Cargando…

BSCI-06. Phage display biopanning identifies Amot regulating cell motility in brain metastasis-initiating cells

OBJECTIVE: Metastatic brain tumors (MBTs) are the most common type of malignant brain tumors. Due to the deviation of MBTs from the parental tumors, the effective therapies for primary tumors often are not working in brain metastases. Even more new intracranial lesions were developed though the prim...

Descripción completa

Detalles Bibliográficos
Autores principales: She, Chunhua, Potez, Marine, Kim, JongMyung, Liu, James
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Oxford University Press 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8351267/
http://dx.doi.org/10.1093/noajnl/vdab071.005
_version_ 1783735937975451648
author She, Chunhua
Potez, Marine
Kim, JongMyung
Liu, James
author_facet She, Chunhua
Potez, Marine
Kim, JongMyung
Liu, James
author_sort She, Chunhua
collection PubMed
description OBJECTIVE: Metastatic brain tumors (MBTs) are the most common type of malignant brain tumors. Due to the deviation of MBTs from the parental tumors, the effective therapies for primary tumors often are not working in brain metastases. Even more new intracranial lesions were developed though the primary lesion was controlled. The occurrence of brain metastasis-initiating cells (BMICs) suggested the possibility of its spread intracranially. Here we aimed to explore the biological behavior in cell motility of BMICs and understand the potential mechanisms. METHODS: In vitro and in vivo phage display biopanning strategies were used to isolate dodecapeptides that specifically target BMICs by selecting against primary lung cancer cells and normal brain cells. In silico analysis was used to derive specific protein targets in BMICs. Potential targets were narrowed down through analysis in patient databases and verified for their presence in BMIC through RT-PCR. Cell migration and adhesion in BMICs were analyzed using Transwell, scratch, and adhesion assays. Protein expression and cell morphology were detected by immunofluorescence. Immune blot was performed to detect the epithelial-mesenchymal related molecules and explore protein-protein interactions. RESULTS: In silico analysis of BMICs specific peptides revealed Angiomotin (Amot) as a potential target in BMICs. Amot was found to be overexpressed in BMICs compared to primary lung cancer cells. Kaplan-Meier analysis demonstrated Amot was negatively correlated with overall survival among lung adenocarcinoma patients. Knockdown of AMOT in BMICs decreased the capability of cell migration and adhesion, through the downregulation of E-Cadherin. Amot was found to maintain the E-Cadherin in BMICs through reducing ubiquitination of E-Cadherin. Furthermore, the knockdown of E-Cadherin decreased cell migration and adhesion due to the decrease in cdc42 activity. CONCLUSIONS: Amot plays a role in promoting migration and adhesion in BMICs through preservation of E-Cadherin.
format Online
Article
Text
id pubmed-8351267
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Oxford University Press
record_format MEDLINE/PubMed
spelling pubmed-83512672021-08-09 BSCI-06. Phage display biopanning identifies Amot regulating cell motility in brain metastasis-initiating cells She, Chunhua Potez, Marine Kim, JongMyung Liu, James Neurooncol Adv Supplement Abstracts OBJECTIVE: Metastatic brain tumors (MBTs) are the most common type of malignant brain tumors. Due to the deviation of MBTs from the parental tumors, the effective therapies for primary tumors often are not working in brain metastases. Even more new intracranial lesions were developed though the primary lesion was controlled. The occurrence of brain metastasis-initiating cells (BMICs) suggested the possibility of its spread intracranially. Here we aimed to explore the biological behavior in cell motility of BMICs and understand the potential mechanisms. METHODS: In vitro and in vivo phage display biopanning strategies were used to isolate dodecapeptides that specifically target BMICs by selecting against primary lung cancer cells and normal brain cells. In silico analysis was used to derive specific protein targets in BMICs. Potential targets were narrowed down through analysis in patient databases and verified for their presence in BMIC through RT-PCR. Cell migration and adhesion in BMICs were analyzed using Transwell, scratch, and adhesion assays. Protein expression and cell morphology were detected by immunofluorescence. Immune blot was performed to detect the epithelial-mesenchymal related molecules and explore protein-protein interactions. RESULTS: In silico analysis of BMICs specific peptides revealed Angiomotin (Amot) as a potential target in BMICs. Amot was found to be overexpressed in BMICs compared to primary lung cancer cells. Kaplan-Meier analysis demonstrated Amot was negatively correlated with overall survival among lung adenocarcinoma patients. Knockdown of AMOT in BMICs decreased the capability of cell migration and adhesion, through the downregulation of E-Cadherin. Amot was found to maintain the E-Cadherin in BMICs through reducing ubiquitination of E-Cadherin. Furthermore, the knockdown of E-Cadherin decreased cell migration and adhesion due to the decrease in cdc42 activity. CONCLUSIONS: Amot plays a role in promoting migration and adhesion in BMICs through preservation of E-Cadherin. Oxford University Press 2021-08-09 /pmc/articles/PMC8351267/ http://dx.doi.org/10.1093/noajnl/vdab071.005 Text en © The Author(s) 2021. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology. https://creativecommons.org/licenses/by-nc/4.0/This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (http://creativecommons.org/licenses/by-nc/4.0/ (https://creativecommons.org/licenses/by-nc/4.0/) ), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com
spellingShingle Supplement Abstracts
She, Chunhua
Potez, Marine
Kim, JongMyung
Liu, James
BSCI-06. Phage display biopanning identifies Amot regulating cell motility in brain metastasis-initiating cells
title BSCI-06. Phage display biopanning identifies Amot regulating cell motility in brain metastasis-initiating cells
title_full BSCI-06. Phage display biopanning identifies Amot regulating cell motility in brain metastasis-initiating cells
title_fullStr BSCI-06. Phage display biopanning identifies Amot regulating cell motility in brain metastasis-initiating cells
title_full_unstemmed BSCI-06. Phage display biopanning identifies Amot regulating cell motility in brain metastasis-initiating cells
title_short BSCI-06. Phage display biopanning identifies Amot regulating cell motility in brain metastasis-initiating cells
title_sort bsci-06. phage display biopanning identifies amot regulating cell motility in brain metastasis-initiating cells
topic Supplement Abstracts
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8351267/
http://dx.doi.org/10.1093/noajnl/vdab071.005
work_keys_str_mv AT shechunhua bsci06phagedisplaybiopanningidentifiesamotregulatingcellmotilityinbrainmetastasisinitiatingcells
AT potezmarine bsci06phagedisplaybiopanningidentifiesamotregulatingcellmotilityinbrainmetastasisinitiatingcells
AT kimjongmyung bsci06phagedisplaybiopanningidentifiesamotregulatingcellmotilityinbrainmetastasisinitiatingcells
AT liujames bsci06phagedisplaybiopanningidentifiesamotregulatingcellmotilityinbrainmetastasisinitiatingcells