Network


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

Hotspot


Dive into the research topics where Genbin Shi is active.

Publication


Featured researches published by Genbin Shi.


Structure | 2000

Catalytic Center Assembly of Hppk as Revealed by the Crystal Structure of a Ternary Complex at 1.25 A Resolution

Jaroslaw Blaszczyk; Genbin Shi; Honggao Yan; Xinhua Ji

BACKGROUND Folates are essential for life. Unlike mammals, most microorganisms must synthesize folates de novo. 6-Hydroxymethyl-7, 8-dihydropterin pyrophosphokinase (HPPK) catalyzes pyrophosphoryl transfer from ATP to 6-hydroxymethyl-7,8-dihydropterin (HP), the first reaction in the folate pathway, and therefore is an ideal target for developing novel antimicrobial agents. HPPK from Escherichia coli is a 158-residue thermostable protein that provides a convenient model system for mechanistic studies. Crystal structures have been reported for HPPK without bound ligand, containing an HP analog, and complexed with an HP analog, two Mg(2+) ions, and ATP. RESULTS We present the 1.25 A crystal structure of HPPK in complex with HP, two Mg(2+) ions, and AMPCPP (an ATP analog that inhibits the enzymatic reaction). This structure demonstrates that the enzyme seals the active center where the reaction occurs. The comparison with unligated HPPK reveals dramatic conformational changes of three flexible loops and many sidechains. The coordination of Mg(2+) ions has been defined and the roles of 26 residues have been derived. CONCLUSIONS HPPK-HP-MgAMPCPP mimics most closely the natural ternary complex of HPPK and provides details of protein-substrate interactions. The coordination of the two Mg(2+) ions helps create the correct geometry for the one-step reaction of pyrophosphoryl transfer, for which we suggest an in-line single displacement mechanism with some associative character in the transition state. The rigidity of the adenine-binding pocket and hydrogen bonds are responsible for adenosine specificity. The nonconserved residues that interact with the substrate might be responsible for the species-dependent properties of an isozyme.


Structure | 1999

Crystal structure of 6-hydroxymethyl-7,8-dihydropterin pyrophosphokinase, a potential target for the development of novel antimicrobial agents

Bing Xiao; Genbin Shi; Xin Chen; Honggao Yan; Xinhua Ji

BACKGROUND Folate cofactors are essential for life. Mammals derive folates from their diet, whereas most microorganisms must synthesize folates de novo. Enzymes of the folate pathway therefore provide ideal targets for the development of antimicrobial agents. 6-Hydroxymethyl-7,8-dihydropterin pyrophosphokinase (HPPK) catalyzes the transfer of pyrophosphate from ATP to 6-hydroxymethyl-7,8-dihydropterin (HP), the first reaction in the folate biosynthetic pathway. RESULTS The crystal structure of HPPK from Escherichia coli has been determined at 1.5 A resolution with a crystallographic R factor of 0.182. The HPPK molecule has a novel three-layered alpha beta alpha fold that creates a valley approximately 26 A long, 10 A wide and 10 A deep. The active center of HPPK is located in the valley and the substrate-binding sites have been identified with the aid of NMR spectroscopy. The HP-binding site is located at one end of the valley, near Asn55, and is sandwiched between two aromatic sidechains. The ATP-binding site is located at the other end of the valley. The adenine base of ATP is positioned near Leu111 and the ribose and the triphosphate extend across and reach the vicinity of HP. CONCLUSIONS The HPPK structure provides a framework to elucidate structure/function relationships of the enzyme and to analyze mechanisms of pyrophosphoryl transfer. Furthermore, this work may prove useful in the structure-based design of new antimicrobial agents.


Bioorganic & Medicinal Chemistry | 2012

Bisubstrate analogue inhibitors of 6-hydroxymethyl-7,8-dihydropterin pyrophosphokinase: New design with improved properties

Genbin Shi; Gary Shaw; Yu He Liang; Priadarsini Subburaman; Yue Li; Yan Wu; Honggao Yan; Xinhua Ji

6-Hydroxymethyl-7,8-dihydropterin pyrophosphokinase (HPPK), a key enzyme in the folate biosynthetic pathway, catalyzes the pyrophosphoryl transfer from ATP to 6-hydroxymethyl-7,8-dihydropterin. The enzyme is essential for microorganisms, is absent from humans, and is not the target for any existing antibiotics. Therefore, HPPK is an attractive target for developing novel antimicrobial agents. Previously, we characterized the reaction trajectory of HPPK-catalyzed pyrophosphoryl transfer and synthesized a series of bisubstrate analog inhibitors of the enzyme by linking 6-hydroxymethylpterin to adenosine through 2, 3, or 4 phosphate groups. Here, we report a new generation of bisubstrate analog inhibitors. To improve protein binding and linker properties of such inhibitors, we have replaced the pterin moiety with 7,7-dimethyl-7,8-dihydropterin and the phosphate bridge with a piperidine linked thioether. We have synthesized the new inhibitors, measured their K(d) and IC(50) values, determined their crystal structures in complex with HPPK, and established their structure-activity relationship. 6-Carboxylic acid ethyl ester-7,7-dimethyl-7,8-dihydropterin, a novel intermediate that we developed recently for easy derivatization at position 6 of 7,7-dimethyl-7,8-dihydropterin, offers a much high yield for the synthesis of bisubstrate analogs than that of previously established procedure.


