNA HAND2-AS1 inhibits invasion and metastasis in endometrioid endometrial carcinoma through inactivating neuromedin U. Cancer Lett. 2018;413:23–34.PubMedCrossRef
42.
go back to reference Martinez VG, Crown J, Porter RK, O’Driscoll L. Neuromedin U alters bioenergetics and expands the cancer stem cell phenotype in HER2-positive breast cancer. Int J Cancer. 2017;140(12):2771–84.PubMedCrossRef Martinez VG, Crown J, Porter RK, O’Driscoll L. Neuromedin U alters bioenergetics and expands the cancer stem cell phenotype in HER2-positive breast cancer. Int J Cancer. 2017;140(12):2771–84.PubMedCrossRef
43.
go back to reference Rani S, Corcoran C, Shiels L, Germano S, Breslin S, Madden S, et al. Neuromedin U: a candidate biomarker and therapeutic target to predict and overcome resistance to HER-tyrosine kinase inhibitors. Cancer Res. 2014;74(14):3821–33.PubMedCrossRef Rani S, Corcoran C, Shiels L, Germano S, Breslin S, Madden S, et al. Neuromedin U: a candidate biomarker and therapeutic target to predict and overcome resistance to HER-tyrosine kinase inhibitors. Cancer Res. 2014;74(14):3821–33.PubMedCrossRef
44.
go back to reference Derwinger K, Kodeda K, Bexe-Lindskog E, Taflin H. Tumour differentiation grade is associated with TNM staging and the risk of node metastasis in colorectal cancer. Acta Oncol. 2010;49(1):57–62.PubMedCrossRef Derwinger K, Kodeda K, Bexe-Lindskog E, Taflin H. Tumour differentiation grade is associated with TNM staging and the risk of node metastasis in colorectal cancer. Acta Oncol. 2010;49(1):57–62.PubMedCrossRef
45.
46.
go back to reference Leijssen LGJ, Dinaux AM, Taylor MS, Deshpande V, Kunitake H, Bordeianou LG, et al. Perineural Invasion Is a Prognostic but not a Predictive Factor in Nonmetastatic Colon Cancer. Dis Colon Rectum. 2019;62(10):1212–21.PubMedCrossRef Leijssen LGJ, Dinaux AM, Taylor MS, Deshpande V, Kunitake H, Bordeianou LG, et al. Perineural Invasion Is a Prognostic but not a Predictive Factor in Nonmetastatic Colon Cancer. Dis Colon Rectum. 2019;62(10):1212–21.PubMedCrossRef
48.
go back to reference Yoshimura K, Meckel KF, Laird LS, Chia CY, Park JJ, Olino KL, et al. Integrin alpha2 mediates selective metastasis to the liver. Cancer Res. 2009;69(18):7320–8.PubMedPubMedCentralCrossRef Yoshimura K, Meckel KF, Laird LS, Chia CY, Park JJ, Olino KL, et al. Integrin alpha2 mediates selective metastasis to the liver. Cancer Res. 2009;69(18):7320–8.PubMedPubMedCentralCrossRef
49.
go back to reference Yu M, Chu S, Fei B, Fang X, Liu Z. O-GlcNAcylation of ITGA5 facilitates the occurrence and development of colorectal cancer. Exp Cell Res. 2019;382(2):111464.PubMedCrossRef Yu M, Chu S, Fei B, Fang X, Liu Z. O-GlcNAcylation of ITGA5 facilitates the occurrence and development of colorectal cancer. Exp Cell Res. 2019;382(2):111464.PubMedCrossRef
50.
go back to reference Beaulieu JF. Integrin alpha6beta4 in Colorectal Cancer: Expression, Regulation, Functional Alterations and Use as a Biomarker. Basel: Cancers; 2019;12(1):41. Beaulieu JF. Integrin alpha6beta4 in Colorectal Cancer: Expression, Regulation, Functional Alterations and Use as a Biomarker. Basel: Cancers; 2019;12(1):41.
51.
go back to reference Viana Lde S, Affonso RJ Jr, Silva SR, Denadai MV, Matos D, Salinas de Souza C, et al. Relationship between the expression of the extracellular matrix genes SPARC, SPP1, FN1, ITGA5 and ITGAV and clinicopathological parameters of tumor progression and colorectal cancer dissemination. Oncology. 2013;84(2):81–91.PubMedCrossRef Viana Lde S, Affonso RJ Jr, Silva SR, Denadai MV, Matos D, Salinas de Souza C, et al. Relationship between the expression of the extracellular matrix genes SPARC, SPP1, FN1, ITGA5 and ITGAV and clinicopathological parameters of tumor progression and colorectal cancer dissemination. Oncology. 2013;84(2):81–91.PubMedCrossRef
52.
go back to reference Liu QZ, Gao XH, Chang WJ, Gong HF, Fu CG, Zhang W, et al. Expression of ITGB1 predicts prognosis in colorectal cancer: a large prospective study based on tissue microarray. Int J Clin Exp Pathol. 2015;8(10):12802–10.PubMedPubMedCentral Liu QZ, Gao XH, Chang WJ, Gong HF, Fu CG, Zhang W, et al. Expression of ITGB1 predicts prognosis in colorectal cancer: a large prospective study based on tissue microarray. Int J Clin Exp Pathol. 2015;8(10):12802–10.PubMedPubMedCentral
53.
go back to reference Peng C, Zou X, Xia W, Gao H, Li Z, Liu N, et al. Integrin alphavbeta6 plays a bi-directional regulation role between colon cancer cells and cancer-associated fibroblasts. Biosci Rep. 2018;38(6):BSR20180243. Peng C, Zou X, Xia W, Gao H, Li Z, Liu N, et al. Integrin alphavbeta6 plays a bi-directional regulation role between colon cancer cells and cancer-associated fibroblasts. Biosci Rep. 2018;38(6):BSR20180243.
54.
go back to reference Yoshioka T, Nishikawa Y, Ito R, Kawamata M, Doi Y, Yamamoto Y, et al. Significance of integrin alphavbeta5 and erbB3 in enhanced cell migration and liver metastasis of colon carcinomas stimulated by hepatocyte-derived heregulin. Cancer Sci. 2010;101(9):2011–8.PubMedCrossRef Yoshioka T, Nishikawa Y, Ito R, Kawamata M, Doi Y, Yamamoto Y, et al. Significance of integrin alphavbeta5 and erbB3 in enhanced cell migration and liver metastasis of colon carcinomas stimulated by hepatocyte-derived heregulin. Cancer Sci. 2010;101(9):2011–8.PubMedCrossRef
Metadata
Title
Neuromedin U induces an invasive phenotype in CRC cells expressing the NMUR2 receptor
Authors
Patrycja Przygodzka
Ewelina Sochacka
Kamila Soboska
Marcin Pacholczyk
Izabela Papiewska-Pająk
Tomasz Przygodzki
Przemysław Płociński
Steven Ballet
An De Prins
Joanna Boncela
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-021-02073-8

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 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