Skip to main content
Top
Published in: Cancer Cell International 1/2018

Open Access 01-12-2018 | Primary research

Neuropeptide-induced modulation of carcinogenesis in a metastatic breast cancer cell line (MDA-MB-231LUC+)

Authors: Silvia Gutierrez, M. Danilo Boada

Published in: Cancer Cell International | Issue 1/2018

Login to get access

Abstract

Background

Metastatic cancer to bone is well-known to produce extreme pain. It has been suggested that the magnitude of this perceived pain is associated with disease progression and poor prognosis. These data suggest a potential cross-talk between cancer cells and nociceptors that contribute not only to pain, but also to cancer aggressiveness although the underlying mechanisms are yet to be stablished.

Methods

The in vitro dose dependent effect of neuropeptides (NPs) (substance P [SP], calcitonin gene-related peptide and neurokinin A [NKA]) and/or its combination, on the migration and invasion of MDA-MB-231LUC+ were assessed by wound healing and collagen-based cell invasion assays, respectively. The effect of NPs on the expression of its receptors (SP [NK1] and neurokinin A receptors [NK2], CALCRL and RAMP1) and kininogen (high-molecular-weight kininogen) release to the cell culture supernatant of MDA-MB-231LUC+, were measured using western-blot analysis and an ELISA assay, respectively. Statistical significance was tested using one-way ANOVA, repeated measures ANOVA, or the paired t-test. Post-hoc testing was performed with correction for multiple comparisons as appropriate.

Results

Our data show that NPs strongly modify the chemokinetic capabilities of a cellular line commonly used as a model of metastatic cancer to bone (MDA-MB-231LUC+) and increased the expression of their receptors (NK1R, NK2R, RAMP1, and CALCRL) on these cells. Finally, we demonstrate that NPs also trigger the acute release of HMWK (Bradykinin precursor) by MDA-MB-231LUC+, a molecule with both tumorigenic and pro-nociceptive activity.

Conclusions

Based on these observations we conclude that NPs exposure modulates this breast cancer cellular line aggressiveness by increasing its ability to migrate and invade new tissues. Furthermore, these results also support the pro nociceptive and cancer promoter role of the peripheral nervous system, during the initial stages of the disease.
Literature
1.
go back to reference Reyes-Gibby CC, Anderson KO, Merriman KW, Todd KH, Shete SS, Hanna EY. Survival patterns in squamous cell carcinoma of the head and neck: pain as an independent prognostic factor for survival. J Pain. 2014;15:1015–22.CrossRef Reyes-Gibby CC, Anderson KO, Merriman KW, Todd KH, Shete SS, Hanna EY. Survival patterns in squamous cell carcinoma of the head and neck: pain as an independent prognostic factor for survival. J Pain. 2014;15:1015–22.CrossRef
2.
go back to reference Schmidt BL, Hamamoto DT, Simone DA, Wilcox GL. Mechanism of cancer pain. Mol Interv. 2010;10:164–78.CrossRef Schmidt BL, Hamamoto DT, Simone DA, Wilcox GL. Mechanism of cancer pain. Mol Interv. 2010;10:164–78.CrossRef
3.
go back to reference Lam DK, Schmidt BL. Orofacial pain onset predicts transition to head and neck cancer. Pain. 2011;152:1206–9.CrossRef Lam DK, Schmidt BL. Orofacial pain onset predicts transition to head and neck cancer. Pain. 2011;152:1206–9.CrossRef
4.
go back to reference Schmidt BL. The neurobiology of cancer pain. Neuroscientist. 2014;20(5):546–62.CrossRef Schmidt BL. The neurobiology of cancer pain. Neuroscientist. 2014;20(5):546–62.CrossRef
5.
go back to reference Sato J, Fujiwara M, Kawakami T, Sumiishi A, Sakata S, Sakamoto A, et al. Fascin expression in dendritic cells and tumor epithelium in thymoma and thymic carcinoma. Oncol Lett. 2011;2:1025–32.CrossRef Sato J, Fujiwara M, Kawakami T, Sumiishi A, Sakata S, Sakamoto A, et al. Fascin expression in dendritic cells and tumor epithelium in thymoma and thymic carcinoma. Oncol Lett. 2011;2:1025–32.CrossRef
6.
go back to reference Oliveira KG, von Zeidler SV, Lamas AZ, Podestá JR, Sena A, Souza ED, et al. Relationship of inflammatory markers and pain in patients with head and neck cancer prior to anticancer therapy. Braz J Med Biol Res. 2014;47:600–4.CrossRef Oliveira KG, von Zeidler SV, Lamas AZ, Podestá JR, Sena A, Souza ED, et al. Relationship of inflammatory markers and pain in patients with head and neck cancer prior to anticancer therapy. Braz J Med Biol Res. 2014;47:600–4.CrossRef
7.
go back to reference Mantyh PW. The neurobiology of skeletal pain. Eur J Neurosci. 2014;39:508–19.CrossRef Mantyh PW. The neurobiology of skeletal pain. Eur J Neurosci. 2014;39:508–19.CrossRef
8.
go back to reference Kuol N, Stojanovska L, Apostolopoulos V, Nurgali K. Role of the nervous system in cancer metastasis. J Exp Clin Cancer Res. 2018;37(1):5.CrossRef Kuol N, Stojanovska L, Apostolopoulos V, Nurgali K. Role of the nervous system in cancer metastasis. J Exp Clin Cancer Res. 2018;37(1):5.CrossRef
9.
go back to reference Rosso M, Muñoz M, Berger M. The role of neurokinin-1 receptor in the microenvironment of inflammation and cancer. Sci World J. 2012;2012:381434. Rosso M, Muñoz M, Berger M. The role of neurokinin-1 receptor in the microenvironment of inflammation and cancer. Sci World J. 2012;2012:381434.
10.
go back to reference Coveñas R, Muñoz M. Cancer progression and substance P. Histol Histopathol. 2014;29:881–90.PubMed Coveñas R, Muñoz M. Cancer progression and substance P. Histol Histopathol. 2014;29:881–90.PubMed
11.
go back to reference Muñoz M, Coveñas R. Involvement of substance P and the NK-1 receptor in cancer progression. Peptides. 2013;48:1–9.CrossRef Muñoz M, Coveñas R. Involvement of substance P and the NK-1 receptor in cancer progression. Peptides. 2013;48:1–9.CrossRef
12.
go back to reference Muñoz M, Coveñas R. Involvement of substance P and the NK-1 receptor in pancreatic cancer. World J Gastroenterol. 2014;20:2321–34.CrossRef Muñoz M, Coveñas R. Involvement of substance P and the NK-1 receptor in pancreatic cancer. World J Gastroenterol. 2014;20:2321–34.CrossRef
13.
