scholarly journals Identification of a Crucial Histidine Involved in Metal Transport Activity in the Arabidopsis Cation/H+ Exchanger CAX1

2005 ◽  
Vol 280 (34) ◽  
pp. 30136-30142 ◽  
Author(s):  
Toshiro Shigaki ◽  
Bronwyn J. Barkla ◽  
Maria Cristina Miranda-Vergara ◽  
Jian Zhao ◽  
Omar Pantoja ◽  
...  
2015 ◽  
Vol 34 (9) ◽  
pp. 1615-1628 ◽  
Author(s):  
Shuqin Xiang ◽  
Shanshan Feng ◽  
Yuxiu Zhang ◽  
Jinjuan Tan ◽  
Shuang Liang ◽  
...  

1999 ◽  
Vol 344 (1) ◽  
pp. 211-219 ◽  
Author(s):  
Mitsuaki TABUCHI ◽  
Tsutomu YOSHIDA ◽  
Kaoru TAKEGAWA ◽  
Fumio KISHI

The Bcg/Ity/Lsh locus in the mouse genome regulates macrophage activation for antimicrobial activity against intracellular pathogens, and the positional cloning of this locus identified the Nramp1 (natural resistance-associated macrophage protein) gene. Nramp2 was initially isolated as a homologue of Nramp1. Recently, the rat divalent metal transporter DMT1 was identified electrophysiologically, and was found to be an isoform of Nramp2, a mutation which was subsequently identified in rats suffering from hereditary iron-deficiency anaemia. Despite the 64% amino acid sequence identity of Nramp1 and Nramp2, no divalent metal transport activity has yet been detected from Nramp1, and the function of Nramp1 on the molecular level is still unclear. To investigate the divalent metal transport activity of NRAMP molecules, we constructed four chimeric NRAMP genes by swapping the domains of human NRAMP1 and NRAMP2 with each other. The functional characteristics of wild-type NRAMP1, NRAMP2 and their chimeras were determined by expression in the divalent metal transporter-disrupted strain of fission yeast, pdt1δ, and we analysed the divalent metal transport activity by complementation of the EGTA- and pH-sensitive phenotype of pdt1δ. Replacement of the N-terminal cytoplasmic domain of NRAMP2 with the NRAMP1 counterpart resulted in inactive chimeras, indicating that the functional difference between NRAMP1 and NRAMP2 is located in this region. However, results obtained with the reverse construct and other chimeras indicated that these regions are not solely responsible for the differences in EGTA- and pH-sensitivity of NRAMP1 and NRAMP2. These findings indicate that NRAMP1 itself cannot represent the divalent metal transport activity in S. pombe and the additional protein segments of the molecules located elsewhere in NRAMP1 are also functionally distinct from their NRAMP2 counterparts.


Diabetes ◽  
1992 ◽  
Vol 41 (5) ◽  
pp. 592-597 ◽  
Author(s):  
N. Inagaki ◽  
K. Yasuda ◽  
G. Inoue ◽  
Y. Okamoto ◽  
H. Yano ◽  
...  

2018 ◽  
Vol 14 (2) ◽  
pp. 149-152
Author(s):  
Jie Chen ◽  
Vai H. Fong ◽  
Amandio Vieira

2016 ◽  
Author(s):  
Alison E. O'Connor ◽  
◽  
Aaron J. Beck ◽  
Aaron J. Beck ◽  
Elizabeth A. Canuel ◽  
...  

Function ◽  
2021 ◽  
Author(s):  
Bruce R Stevens ◽  
J Clive Ellory ◽  
Robert L Preston

Abstract The SARS-CoV-2 receptor, Angiotensin Converting Enzyme-2 (ACE2), is expressed at levels of greatest magnitude in the small intestine as compared to all other human tissues. Enterocyte ACE2 is co-expressed as the apical membrane trafficking partner obligatory for expression and activity of the B0AT1 sodium-dependent neutral amino acid transporter. These components are assembled as an [ACE2: B0AT1]2 dimer-of-heterodimers quaternary complex that putatively steers SARS-CoV-2 tropism in the gastrointestinal (GI) tract. GI clinical symptomology is reported in about half of COVID-19 patients, and can be accompanied by gut shedding of virion particles. We hypothesized that within this 4-mer structural complex, each [ACE2: B0AT1] heterodimer pair constitutes a physiological “functional unit.” This was confirmed experimentally by employing purified lyophilized enterocyte brush border membrane vesicles that were exposed to increasing doses of high-energy electron radiation from a 16 MeV linear accelerator. Based on established target theory, the results indicated the presence of Na+-dependent neutral amino acid influx transport activity functional unit with target size mw = 183.7 ± 16.8 kDa in situ in intact apical membranes. Each thermodynamically stabilized [ACE2: B0AT1] heterodimer functional unit manifests the transport activity within the whole ∼345 kDa [ACE2: B0AT1]2 dimer-of-heterodimers quaternary structural complex. The results are consistent with our prior molecular docking modeling and gut-lung axis approaches to understanding COVID-19. These findings advance the understanding of the physiology of B0AT1 interaction with ACE2 in the gut, and thereby potentially contribute to translational developments designed to treat or mitigate COVID-19 variant outbreaks and/or GI symptom persistence in long-haul Post-Acute Sequelae of SARS-CoV-2 (PASC).


1992 ◽  
Vol 267 (24) ◽  
pp. 16951-16956
Author(s):  
D Vijayalakshmi ◽  
L Dagnino ◽  
J.A. Belt ◽  
W.P. Gati ◽  
C.E. Cass ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document