DSpace logo

Please use this identifier to cite or link to this item: http://dspace.bits-pilani.ac.in:8080/jspui/xmlui/handle/123456789/13886
Full metadata record
DC FieldValueLanguage
dc.contributor.authorShrivastava, Richa-
dc.date.accessioned2024-01-19T06:55:11Z-
dc.date.available2024-01-19T06:55:11Z-
dc.date.issued2016-07-
dc.identifier.urihttps://www.tandfonline.com/doi/full/10.1080/2162402X.2016.1196299-
dc.identifier.urihttp://dspace.bits-pilani.ac.in:8080/jspui/xmlui/handle/123456789/13886-
dc.description.abstractThe potential of a tumor cell to metastasize profoundly depends on its microenvironment, or “niche” interactions with local components. Tumor-associated-macrophages (TAMs) are the most abundant subpopulation of tumor stroma and represent a key component of tumor microenvironment. The dynamic interaction of cancer cells with neighboring TAMs actively drive cancer progression and metastatic transformation through intercellular signaling networks that need better elucidation. Thus, current study was planned for discerning paracrine communication networks operational between TAMs, and breast cancer cells with special reference to cancer cell invasion and dissemination to distant sites. Here, we report role of MIP-1β in enhancing invasive potential of metastatic breast cancer MDA-MB-231 and MDA-MB-468 cells. In addition, the poorly metastatic MCF-7 cells were also rendered invasive by MIP-1β. The MIP-1β-driven cancer cell invasion was dependent on upregulated expression levels of MYO3A gene, which encodes an unconventional myosin super-family protein harboring a kinase domain. Ex ovo study employing Chick-embryo-model and in vivo Syngenic 4T1/BALB/c mice-model further corroborated aforementioned in vitro findings, thereby substantiating their physiological relevance. Concordantly, human breast cancer specimen exhibited significant association between mRNA expression levels of MIP-1β and MYO3A. Both, MIP-1β and MYO3A exhibited positive correlation with MMP9, an established molecular determinant of cancer cell invasion. Higher expression of these genes correlated with poor survival of breast cancer patients. Collectively, these results point toward so far undisclosed MIP-1β/MYO3A axis being operational during metastasis, wherein macrophage-derived MIP-1β potentiated cancer cell invasion and metastasis via up regulation of MYO3A gene within cancer cells. Our study exposes opportunities for devising potential anti-metastatic strategies for efficient clinical management of breast cancer.en_US
dc.language.isoenen_US
dc.publisherTaylor & Francisen_US
dc.subjectPharmacyen_US
dc.subjectInvadopodiaen_US
dc.subjectInvasionen_US
dc.subjectMigrationen_US
dc.subjectMIP-1βen_US
dc.subjectMYO3en_US
dc.subjectAMMP-9en_US
dc.titleMacrophages promote matrix protrusive and invasive function of breast cancer cells via MIP-1β dependent upregulation of MYO3A gene in breast cancer cellsen_US
dc.typeArticleen_US
Appears in Collections:Department of Pharmacy

Files in This Item:
There are no files associated with this item.


Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.