check
RGS7 is recurrently mutated in melanoma and promotes migration and invasion of human cancer cells | Biochemistry, Food Science and Nutrition

Publications by year

<embed>
Copy and paste this code to your website.

Publications by Authors

Recent Publications

More<embed>
Copy and paste this code to your website.

Contact Us

Head of Institute: Prof. Ido Braslavsky

Administrative manager: Rakefet Kalev

Office Address:
Institute of Biochemistry, Food Science and Nutrition,
Robert H. Smith Faculty of Agriculture, Food and Environment,
The Hebrew University of Jerusalem, 
Herzl 229, Rehovot 7610001, ISRAEL

Tel: +972 - (0)8-9489385
Fax: +972 - (0)8-9363208
Email Address: rakefetk@savion.huji.ac.il

RGS7 is recurrently mutated in melanoma and promotes migration and invasion of human cancer cells

Citation:

Qutob, N. ; Masuho, I. ; Alon, M. ; Emmanuel, R. ; Cohen, I. ; Di Pizio, A. ; Madore, J. ; Elkahloun, A. ; Ziv, T. ; Levy, R. ; et al. Rgs7 Is Recurrently Mutated In Melanoma And Promotes Migration And Invasion Of Human Cancer Cells. Scientific Reports 2018, 8, 653.

Date Published:

2018

Abstract:

Analysis of 501 melanoma exomes revealed RGS7, which encodes a GTPase-accelerating protein (GAP), to be a tumor-suppressor gene. RGS7 was mutated in 11% of melanomas and was found to harbor three recurrent mutations (p.R44C, p.E383K and p.R416Q). Structural modeling of the most common recurrent mutation of the three (p.R44C) predicted that it destabilizes the protein due to the loss of an H-bond and salt bridge network between the mutated position and the serine and aspartic acid residues at positions 58 as 61, respectively. We experimentally confirmed this prediction showing that the p.R44C mutant protein is indeed destabilized. We further show RGS7 p.R44C has weaker catalytic activity for its substrate Gαo, thus providing a dual mechanism for its loss of function. Both of these effects are expected to contribute to loss of function of RGS7 resulting in increased anchorage-independent growth, migration and invasion of melanoma cells. By mutating position 56 in the R44C mutant from valine to cysteine, thereby enabling the formation of a disulfide bridge between the two mutated positions, we slightly increased the catalytic activity and reinstated protein stability, leading to the rescue of RGS7′s function as a tumor suppressor. Our findings identify RGS7 as a novel melanoma driver and point to the clinical relevance of using strategies to stabilize the protein and, thereby, restore its function.

Publisher's Version

Last updated on 07/11/2019