Journal of Molecular Graphics & Modelling | 2000

Structure and dynamics of 6-hydroxymethyl-7,8-dihydropterin pyrophosphokinase.

Honggao Yan; Jaroslaw Blaszczyk; Bing Xiao; Genbin Shi; Xinhua Ji

Folates are essential for life. Unlike mammals, most microorganisms must synthesize folates de novo. 6-Hydroxymethyl-7,8-dihydropterin pyrophosphokinase (HPPK) catalyzes pyrophosphoryl transfer from ATP to 6-hydroxymethyl-7,8-dihydropterin (HP), the first reaction in folate pathway, and therefore, is an ideal target for developing novel antimicrobial agents. Because of its small size and high thermal stability, E. coli HPPK is also an excellent model enzyme for studying the mechanisms of enzymatic pyrophosphoryl transfer. We have determined the crystal structures of HPPK in the unligated form and in complex with HP, two Mg2+ ions, and AMPCPP (an ATP analog that inhibits the enzymatic reaction). Comparison of the two crystal structures reveals dramatic conformational changes of three flexible loops and many side chains and possible roles of the active site residues.


FEBS Journal | 2014

Structural enzymology and inhibition of the bi-functional folate pathway enzyme HPPK-DHPS from the biowarfare agent Francisella tularensis.

Gary X. Shaw; Yue Li; Genbin Shi; Yan Wu; Scott Cherry; Danielle Needle; Di Zhang; Joseph E. Tropea; David S. Waugh; Honggao Yan; Xinhua Ji

Two valid targets for antibiotic development, 6‐hydroxymethyl‐7,8‐dihydropterin pyrophosphokinase (HPPK) and dihydropteroate synthase (DHPS), catalyze consecutive reactions in folate biosynthesis. In Francisella tularensis (Ft), these two activities are contained in a single protein, FtHPPK–DHPS. Although Pemble et al. (PLoS One 5, e14165) determined the structure of FtHPPK–DHPS, they were unable to measure the kinetic parameters of the enzyme. In this study, we elucidated the binding and inhibitory activities of two HPPK inhibitors (HP‐18 and HP‐26) against FtHPPK–DHPS, determined the structure of FtHPPK–DHPS in complex with HP‐26, and measured the kinetic parameters for the dual enzymatic activities of FtHPPK–DHPS. The biochemical analyses showed that HP‐18 and HP‐26 have significant isozyme selectivity, and that FtHPPK–DHPS is unique in that the catalytic efficiency of its DHPS activity is only 1/260,000 of that of Escherichia coli DHPS. Sequence and structural analyses suggest that HP‐26 is an excellent lead for developing therapeutic agents for tularemia, and that the very low DHPS activity is due, at least in part, to the lack of a key residue that interacts with the substrate p‐aminobenzoic acid (pABA). A BLAST search of the genomes of ten F. tularensis strains indicated that the bacterium contains a single FtHPPK–DHPS. The marginal DHPS activity and the single copy existence of FtHPPK–DHPS in F. tularensis make this bacterium more vulnerable to DHPS inhibitors. Current sulfa drugs are ineffective against tularemia; new inhibitors targeting the unique pABA‐binding pocket may be effective and less subject to resistance because any mutations introducing resistance may make the marginal DHPS activity unable to support the growth of F. tularensis.


Journal of Medicinal Chemistry | 2001

Bisubstrate Analogue Inhibitors of 6-Hydroxymethyl-7,8-dihydropterin Pyrophosphokinase: Synthesis and Biochemical and Crystallographic Studies†

Genbin Shi; Jaroslaw Blaszczyk; Xinhua Ji; Honggao Yan


Journal of Biological Chemistry | 2001

Unusual Conformational Changes in 6-Hydroxymethyl-7,8-dihydropterin Pyrophosphokinase as Revealed by X-ray Crystallography and NMR

Bing Xiao; Genbin Shi; Jinhai Gao; Jaroslaw Blaszczyk; Qin Liu; Xinhua Ji; Honggao Yan


Biochemistry | 2003

Dynamic roles of arginine residues 82 and 92 of Escherichia coli 6-hydroxymethyl-7,8-dihydropterin pyrophosphokinase: crystallographic studies.

Jaroslaw Blaszczyk; Yue Li; Genbin Shi; Honggao Yan; Xinhua Ji


Structure | 2004

REACTION TRAJECTORY OF PYROPHOSPHORYL TRANSFER CATALYZED BY 6-HYDROXYMETHYL-7,8-DIHYDROPTERIN PYROPHOSPHOKINASE

Jaroslaw Blaszczyk; Genbin Shi; Yue Li; Honggao Yan; Xinhua Ji


Biochemistry | 2002

Chemical transformation is not rate-limiting in the reaction catalyzed by Escherichia coli 6-hydroxymethyl-7,8-dihydropterin pyrophosphokinase.

Yue Li; Yunchen Gong; Genbin Shi; Jaroslaw Blaszczyk; Xinhua Ji; Honggao Yan

Collaboration


Dive into the Genbin Shi's collaboration.

Top Co-Authors

Avatar

Honggao Yan

Michigan State University

View shared research outputs
Top Co-Authors

Avatar

Xinhua Ji

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jaroslaw Blaszczyk

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yue Li

Michigan State University

View shared research outputs
Top Co-Authors

Avatar

Bing Xiao

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yan Wu

Michigan State University

View shared research outputs
Top Co-Authors

Avatar

Yunchen Gong

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gary Shaw

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge