Cargando…

The Prolactin Inducible Protein Modulates Antitumor Immune Responses and Metastasis in a Mouse Model of Triple Negative Breast Cancer

The prolactin inducible protein (PIP) is expressed to varying degrees in more than 90% of breast cancers (BCs). Although high levels of PIP expression in BC has been shown to correlate with better prognosis and patient response to chemotherapy, some studies suggest that PIP may also play a role in m...

Descripción completa

Detalles Bibliográficos
Autores principales: Edechi, Chidalu A., Ikeogu, Nnamdi M., Akaluka, Gloria N., Terceiro, Lucas E. L., Machado, Mikayla, Salako, Enitan S., Barazandeh, Aida F., Kung, Sam K. P., Uzonna, Jude E., Myal, Yvonne
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7994859/
https://www.ncbi.nlm.nih.gov/pubmed/33777801
http://dx.doi.org/10.3389/fonc.2021.639859
_version_ 1783669844731756544
author Edechi, Chidalu A.
Ikeogu, Nnamdi M.
Akaluka, Gloria N.
Terceiro, Lucas E. L.
Machado, Mikayla
Salako, Enitan S.
Barazandeh, Aida F.
Kung, Sam K. P.
Uzonna, Jude E.
Myal, Yvonne
author_facet Edechi, Chidalu A.
Ikeogu, Nnamdi M.
Akaluka, Gloria N.
Terceiro, Lucas E. L.
Machado, Mikayla
Salako, Enitan S.
Barazandeh, Aida F.
Kung, Sam K. P.
Uzonna, Jude E.
Myal, Yvonne
author_sort Edechi, Chidalu A.
collection PubMed
description The prolactin inducible protein (PIP) is expressed to varying degrees in more than 90% of breast cancers (BCs). Although high levels of PIP expression in BC has been shown to correlate with better prognosis and patient response to chemotherapy, some studies suggest that PIP may also play a role in metastasis. Here, we investigated the role of PIP in BC using the well-established 4T1 and E0771 mouse BC cell lines. Stable expression of PIP in both cell lines did not significantly alter their proliferation, migration, and response to anticancer drugs in vitro compared to empty vector control. To assess the effect of PIP expression on breast tumorigenesis in vivo, the 4T1 syngeneic transplantable mouse model was utilized. In immunocompetent syngeneic BALB/c mice, PIP-expressing 4T1 primary tumors displayed delayed tumor onset and reduced tumor growth, and this was associated with higher percentages of natural killer cells and reduced percentages of type 2 T-helper cells in the tumor environment. The delayed tumor onset and growth were abrogated in immunodeficient mice, suggesting that PIP-mediated modulation of primary tumor growth involves an intact immune system. Paradoxically, we also observed that PIP expression was associated with a higher number of 4T1 colonies in the lungs in both the immunocompetent and immunodeficient mice. Gene expression analysis of PIP-expressing 4T1 cells (4T1-PIP) revealed that genes associated with tumor metastasis such as CCL7, MMP3 and MMP13, were significantly upregulated in 4T1-PIP cells when compared to the empty vector control (4T1-EV) cells. Collectively, these studies strongly suggest that PIP may possess a double-edge sword effect in BC, enhancing both antitumor immunity as well as metastasis.
format Online
Article
Text
id pubmed-7994859
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-79948592021-03-27 The Prolactin Inducible Protein Modulates Antitumor Immune Responses and Metastasis in a Mouse Model of Triple Negative Breast Cancer Edechi, Chidalu A. Ikeogu, Nnamdi M. Akaluka, Gloria N. Terceiro, Lucas E. L. Machado, Mikayla Salako, Enitan S. Barazandeh, Aida F. Kung, Sam K. P. Uzonna, Jude E. Myal, Yvonne Front Oncol Oncology The prolactin inducible protein (PIP) is expressed to varying degrees in more than 90% of breast cancers (BCs). Although high levels of PIP expression in BC has been shown to correlate with better prognosis and patient response to chemotherapy, some studies suggest that PIP may also play a role in metastasis. Here, we investigated the role of PIP in BC using the well-established 4T1 and E0771 mouse BC cell lines. Stable expression of PIP in both cell lines did not significantly alter their proliferation, migration, and response to anticancer drugs in vitro compared to empty vector control. To assess the effect of PIP expression on breast tumorigenesis in vivo, the 4T1 syngeneic transplantable mouse model was