go back to reference Muñoz M, Coveñas R. Involvement of substance P and the NK-1 receptor in human pathology. Amino Acids. 2014;46:1727–50.CrossRef Muñoz M, Coveñas R. Involvement of substance P and the NK-1 receptor in human pathology. Amino Acids. 2014;46:1727–50.CrossRef
14.
go back to reference Singh D, Joshi DD, Hameed M, Qian J, Gascón P, Maloof PB, et al. Increased expression of preprotachykinin-I and neurokinin receptors in human breast cancer cells: implications for bone marrow metastasis. Proc Natl Acad Sci USA. 2000;97:388–93.CrossRef Singh D, Joshi DD, Hameed M, Qian J, Gascón P, Maloof PB, et al. Increased expression of preprotachykinin-I and neurokinin receptors in human breast cancer cells: implications for bone marrow metastasis. Proc Natl Acad Sci USA. 2000;97:388–93.CrossRef
15.
go back to reference Rao G, Patel PS, Idler SP, Maloof P, Gascon P, Potian JA, et al. Facilitating role of preprotachykinin-I gene in the integration of breast cancer cells within the stromal compartment of the bone marrow: a model of early cancer progression. Cancer Res. 2004;64:2874–81.CrossRef Rao G, Patel PS, Idler SP, Maloof P, Gascon P, Potian JA, et al. Facilitating role of preprotachykinin-I gene in the integration of breast cancer cells within the stromal compartment of the bone marrow: a model of early cancer progression. Cancer Res. 2004;64:2874–81.CrossRef
16.
go back to reference Castro TA, Cohen MC, Rameshwar P. The expression of neurokinin-1 and preprotachykinin-1 in breast cancer cells depends on the relative degree of invasive and metastatic potential. Clin Exp Metastasis. 2005;22:621–8.CrossRef Castro TA, Cohen MC, Rameshwar P. The expression of neurokinin-1 and preprotachykinin-1 in breast cancer cells depends on the relative degree of invasive and metastatic potential. Clin Exp Metastasis. 2005;22:621–8.CrossRef
17.
go back to reference Singh AS, Caplan A, Corcoran KE, Fernandez JS, Preziosi M, Rameshwar P. Oncogenic and metastatic properties of preprotachykinin-I and neurokinin-1 genes. Vascul Pharmacol. 2006;45:235–42.CrossRef Singh AS, Caplan A, Corcoran KE, Fernandez JS, Preziosi M, Rameshwar P. Oncogenic and metastatic properties of preprotachykinin-I and neurokinin-1 genes. Vascul Pharmacol. 2006;45:235–42.CrossRef
18.
go back to reference Huang SC, Korlipara VL. Neurokinin-1 receptor antagonists: a comprehensive patent survey. Expert Opin Ther Pat. 2010;20:1019–45.CrossRef Huang SC, Korlipara VL. Neurokinin-1 receptor antagonists: a comprehensive patent survey. Expert Opin Ther Pat. 2010;20:1019–45.CrossRef
19.
go back to reference Rupniak NMJ, Perdona E, Griffante C, Cavallini P, Sava A, Ricca DJ, Thor KB, Burgard EC, Corsi M. Affinity, potency, efficacy, and selectivity of neurokinin A analogs at human recombinant NK2 and NK1 receptors. PLoS ONE. 2018;13(10):e0205894.CrossRef Rupniak NMJ, Perdona E, Griffante C, Cavallini P, Sava A, Ricca DJ, Thor KB, Burgard EC, Corsi M. Affinity, potency, efficacy, and selectivity of neurokinin A analogs at human recombinant NK2 and NK1 receptors. PLoS ONE. 2018;13(10):e0205894.CrossRef
20.
go back to reference Barwell J, Gingell JJ, Watkins HA, Archbold JK, Poyner DR, Hay DL. Calcitonin and calcitonin receptor-like receptors: common themes with family B GPCRs? Br J Pharmacol. 2012;166(1):51–65.CrossRef Barwell J, Gingell JJ, Watkins HA, Archbold JK, Poyner DR, Hay DL. Calcitonin and calcitonin receptor-like receptors: common themes with family B GPCRs? Br J Pharmacol. 2012;166(1):51–65.CrossRef
21.
go back to reference Wang G, Ye Y, Zhang X, Song J. Bradykinin stimulates IL-6 production and cell invasion in colorectal cancer cells. Oncol Rep. 2014;32:1709–14.CrossRef Wang G, Ye Y, Zhang X, Song J. Bradykinin stimulates IL-6 production and cell invasion in colorectal cancer cells. Oncol Rep. 2014;32:1709–14.CrossRef
22.
go back to reference Drell TL IV, Joseph J, Lang K, Niggemann B, Zaenker KS, Entschladen F. Effects of neurotransmitters on the chemokinesis and chemotaxis of MDA-MB-468 human breast carcinoma cells. Breast Cancer Res Treat. 2003;80:63–70.CrossRef Drell TL IV, Joseph J, Lang K, Niggemann B, Zaenker KS, Entschladen F. Effects of neurotransmitters on the chemokinesis and chemotaxis of MDA-MB-468 human breast carcinoma cells. Breast Cancer Res Treat. 2003;80:63–70.CrossRef
