Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jai Moo Shin is active.

Publication


Featured researches published by Jai Moo Shin.


Alimentary Pharmacology & Therapeutics | 2006

Review article: the clinical pharmacology of proton pump inhibitors

Gary S. Sachs; Jai Moo Shin; Colin W. Howden

Proton pump inhibitors inhibit the gastric H+/K+‐ATPase via covalent binding to cysteine residues of the proton pump. All proton pump inhibitors must undergo acid accumulation in the parietal cell through protonation, followed by activation mediated by a second protonation at the active secretory canaliculus of the parietal cell.


Journal of Neurogastroenterology and Motility | 2013

Pharmacokinetics and Pharmacodynamics of the Proton Pump Inhibitors

Jai Moo Shin; Nayoung Kim

Proton pump inhibitor (PPI) is a prodrug which is activated by acid. Activated PPI binds covalently to the gastric H+, K+-ATPase via disulfide bond. Cys813 is the primary site responsible for the inhibition of acid pump enzyme, where PPIs bind. Omeprazole was the first PPI introduced in market, followed by pantoprazole, lansoprazole and rabeprazole. Though these PPIs share the core structures benzimidazole and pyridine, their pharmacokinetics and pharmacodynamics are a little different. Several factors must be considered in understanding the pharmacodynamics of PPIs, including: accumulation of PPI in the parietal cell, the proportion of the pump enzyme located at the canaliculus, de novo synthesis of new pump enzyme, metabolism of PPI, amounts of covalent binding of PPI in the parietal cell, and the stability of PPI binding. PPIs have about 1hour of elimination half-life. Area under the plasmic concentration curve and the intragastric pH profile are very good indicators for evaluating PPI efficacy. Though CYP2C19 and CYP3A4 polymorphism are major components of PPI metabolism, the pharmacokinetics and pharmacodynamics of racemic mixture of PPIs depend on the CYP2C19 genotype status. S-omeprazole is relatively insensitive to CYP2C19, so better control of the intragastric pH is achieved. Similarly, R-lansoprazole was developed in order to increase the drug activity. Delayed-release formulation resulted in a longer duration of effective concentration of R-lansoprazole in blood, in addition to metabolic advantage. Thus, dexlansoprazole showed best control of the intragastric pH among the present PPIs. Overall, PPIs made significant progress in the management of acid-related diseases and improved health-related quality of life.


Journal of Clinical Gastroenterology | 2007

The Gastric H,K ATPase as a Drug Target: Past, Present, and Future

George Sachs; Jai Moo Shin; Olga Vagin; Nils Lambrecht; Iskandar Yakubov; Keith Munson

The recent progress in therapy if acid disease has relied heavily on the performance of drugs targeted against the H,K ATPase of the stomach and the H2 receptor antagonists. It has become apparent in the last decade that the proton pump is the target that has the likelihood of being the most sustainable area of therapeutic application in the regulation of acid suppression. The process of activation of acid secretion requires a change in location of the ATPase from cytoplasmic tubules into the microvilli of the secretory canaliculus of the parietal cell. Stimulation of the resting parietal cell, with involvement of F-actin and ezrin does not use significant numbers of SNARE proteins, because their message is depleted in the pure parietal cell transcriptome. The cell morphology and gene expression suggest a tubule fusion-eversion event. As the active H,K ATPase requires efflux of KCl for activity we have, using the transcriptome derived from 99% pure parietal cells and immunocytochemistry, provided evidence that the KCl pathway is mediated by a KCQ1/KCNE2 complex for supplying K+ and CLIC6 for supplying the accompanying Cl−. The pump has been modeled on the basis of the structures of different conformations of the sr Ca ATPase related to the catalytic cycle. These models use the effects of site directed mutations and identification of the binding domain of the K competitive acid pump antagonists or the defined site of binding for the covalent class of proton pump inhibitors. The pump undergoes conformational changes associated with phosphorylation to allow the ion binding site to change exposure from cytoplasmic to luminal exposure. We have been able to postulate that the very low gastric pH is achieved by lysine 791 motion extruding the hydronium ion bound to carboxylates in the middle of the membrane domain. These models also allow description of the K+ entry to form the K+ liganded form of the enzyme and the reformation of the ion site inward conformation thus relating the catalytic cycle of the pump to conformational models. The mechanism of action of the proton pump inhibitor class of drug is discussed along with the cysteines covalently bound with these inhibitors. The review concludes with a discussion of the mechanism of action and binding regions of a possible new class of drug for acid control, the K+ competitive acid pump antagonists.