utilized. In immunocompetent syngeneic BALB/c mice, PIP-expressing 4T1 primary tumors displayed delayed tumor onset and reduced tumor growth, and this was associated with higher percentages of natural killer cells and reduced percentages of type 2 T-helper cells in the tumor environment. The delayed tumor onset and growth were abrogated in immunodeficient mice, suggesting that PIP-mediated modulation of primary tumor growth involves an intact immune system. Paradoxically, we also observed that PIP expression was associated with a higher number of 4T1 colonies in the lungs in both the immunocompetent and immunodeficient mice. Gene expression analysis of PIP-expressing 4T1 cells (4T1-PIP) revealed that genes associated with tumor metastasis such as CCL7, MMP3 and MMP13, were significantly upregulated in 4T1-PIP cells when compared to the empty vector control (4T1-EV) cells. Collectively, these studies strongly suggest that PIP may possess a double-edge sword effect in BC, enhancing both antitumor immunity as well as metastasis. Frontiers Media S.A. 2021-03-12 /pmc/articles/PMC7994859/ /pubmed/33777801 http://dx.doi.org/10.3389/fonc.2021.639859 Text en Copyright © 2021 Edechi, Ikeogu, Akaluka, Terceiro, Machado, Salako, Barazandeh, Kung, Uzonna and Myal http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Oncology
Edechi, Chidalu A.
Ikeogu, Nnamdi M.
Akaluka, Gloria N.
Terceiro, Lucas E. L.
Machado, Mikayla
Salako, Enitan S.
Barazandeh, Aida F.
Kung, Sam K. P.
Uzonna, Jude E.
Myal, Yvonne
The Prolactin Inducible Protein Modulates Antitumor Immune Responses and Metastasis in a Mouse Model of Triple Negative Breast Cancer
title The Prolactin Inducible Protein Modulates Antitumor Immune Responses and Metastasis in a Mouse Model of Triple Negative Breast Cancer
title_full The Prolactin Inducible Protein Modulates Antitumor Immune Responses and Metastasis in a Mouse Model of Triple Negative Breast Cancer
title_fullStr The Prolactin Inducible Protein Modulates Antitumor Immune Responses and Metastasis in a Mouse Model of Triple Negative Breast Cancer
title_full_unstemmed The Prolactin Inducible Protein Modulates Antitumor Immune Responses and Metastasis in a Mouse Model of Triple Negative Breast Cancer
title_short The Prolactin Inducible Protein Modulates Antitumor Immune Responses and Metastasis in a Mouse Model of Triple Negative Breast Cancer
title_sort prolactin inducible protein modulates antitumor immune responses and metastasis in a mouse model of triple negative breast cancer
topic Oncology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7994859/
https://www.ncbi.nlm.nih.gov/pubmed/33777801
http://dx.doi.org/10.3389/fonc.2021.639859
work_keys_str_mv AT edechichidalua theprolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT ikeogunnamdim theprolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT akalukaglorian theprolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT terceirolucasel theprolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT machadomikayla theprolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT salakoenitans theprolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT barazandehaidaf theprolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT kungsamkp theprolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT uzonnajudee theprolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT myalyvonne theprolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT edechichidalua prolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT ikeogunnamdim prolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT akalukaglorian prolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT terceirolucasel prolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT machadomikayla prolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT salakoenitans prolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT barazandehaidaf prolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT kungsamkp prolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT uzonnajudee prolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer
AT myalyvonne prolactininducibleproteinmodulatesantitumorimmuneresponsesandmetastasisinamousemodeloftriplenegativebreastcancer