The gastric H+, K+ ATPase catalyzes the final step of gastric acid secretion thereby generating a proton gradient across the canalicular membrane of greater than a million fold. It has been a focus of this laboratory for 30 years. Our interest in recent years has been the structure-function relationships of this pump and the mechanism of inhibition of this pump by covalent inhibitors (the proton pump inhibitors, PPIs) and the K+ -competitive reversible inhibitors (the acid pump antagonists, APAs).In the proposed studies, we plan to continue our site-directed mutagenesis approach coupled with detailed homology analysis and modeling using the 4 available crystal structures of the SERCA Ca ATPase as a template to better define transport by the gastric acid pump. The Ca ATPase although only 29% homologous to the H+-K+-ATPase, has a very similar overall structure and also uses carboxylic acid clusters in the membrane domain as the ion transport- binding and export-import sites as does the Na, K+ ATPase that is 75% homologous to the H+,K+ ATPase. We plan to delineate pathways for transport of hydronium ion from cytoplasm to lumen and K+ from lumen to cytoplasm by analyzing various enzyme activities including phosphorylation and dephosphorylation of selected site mutants. We have developed a hypothesis that the unique Lys791 insertion into one cluster of acids (D814, E820, E795) allows release of proton at the required pH~1.0 and K+ binding to luminal carbonyl groups displaces two of the carboxylic acids bound to lys 791, thereby allowing return ofIys791 and replacement by K+ at this site. New mutants will further define the ion transport pathways of the H+,K+ ATPase by homology modeling. Since acid- pump antagonists are in final clinical trials, we plan to define their site of binding to the enzyme more precisely by synthesizing a new class of compounds and identifying the amino acid residues whose mutation alters the affinity or nature of inhibition by these new compounds as we have done for the now classical imidazo-1,2a prydine class that often show unexpected negative side effects. Since an important step of acid secretory regulation involves a morphological transformation of the parietal cell wherein the ATPase moves from a cytoplasmic membrane location to the microvilli of the secretory canaliculus, we will continue our study of trafficking and sorting of the stably expressed ? subunit of the enzyme in polarized gastric cells. In addition, we will study the distribution of a YFP- ? subunit knock in- construct in the mouse stomach, living mouse gastric glands and in other tissuess such as the kidney where the enzyme is expressed but function is unknown. The scaffold proteins interacting with the (3 subunit will be elucidated using the the split ubiquitin method which is capable of defining those proteins associated with a particular membrane inserted protein. These studies will aid in clarifying the role of translocation of the pump in regulation of acid secretion. The results of the proposed research will further improve the agents used for the treatment of acid-related diseases and also our knowledge of the ATPase and cellular events involved in regulation of its activity. ? ?

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK058333-08
Application #
7216934
Study Section
Clinical and Integrative Gastrointestinal Pathobiology Study Section (CIGP)
Program Officer
Hamilton, Frank A
Project Start
2000-09-15
Project End
2010-08-31
Budget Start
2006-09-01
Budget End
2007-08-31
Support Year
8
Fiscal Year
2006
Total Cost
$248,539
Indirect Cost
Name
University of California Los Angeles
Department
Physiology
Type
Schools of Medicine
DUNS #
092530369
City
Los Angeles
State
CA
Country
United States
Zip Code
90095
Sachs, G; Marcus, E A; Wen, Y et al. (2018) Editorial: control of acid secretion. Aliment Pharmacol Ther 48:682-683
Wen, Yi; Scott, David R; Vagin, Olga et al. (2018) Measurement of Internal pH in Helicobacter pylori by Using Green Fluorescent Protein Fluorimetry. J Bacteriol 200:
Wen, Yi; Feng, Jing; Sachs, George (2013) Helicobacter pylori 5'ureB-sRNA, a cis-encoded antisense small RNA, negatively regulates ureAB expression by transcription termination. J Bacteriol 195:444-52
Strugatsky, David; McNulty, Reginald; Munson, Keith et al. (2013) Structure of the proton-gated urea channel from the gastric pathogen Helicobacter pylori. Nature 493:255-8
Tokhtaeva, Elmira; Clifford, Rebecca J; Kaplan, Jack H et al. (2012) Subunit isoform selectivity in assembly of Na,K-ATPase ?-? heterodimers. J Biol Chem 287:26115-25
Tokhtaeva, Elmira; Sachs, George; Sun, Haiying et al. (2012) Identification of the amino acid region involved in the intercellular interaction between the *1 subunits of Na+/K+ -ATPase. J Cell Sci 125:1605-16
Tokhtaeva, Elmira; Sachs, George; Souda, Puneet et al. (2011) Epithelial junctions depend on intercellular trans-interactions between the Na,K-ATPase ?? subunits. J Biol Chem 286:25801-12
Goebel, M; Stengel, A; Lambrecht, N W G et al. (2011) Selective gene expression by rat gastric corpus epithelium. Physiol Genomics 43:237-54
Shin, Jai Moo; Munson, Keith; Sachs, George (2011) Gastric H+,K+-ATPase. Compr Physiol 1:2141-53
Shin, J M; Vagin, O; Munson, K et al. (2011) Erratum to: Molecular mechanisms in therapy of acid-related diseases. Cell Mol Life Sci 68:921

Showing the most recent 10 out of 43 publications