The MSP‐RON axis stimulates cancer cell growth in models of triple negative breast cancer

Triple‐negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with poor prognosis and high rates of relapse. The lack of actionable targets for TNBC has contributed to the high mortality rates of this disease, and new candidate molecules for potential manipulation are urgently...

Full description

Bibliographic Details
Main Authors: Rhona Millar, Anna Kilbey, Sarah‐Jane Remak, Tesa M. Severson, Sandeep Dhayade, Emma Sandilands, Kyla Foster, David M. Bryant, Karen Blyth, Seth B. Coffelt
Format: Article
Language:English
Published: Wiley 2020-08-01
Series:Molecular Oncology
Subjects:
MSP
RON
Online Access:https://doi.org/10.1002/1878-0261.12734
id doaj-1bd5d6c4e71a47f29ed8742bf973a817
record_format Article
spelling doaj-1bd5d6c4e71a47f29ed8742bf973a8172020-11-25T03:54:28ZengWileyMolecular Oncology1574-78911878-02612020-08-011481868188010.1002/1878-0261.12734The MSP‐RON axis stimulates cancer cell growth in models of triple negative breast cancerRhona Millar0Anna Kilbey1Sarah‐Jane Remak2Tesa M. Severson3Sandeep Dhayade4Emma Sandilands5Kyla Foster6David M. Bryant7Karen Blyth8Seth B. Coffelt9Institute of Cancer Sciences University of Glasgow Glasgow UKInstitute of Cancer Sciences University of Glasgow Glasgow UKInstitute of Cancer Sciences University of Glasgow Glasgow UKDivision of Oncogenesis Netherlands Cancer Institute Amsterdam The NetherlandsCancer Research UK Beatson Institute Glasgow UKInstitute of Cancer Sciences University of Glasgow Glasgow UKInstitute of Cancer Sciences University of Glasgow Glasgow UKInstitute of Cancer Sciences University of Glasgow Glasgow UKInstitute of Cancer Sciences University of Glasgow Glasgow UKInstitute of Cancer Sciences University of Glasgow Glasgow UKTriple‐negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with poor prognosis and high rates of relapse. The lack of actionable targets for TNBC has contributed to the high mortality rates of this disease, and new candidate molecules for potential manipulation are urgently required. Here, we show that macrophage‐stimulating protein (MSP) and its tyrosine kinase receptor, Recepteur d’origine nantais (RON), are potent drivers of cancer cell growth and tumor progression in a mouse model of TNBC driven by the loss of Trp53 and Brca1. After comparison of two genetically engineered mouse models of TNBC, we found that mammary tumors from K14‐Cre;Brca1F/F;Trp53F/F (KB1P) mice exhibit high endogenous levels of MSP and RON expression. We show that MSP stimulates serine/threonine kinase 1 and extracellular regulated MAPK activation as well as cancer cell growth in cell lines derived from the two mouse models, while genetic and pharmacological inhibition of RON prevents these effects. Similarly, KB1P tumor progression in mice was robustly attenuated by treatment with a RON inhibitor with accompanied reduction in the proliferation marker, Ki‐67. Analysis of human gene expression data confirmed that the genes encoding MSP and RON are robustly expressed in human TNBC as well as other subsets of breast cancer. Our findings uncover a mouse model where MSP expression and RON expression are naturally increased, and they provide evidence that this receptor and its ligand are viable candidate molecules for targeted treatment of breast cancer.https://doi.org/10.1002/1878-0261.12734breast cancermouse modelsMSPMST1RRONtherapeutic target
collection DOAJ
language English
format Article
sources DOAJ
author Rhona Millar
Anna Kilbey
Sarah‐Jane Remak
Tesa M. Severson
Sandeep Dhayade
Emma Sandilands
Kyla Foster
David M. Bryant
Karen Blyth
Seth B. Coffelt
spellingShingle Rhona Millar
Anna Kilbey
Sarah‐Jane Remak
Tesa M. Severson
Sandeep Dhayade
Emma Sandilands
Kyla Foster
David M. Bryant
Karen Blyth
Seth B. Coffelt
The MSP‐RON axis stimulates cancer cell growth in models of triple negative breast cancer
Molecular Oncology
breast cancer
mouse models
MSP
MST1R
RON
therapeutic target
author_facet Rhona Millar
Anna Kilbey
Sarah‐Jane Remak
Tesa M. Severson
Sandeep Dhayade
Emma Sandilands
Kyla Foster
David M. Bryant
Karen Blyth
Seth B. Coffelt
author_sort Rhona Millar
title The MSP‐RON axis stimulates cancer cell growth in models of triple negative breast cancer
title_short The MSP‐RON axis stimulates cancer cell growth in models of triple negative breast cancer
title_full The MSP‐RON axis stimulates cancer cell growth in models of triple negative breast cancer
title_fullStr The MSP‐RON axis stimulates cancer cell growth in models of triple negative breast cancer
title_full_unstemmed The MSP‐RON axis stimulates cancer cell growth in models of triple negative breast cancer
title_sort msp‐ron axis stimulates cancer cell growth in models of triple negative breast cancer
publisher Wiley
series Molecular Oncology
issn 1574-7891
1878-0261
publishDate 2020-08-01
description Triple‐negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with poor prognosis and high rates of relapse. The lack of actionable targets for TNBC has contributed to the high mortality rates of this disease, and new candidate molecules for potential manipulation are urgently required. Here, we show that macrophage‐stimulating protein (MSP) and its tyrosine kinase receptor, Recepteur d’origine nantais (RON), are potent drivers of cancer cell growth and tumor progression in a mouse model of TNBC driven by the loss of Trp53 and Brca1. After comparison of two genetically engineered mouse models of TNBC, we found that mammary tumors from K14‐Cre;Brca1F/F;Trp53F/F (KB1P) mice exhibit high endogenous levels of MSP and RON expression. We show that MSP stimulates serine/threonine kinase 1 and extracellular regulated MAPK activation as well as cancer cell growth in cell lines derived from the two mouse models, while genetic and pharmacological inhibition of RON prevents these effects. Similarly, KB1P tumor progression in mice was robustly attenuated by treatment with a RON inhibitor with accompanied reduction in the proliferation marker, Ki‐67. Analysis of human gene expression data confirmed that the genes encoding MSP and RON are robustly expressed in human TNBC as well as other subsets of breast cancer. Our findings uncover a mouse model where MSP expression and RON expression are naturally increased, and they provide evidence that this receptor and its ligand are viable candidate molecules for targeted treatment of breast cancer.
topic breast cancer
mouse models
MSP
MST1R
RON
therapeutic target
url https://doi.org/10.1002/1878-0261.12734
work_keys_str_mv AT rhonamillar themspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT annakilbey themspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT sarahjaneremak themspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT tesamseverson themspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT sandeepdhayade themspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT emmasandilands themspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT kylafoster themspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT davidmbryant themspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT karenblyth themspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT sethbcoffelt themspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT rhonamillar mspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT annakilbey mspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT sarahjaneremak mspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT tesamseverson mspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT sandeepdhayade mspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT emmasandilands mspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT kylafoster mspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT davidmbryant mspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT karenblyth mspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
AT sethbcoffelt mspronaxisstimulatescancercellgrowthinmodelsoftriplenegativebreastcancer
_version_ 1724473587908214784