Apasu, Jonathan Edem: The influence of neuronal calcium sensor 1 on metastasis of triple-negative breast cancer. - Bonn, 2020. - Dissertation, Rheinische Friedrich-Wilhelms-Universität Bonn.
Online-Ausgabe in bonndoc: https://nbn-resolving.org/urn:nbn:de:hbz:5-59858
@phdthesis{handle:20.500.11811/8666,
urn: https://nbn-resolving.org/urn:nbn:de:hbz:5-59858,
author = {{Jonathan Edem Apasu}},
title = {The influence of neuronal calcium sensor 1 on metastasis of triple-negative breast cancer},
school = {Rheinische Friedrich-Wilhelms-Universität Bonn},
year = 2020,
month = oct,

note = {At present, breast cancer is the leading global cause of cancer morbidity and mortality in women. The most aggressive subtype - triple-negative breast cancer - evades current targeted therapy schemes and lacks sufficient prognostic markers to predict disease progression. Hence, there is an urgent clinical need for novel prognostic and therapeutic biomarkers.
High expression of NCS1 has been linked to poor outcomes in breast cancer patients as well as in patients suffering from liver cancer. Recent investigations suggest that high NCS1 levels accelerate metastatic spread of breast cancer by promoting crucial pro-metastatic capabilities. Moreover, previous in vitro studies provide strong evidence of increased cancer cell invasion, increased cancer cell survival and altered cancer cell adhesion due to high NCS1 levels. To test if these metastasis enhancing phenotypes also confer a functional effect in vivo, the effect of increased NCS1 on the metastatic capabilities of triple-negative breast cancer cells were investigated in a mouse xenograft model.
By utilizing a luciferase, photon-flux based, life imaging system, significantly accelerated lung metastasis formation by NCS1 overexpressing breast cancer cells, as compared to cells with a control level of NCS1, was demonstrated. Histopathological assessment of the developed lung metastases further delivered profound evidence that NCS1 overexpression facilitated early metastatic spread to the lungs and additionally increased cancer cell survival in mature metastases by preventing necrosis.
In an attempt to elucidate the metastasis promoting mechanisms, in vitro experiments were conducted. Herein, a migration favoring cellular phenotype of NCS1 overexpressing cells, as well as close association of NCS1 with actin in cellular protrusions, were successfully demonstrated. The observation of increased cell motility of NCS1 overexpressing cells in 2-D and 3-D motility assays confirmed a functional effect of this phenotype.
NCS1, as a calcium binding protein, plays a crucial role in calcium homeostasis. Calcium is by now recognized as an important second messenger in cancer progression and metastasis. Consequently, in this dissertation, the influence of NCS1 on the PI3K / AKT pathway, the direct interaction of NCS1 with the InsP3R and the function of NCS1 in the LIMK1- pathway were suggested as possible routes through which NCS1 might promote cancer progression.
In sum, the investigations are supportive of the hypothesis that NCS1 increases metastatic capabilities of triple-negative breast cancer in vivo. Clinical evidence, as well as multiple in vitro and in vivo experiments suggest adverse outcomes for cancer patients with high NCS1 levels. Therefore, NCS1 might qualify as a novel prognostic biomarker for triple-negative breast cancer.
Previously reported interactions between NCS1 and chemotherapeutic drugs emphasize the relevance to further evaluate possible therapeutic targeting of NCS1 in cancer. This could lead to the discovery of urgently needed, novel pharmacologic therapy schemes in the treatment of triple-negative breast cancer.},

url = {https://hdl.handle.net/20.500.11811/8666}
}

The following license files are associated with this item:

InCopyright