Biochimica et Biophysica Acta | 1993

The site of action of pantoprazole in the gastric H+/K+-ATPase

Jai Moo Shin; Marie Besancon; Alexander Simon; George Sachs

Pantoprazole is a pyridinyl-2-methylenesulfinyl-2-benzimidazole derivative. This compound inhibits the vesicular gastric H+/K(+)-ATPase (cytoplasmic side out) under acid transporting conditions by accumulating in the acid space generated by the pump. Pantoprazole is then converted in an acid-catalysed reaction to a cationic sulfenamide and reacts with cysteines available in or from the acidic extracytoplasmic space. This compound binds to the hog gastric H+/K(+)-ATPase with a stoichiometry of 3 nmol per mg protein, resulting in 94% inhibition of ATPase activity. Tryptic cleavage of the intact vesicles which had been reacted with [14C]pantoprazole at a 1 to 4 trypsin to protein ratio removed most of the cytoplasmic domain leaving the pairs of membrane spanning segments and their connecting extracytoplasmic loops intact. The peptides remaining in the membrane were dissolved in SDS and available cysteine residues labelled with fluorescein-5-maleimide. The peptides were separated on Tricine gradient gels, transferred to PVDF membranes and identified by fluorescence and radioactivity. From N-terminal sequence, fluorescence and molecular mass, it is concluded that pantoprazole is able to label both Cys-813 and Cys-822. These cysteines are predicted to be located in the extracytoplasmic loop connecting membrane segments 5 and 6 and in membrane segment 6. The major cytoplasmic tryptic cleavage site at this location moved from position 776 in unmodified enzyme to positions 784 and 792 following pantoprazole labelling, showing that the configuration of this region changed with pantoprazole labelling. A similar result was obtained by reduction of the enzyme with dithiothreitol. Covalent binding of the cationic sulfenamide to this region of the enzyme is able to block the conformation necessary for phosphorylation of the enzyme by ATP, accounting for its inhibitory effect on acid secretion.


Journal of Bacteriology | 2002

Proteins Released by Helicobacter pylori In Vitro

Nayoung Kim; David L. Weeks; Jai Moo Shin; David R. Scott; Mary K. Young; George Sachs

Secretion of proteins by Helicobacter pylori may contribute to gastric inflammation and epithelial damage. An in vitro analysis was designed to identify proteins released by mechanisms other than nonspecific lysis. The radioactivity of proteins in the supernatant was compared with that of the intact organism by two-dimensional gel phosphorimaging following a 4-h pulse-chase. The ratio of the amount of UreB, a known cytoplasmic protein, in the supernatant to that in the pellet was found to be 0.25, and this was taken as an index of lysis during the experiments (n = 6). Ratios greater than that of UreB were used to distinguish proteins that were selectively released into the medium. Thus, proteins enriched more than 10-fold in the supernatant compared to UreB were identified by mass spectrometry. Sixteen such proteins were present in the supernatant: VacA; a conserved secreted protein (HP1286); putative peptidyl cis-trans isomerase (HP0175); six proteins encoded by HP0305, HP0231, HP0973, HP0721, HP0129, and HP0902; thioredoxin (HP1458); single-stranded-DNA-binding 12RNP2 precursor (HP0827); histone-like DNA-binding protein HU (HP0835); ribosomal protein L11 (HP1202); a putative outer membrane protein (HP1564); and outer membrane proteins Omp21 (HP0913) and Omp20 (HP0912). All except HP0902, thioredoxin, HP0827, HP0835, and HP1202 had a signal peptide. When nalidixic acid, a DNA synthesis inhibitor, was added to inhibit cell division but not protein synthesis, to decrease possible contamination due to outer membrane shedding, two outer membrane proteins (Omp21 and Omp20) disappeared from the supernatant, and the amount of VacA also decreased. Thus, 13 proteins were still enriched greater than 10-fold in the medium after nalidixic acid treatment, suggesting these were released specifically, possibly by secretion. These proteins may be implicated in H. pylori-induced effects on the gastric epithelium.


