Cardiff University | Prifysgol Caerdydd ORCA
Online Research @ Cardiff 
WelshClear Cookie - decide language by browser settings

Detailed analyses of the crucial functions of Zn transporter proteins in alkaline phosphatase activation

Suzuki, Eisuke, Ogawa, Namino, Takeda, Taka-aki, Nishito, Yukina, Tanaka, Yu-ki, Fujiwara, Takashi, Matsunaga, Mayu, Ueda, Sachiko, Kubo, Naoya, Tsuji, Tokuji, Fukunaka, Ayako, Yamazaki, Tomohiro, Taylor, Kathryn M. ORCID: https://orcid.org/0000-0002-9576-9490, Ogra, Yasumitsu and Kambe, Taiho 2020. Detailed analyses of the crucial functions of Zn transporter proteins in alkaline phosphatase activation. Journal of Biological Chemistry 295 , pp. 5669-5684. 10.1074/jbc.RA120.012610

[thumbnail of Detailed analyses of the crucial functions of Zn transporter proteins in alkaline phosphatase activation.pdf]
Preview
PDF - Accepted Post-Print Version
Download (1MB) | Preview

Abstract

Numerous zinc ectoenzymes are metalated by zinc and activated in the compartments of the early secretory pathway before reaching their destination. Zn transporter (ZNT) proteins located in these compartments are essential for ectoenzyme activation. We have previously reported that ZNT proteins, specifically ZNT5–ZNT6 heterodimers and ZNT7 homodimers, play critical roles in the activation of zinc ectoenzymes such as alkaline phosphatases (ALPs) by mobilizing cytosolic zinc into these compartments. However, this process remains incompletely understood. Here, using genetically engineered chicken DT40 cells, we first determined that Zrt/Irt-like protein (ZIP) transporters that are localized to the compartments of the early secretory pathway play only a minor role in the ALP activation process. These transporters included ZIP7, ZIP9, and ZIP13, performing pivotal functions in maintaining cellular homeostasis by effluxing zinc out of the compartments, Next, using purified ALP proteins, we showed that zinc metalation on ALP produced in DT40 cells lacking ZNT5–ZNT6 heterodimers and ZNT7 homodimers is impaired. Lastly, by genetically disrupting both ZNT5 and ZNT7 in human HAP1 cells, we directly demonstrated that the tissue-nonspecific ALP (TNAP)-activating functions of both ZNT complexes are conserved in human cells. Furthermore, using mutant HAP1 cells, we uncovered a previously unrecognized and unique spatial regulation of ZNT5–ZNT6 heterodimer formation, wherein ZNT5 recruits ZNT6 to the Golgi apparatus to form the heterodimeric complex. These findings fill in major gaps in our understanding of the molecular mechanisms underlying zinc ectoenzyme activation in the compartments of the early secretory pathway.

Item Type: Article
Date Type: Publication
Status: Published
Schools: Pharmacy
Publisher: American Society for Biochemistry and Molecular Biology
ISSN: 0021-9258
Date of First Compliant Deposit: 27 March 2020
Date of Acceptance: 16 March 2020
Last Modified: 06 Nov 2023 21:14
URI: https://orca.cardiff.ac.uk/id/eprint/130613

Citation Data

Cited 16 times in Scopus. View in Scopus. Powered By Scopus® Data

Actions (repository staff only)

Edit Item Edit Item

Downloads

Downloads per month over past year

View more statistics