23.
go back to reference Li J, Wu Y, Schimmel N, Al-Ameen MA, Ghosh G. Breast cancer cells mechanosensing in engineered matrices: correlation with aggressive phenotype. J Mech Behav Biomed Mater. 2016;61:208–20.CrossRef Li J, Wu Y, Schimmel N, Al-Ameen MA, Ghosh G. Breast cancer cells mechanosensing in engineered matrices: correlation with aggressive phenotype. J Mech Behav Biomed Mater. 2016;61:208–20.CrossRef
24.
go back to reference Li J, Zeng Q, Zhang Y, Li X, Hu H, Miao X, et al. Neurokinin-1 receptor mediated breast cancer cell migration by increased expression of MMP-2 and MMP-14. Eur J Cell Biol. 2016;95:368–77.CrossRef Li J, Zeng Q, Zhang Y, Li X, Hu H, Miao X, et al. Neurokinin-1 receptor mediated breast cancer cell migration by increased expression of MMP-2 and MMP-14. Eur J Cell Biol. 2016;95:368–77.CrossRef
25.
go back to reference Lang K, Drell TL IV, Lindecke A, Niggemann B, Kaltschmidt C, Zaenker KS, et al. Induction of a metastatogenic tumor cell type by neurotransmitters and its pharmacological inhibition by established drugs. Int J Cancer. 2004;112:231–8.CrossRef Lang K, Drell TL IV, Lindecke A, Niggemann B, Kaltschmidt C, Zaenker KS, et al. Induction of a metastatogenic tumor cell type by neurotransmitters and its pharmacological inhibition by established drugs. Int J Cancer. 2004;112:231–8.CrossRef
26.
go back to reference Meshki J, Douglas SD, Lai JP, Schwartz L, Kilpatrick LE, Tuluc F. Neurokinin 1 receptor mediates membrane blebbing in HEK293 cells through a Rho/Rho-associated coiled-coil kinase-dependent mechanism. J Biol Chem. 2009;284:9280–9.CrossRef Meshki J, Douglas SD, Lai JP, Schwartz L, Kilpatrick LE, Tuluc F. Neurokinin 1 receptor mediates membrane blebbing in HEK293 cells through a Rho/Rho-associated coiled-coil kinase-dependent mechanism. J Biol Chem. 2009;284:9280–9.CrossRef
27.
go back to reference Mierke CT. Physical view on migration modes. Cell Adhes Migr. 2015;9:367–79.CrossRef Mierke CT. Physical view on migration modes. Cell Adhes Migr. 2015;9:367–79.CrossRef
28.
go back to reference Fackler OT, Grosse R. Cell motility through plasma membrane blebbing. J Cell Biol. 2008;181:879–84.CrossRef Fackler OT, Grosse R. Cell motility through plasma membrane blebbing. J Cell Biol. 2008;181:879–84.CrossRef
29.
go back to reference Bigioni M, Benzo A, Irrissuto C, Maggi CA, Goso C. Role of NK-1 and NK-2 tachykinin receptor antagonism on the growth of human breast carcinoma cell line MDA-MB-231. Anticancer Drugs. 2005;16:1083–9.CrossRef Bigioni M, Benzo A, Irrissuto C, Maggi CA, Goso C. Role of NK-1 and NK-2 tachykinin receptor antagonism on the growth of human breast carcinoma cell line MDA-MB-231. Anticancer Drugs. 2005;16:1083–9.CrossRef
30.
go back to reference van Moorselaar RJ, Voest EE. Angiogenesis in prostate cancer: its role in disease progression and possible therapeutic approaches. Mol Cell Endocrinol. 2002;197:239–50.CrossRef van Moorselaar RJ, Voest EE. Angiogenesis in prostate cancer: its role in disease progression and possible therapeutic approaches. Mol Cell Endocrinol. 2002;197:239–50.CrossRef
31.
go back to reference Papantoniou V, Tsiouris S, Sotiropoulou M, Valsamaki P, Koutsikos J, Ptohis N, et al. The potential role of calcitonin gene-related peptide (CGRP) in breast carcinogenesis and its correlation with 99mTc-(V)DMSA scintimammography. Am J Clin Oncol. 2007;30:420–7.CrossRef Papantoniou V, Tsiouris S, Sotiropoulou M, Valsamaki P, Koutsikos J, Ptohis N, et al. The potential role of calcitonin gene-related peptide (CGRP) in breast carcinogenesis and its correlation with 99mTc-(V)DMSA scintimammography. Am J Clin Oncol. 2007;30:420–7.CrossRef
32.
go back to reference Logan M, Anderson PD, Saab ST, Hameed O, Abdulkadir SA. RAMP1 is a direct NKX3.1 target gene up-regulated in prostate cancer that promotes tumorigenesis. Am J Pathol. 2013;183:951–63.CrossRef Logan M, Anderson PD, Saab ST, Hameed O, Abdulkadir SA. RAMP1 is a direct NKX3.1 target gene up-regulated in prostate cancer that promotes tumorigenesis. Am J Pathol. 2013;183:951–63.CrossRef
33.