Journal of Pharmacology and Experimental Therapeutics | 2011

Characterization of a novel potassium-competitive acid blocker of the gastric H,K-ATPase, 1-[5-(2-fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl]-N-methylmethanamine monofumarate (TAK-438).

Jai Moo Shin; Nobuhiro Inatomi; Keith Munson; David Strugatsky; Elmira Tokhtaeva; Olga Vagin; George Sachs

Inhibition of the gastric H,K-ATPase by the potassium-competitive acid blocker (P-CAB) 1-[5-(2-fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl]-N-methylmethanamine (TAK-438), is strictly K+-competitive with a Ki of 10 nM at pH 7. In contrast to previous P-CABs, this structure has a point positive charge (pKa 9.06) allowing for greater accumulation in parietal cells compared with previous P-CABs [e.g., (8-benzyloxy-2-methyl-imidazo(1,2-a)pyridin-3-yl)acetonitrile (SCH28080), pKa 5.6]. The dissociation rate of the compound from the isolated ATPase is slower than other P-CABs, with the t1/2 being 7.5 h in 20 mM KCl at pH 7. The stoichiometry of binding of TAK-438 to the H,K-ATPase is 2.2 nmol/mg in the presence of Mg-ATP, vanadate, or MgPi. However, TAK-438 also binds enzyme at 1.3 nmol/mg in the absence of Mg2+. Modeling of the H,K-ATPase to the homologous Na,K-ATPase predicts a close approach and hydrogen bonding between the positively charged N-methylamino group and the negatively charged Glu795 in the K+-binding site in contrast to the planar diffuse positive charge of previous P-CABs. This probably accounts for the slow dissociation and high affinity. The model also predicts hydrogen bonding between the hydroxyl of Tyr799 and the oxygens of the sulfonyl group of TAK-438. A Tyr799Phe mutation resulted in a 3-fold increase of the dissociation rate, showing that this hydrogen bonding also contributes to the slow dissociation rate. Hence, this K+-competitive inhibitor of the gastric H,K-ATPase should provide longer-lasting inhibition of gastric acid secretion compared with previous drugs of this class.


Alimentary Pharmacology & Therapeutics | 2000

The control of gastric acid and Helicobacter pylori eradication

Gary S. Sachs; Jai Moo Shin; Keith Munson; Olga Vagin; Nils Lambrecht; David R. Scott; David L. Weeks; K. Melchers

This review focuses on the gastric acid pump as a therapeutic target for the control of acid secretion in peptic ulcer and gastro‐oesophageal reflux disease. The mechanism of the proton pump inhibitors is discussed as well as their clinical use. The biology of Helicobacter pylori as a gastric denizen is then discussed, with special regard to its mechanisms of acid resistance. Here the properties of the products of the urease gene clusters, ureA, B and ureI, E, F, G and H are explored in order to explain the unique location of this pathogen. The dominant requirement for acid resistance is the presence of a proton gated urea transporter, UreI, which increases access of gastric juice urea to the intrabacterial urease 300‐fold. This enables rapid and continuous buffering of the bacterial periplasm to ≈ pH 6.0, allowing acid resistance and growth at acidic pH in the presence of 1 m M urea. A hypothesis for the basis of combination therapy for eradication is also presented.