go back to reference Austin M, Elliott L, Nicolaou N, Grabowska A, Hulse RP. Breast cancer induced nociceptor aberrant growth and collateral sensory axonal branching. Oncotarget. 2017;8:76606–21.CrossRef Austin M, Elliott L, Nicolaou N, Grabowska A, Hulse RP. Breast cancer induced nociceptor aberrant growth and collateral sensory axonal branching. Oncotarget. 2017;8:76606–21.CrossRef
34.
go back to reference Rozengurt E. Neuropeptides as growth factors for normal and cancerous cells. Trends Endocrinol Metab. 2002;13:128–34.CrossRef Rozengurt E. Neuropeptides as growth factors for normal and cancerous cells. Trends Endocrinol Metab. 2002;13:128–34.CrossRef
35.
go back to reference Muñoz M, Coveñas R. Neurokinin-1 receptor antagonists as antitumor drugs in gastrointestinal cancer: a new approach. Saudi J Gastroenterol. 2016;22:260–8.CrossRef Muñoz M, Coveñas R. Neurokinin-1 receptor antagonists as antitumor drugs in gastrointestinal cancer: a new approach. Saudi J Gastroenterol. 2016;22:260–8.CrossRef
36.
go back to reference Muñoz M, Coveñas R. Targeting NK-1 receptors to prevent and treat pancreatic cancer: a new therapeutic approach. Cancers (Basel). 2015;7:1215–32.CrossRef Muñoz M, Coveñas R. Targeting NK-1 receptors to prevent and treat pancreatic cancer: a new therapeutic approach. Cancers (Basel). 2015;7:1215–32.CrossRef
37.
go back to reference Muñoz M, Rosso M, Coveñas R. A new frontier in the treatment of cancer: NK-1 receptor antagonists. Curr Med Chem. 2010;17:504–16.CrossRef Muñoz M, Rosso M, Coveñas R. A new frontier in the treatment of cancer: NK-1 receptor antagonists. Curr Med Chem. 2010;17:504–16.CrossRef
38.
go back to reference Rosso M, Robles-Frías MJ, Coveñas R, Salinas-Martín MV, Muñoz M. The NK-1 receptor is expressed in human primary gastric and colon adenocarcinomas and is involved in the antitumor action of L-733,060 and the mitogenic action of substance P on human gastrointestinal cancer cell lines. Tumour Biol. 2008;29:245–54.CrossRef Rosso M, Robles-Frías MJ, Coveñas R, Salinas-Martín MV, Muñoz M. The NK-1 receptor is expressed in human primary gastric and colon adenocarcinomas and is involved in the antitumor action of L-733,060 and the mitogenic action of substance P on human gastrointestinal cancer cell lines. Tumour Biol. 2008;29:245–54.CrossRef
39.
go back to reference Brener S, González-Moles MA, Tostes D, Esteban F, Gil-Montoya JA, Ruiz-Avila I, et al. A role for the substance P/NK-1 receptor complex in cell proliferation in oral squamous cell carcinoma. Anticancer Res. 2009;29:2323–9.PubMed Brener S, González-Moles MA, Tostes D, Esteban F, Gil-Montoya JA, Ruiz-Avila I, et al. A role for the substance P/NK-1 receptor complex in cell proliferation in oral squamous cell carcinoma. Anticancer Res. 2009;29:2323–9.PubMed
40.
go back to reference Garcia-Recio S, Gascón P. Biological and pharmacological aspects of the NK1-receptor. Biomed Res Int. 2015;2015:495704.CrossRef Garcia-Recio S, Gascón P. Biological and pharmacological aspects of the NK1-receptor. Biomed Res Int. 2015;2015:495704.CrossRef
41.
go back to reference Trafton JA, Abbadie C, Basbaum AI. Differential contribution of substance P and neurokinin A to spinal cord neurokinin-1 receptor signaling in the rat. J Neurosci. 2001;21:3656–64.CrossRef Trafton JA, Abbadie C, Basbaum AI. Differential contribution of substance P and neurokinin A to spinal cord neurokinin-1 receptor signaling in the rat. J Neurosci. 2001;21:3656–64.CrossRef
42.
go back to reference Cueille C, Birot O, Bigard X, Hagner S, Garel JM. Post-transcriptional regulation of CRLR expression during hypoxia. Biochem Biophys Res Commun. 2005;326(1):23–9.CrossRef Cueille C, Birot O, Bigard X, Hagner S, Garel JM. Post-transcriptional regulation of CRLR expression during hypoxia. Biochem Biophys Res Commun. 2005;326(1):23–9.CrossRef
43.
go back to reference Kojima S, Shimo Y. Calcitonin gene-related peptide (CGRP)-enhanced non-adrenergic non-cholinergic contraction of guinea-pig proximal colon. Br J Pharmacol. 1995;115:1290–4.CrossRef Kojima S, Shimo Y. Calcitonin gene-related peptide (CGRP)-enhanced non-adrenergic non-cholinergic contraction of guinea-pig proximal colon. Br J Pharmacol. 1995;115:1290–4.CrossRef
44.
go back to reference Kojima S, Ueda S, Ikeda M, Kamikawa Y. Calcitonin gene-related peptide facilitates serotonin release from guinea-pig colonic mucosa via myenteric neurons and tachykinin NK2/NK3 receptors. Br J Pharmacol. 2004;141:385–90.CrossRef Kojima S, Ueda S, Ikeda M, Kamikawa Y. Calcitonin gene-related peptide facilitates serotonin release from guinea-pig colonic mucosa via myenteric neurons and tachykinin NK2/NK3 receptors. Br J Pharmacol. 2004;141:385–90.CrossRef
45.
go back to reference Zaidi M, Breimer LH, MacIntyre I. Production of calcitonin gene-related peptide from human cancer cells. J Endocrinol. 1989;123:159–65.CrossRef Zaidi M, Breimer LH, MacIntyre I. Production of calcitonin gene-related peptide from human cancer cells. J Endocrinol. 1989;123:159–65.CrossRef
46.
go back to reference Dray A, Perkins M. Bradykinin and inflammatory pain. Trends Neurosci. 1993;16:99–104.CrossRef Dray A, Perkins M. Bradykinin and inflammatory pain. Trends Neurosci. 1993;16:99–104.CrossRef
47.
go back to reference da Costa PL, Sirois P, Tannock IF, Chammas R. The role of kinin receptors in cancer and therapeutic opportunities. Cancer Lett. 2014;345:27–38.CrossRef da Costa PL, Sirois P, Tannock IF, Chammas R. The role of kinin receptors in cancer and therapeutic opportunities. Cancer Lett. 2014;345:27–38.CrossRef
48.
go back to reference Leeb-Lundberg LM, Marceau F, Müller-Esterl W, Pettibone DJ, Zuraw BL. International union of pharmacology. XLV. Classification of the kinin receptor family: from molecular mechanisms to pathophysiological consequences. Pharmacol Rev. 2005;57:27–77.CrossRef Leeb-Lundberg LM, Marceau F, Müller-Esterl W, Pettibone DJ, Zuraw BL. International union of pharmacology. XLV. Classification of the kinin receptor family: from molecular mechanisms to pathophysiological consequences. Pharmacol Rev. 2005;57:27–77.CrossRef
49.
go back to reference Morissette G, Houle S, Gera L, Stewart JM, Marceau F. Antagonist, partial agonist and antiproliferative actions of B-9870 (CU201) as a function of the expression and density of the bradykinin B1 and B2 receptors. Br J Pharmacol. 2007;150:369–79.CrossRef Morissette G, Houle S, Gera L, Stewart JM, Marceau F. Antagonist, partial agonist and antiproliferative actions of B-9870 (CU201) as a function of the expression and density of the bradykinin B1 and B2 receptors. Br J Pharmacol. 2007;150:369–79.CrossRef
50.
go back to reference Ehrenfeld P, Conejeros I, Pavicic MF, Matus CE, Gonzalez CB, Quest AF, et al. Activation of kinin B1 receptor increases the release of metalloproteases-2 and -9 from both estrogen-sensitive and -insensitive breast cancer cells. Cancer Lett. 2011;301:106–18.CrossRef Ehrenfeld P, Conejeros I, Pavicic MF, Matus CE, Gonzalez CB, Quest AF, et al. Activation of kinin B1 receptor increases the release of metalloproteases-2 and -9 from both estrogen-sensitive and -insensitive breast cancer cells. Cancer Lett. 2011;301:106–18.CrossRef
51.
go back to reference Damasceno IZ, Melo KR, Nascimento FD, Souza DS, Araujo MS, Souza SE, et al. Bradykinin release avoids high molecular weight kininogen endocytosis. PLoS ONE. 2015;10:e0121721.CrossRef Damasceno IZ, Melo KR, Nascimento FD, Souza DS, Araujo MS, Souza SE, et al. Bradykinin release avoids high molecular weight kininogen endocytosis. PLoS ONE. 2015;10:e0121721.CrossRef
52.
go back to reference Dendorfer A, Wellhöner P, Braun A, Roscher AA, Dominiak P. Synthesis of kininogen and degradation of bradykinin by PC12 cells. Br J Pharmacol. 1997;122:1585–92.CrossRef Dendorfer A, Wellhöner P, Braun A, Roscher AA, Dominiak P. Synthesis of kininogen and degradation of bradykinin by PC12 cells. Br J Pharmacol. 1997;122:1585–92.CrossRef
Metadata
Title
Neuropeptide-induced modulation of carcinogenesis in a metastatic breast cancer cell line (MDA-MB-231LUC+)
Authors
Silvia Gutierrez
M. Danilo Boada
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2018
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-018-0707-8

Other articles of this Issue 1/2018

Cancer Cell International 1/2018 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