Alimentary Pharmacology & Therapeutics | 2008

Predictable prolonged suppression of gastric acidity with a novel proton pump inhibitor, AGN 201904-Z

Richard H. Hunt; David Armstrong; Mohammad Yaghoobi; Cindy James; Ying Chen; J. Leonard; Jai Moo Shin; E. Lee; D. Tang-Liu; Gary S. Sachs

Background  AGN 201904‐Z is a new, slowly absorbed, acid‐stable pro‐proton pump inhibitor (pro‐PPI) rapidly converted to omeprazole in the systemic circulation giving a prolonged residence time.


Current Gastroenterology Reports | 2010

Novel Approaches to Inhibition of Gastric Acid Secretion

George Sachs; Jai Moo Shin; Richard H. Hunt

The gastric H,K-adenosine triphosphatase (ATPase) is the primary target for treatment of acid-related diseases. Proton pump inhibitors (PPIs) are weak bases composed of two moieties, a substituted pyridine with a primary pKa of about 4.0 that allows selective accumulation in the secretory canaliculus of the parietal cell, and a benzimidazole with a second pKa of about 1.0. Protonation of this benzimidazole activates these prodrugs, converting them to sulfenic acids and/or sulfenamides that react covalently with one or more cysteines accessible from the luminal surface of the ATPase. The maximal pharmacodynamic effect of PPIs as a group relies on cyclic adenosine monophosphate–driven H,K-ATPase translocation from the cytoplasm to the canalicular membrane of the parietal cell. At present, this effect can only be achieved with protein meal stimulation. Because of covalent binding, inhibitory effects last much longer than their plasma half-life. However, the short dwell-time of the drug in the blood and the requirement for acid activation impair their efficacy in acid suppression, particularly at night. All PPIs give excellent healing of peptic ulcer and produce good, but less than satisfactory, results in reflux esophagitis. PPIs combined with antibiotics eradicate Helicobacter pylori, but success has fallen to less than 80%. Longer dwell-time PPIs promise to improve acid suppression and hence clinical outcome. Potassium-competitive acid blockers (P-CABs) are another class of ATPase inhibitors, and at least one is in development. The P-CAB under development has a long duration of action even though its binding is not covalent. PPIs with a longer dwell time or P-CABs with long duration promise to address unmet clinical needs arising from an inability to inhibit nighttime acid secretion, with continued symptoms, delayed healing, and growth suppression of H. pylori reducing susceptibility to clarithromycin and amoxicillin. Thus, novel and more effective suppression of acid secretion would benefit those who suffer from acid-related morbidity, continuing esophageal damage and pain, nonsteroidal anti-inflammatory drug–induced ulcers, and nonresponders to H. pylori eradication.


Cellular and Molecular Life Sciences | 2008

Molecular mechanisms in therapy of acid-related diseases

Jai Moo Shin; Olga Vagin; Keith Munson; M. Kidd; I. M. Modlin; Gary S. Sachs

Abstract.Inhibition of gastric acid secretion is the mainstay of the treatment of gastroesophageal reflux disease and peptic ulceration; therapies to inhibit acid are among the best-selling drugs worldwide. Highly effective agents targeting the histamine H2 receptor were first identified in the 1970s. These were followed by the development of irreversible inhibitors of the parietal cell hydrogen-potassium ATPase (the proton pump inhibitors) that inhibit acid secretion much more effectively. Reviewed here are the chemistry, biological targets and pharmacology of these drugs, with reference to their current and evolving clinical utilities. Future directions in the development of acid inhibitory drugs include modifications of current agents and the emergence of a novel class of agents, the acid pump antagonists.

Collaboration


Dive into the Jai Moo Shin's collaboration.

Top Co-Authors

Avatar

George Sachs

University of California

View shared research outputs
Top Co-Authors

Avatar

Keith Munson

University of California

View shared research outputs
Top Co-Authors

Avatar

Olga Vagin

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marie Besancon

University of California

View shared research outputs
Top Co-Authors

Avatar

David R. Scott

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nils Lambrecht

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nayoung Kim

Seoul National University Bundang Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge