Network


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

Hotspot


Dive into the research topics where Xiayang Qiu is active.

Publication


Featured researches published by Xiayang Qiu.


Journal of Biological Chemistry | 2010

SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1

Michelle Pacholec; John E. Bleasdale; Boris A. Chrunyk; David Cunningham; Declan Flynn; Robert S. Garofalo; David A. Griffith; Matt Griffor; Pat Loulakis; Brandon Pabst; Xiayang Qiu; Brian J. Stockman; Venkataraman Thanabal; Alison H. Varghese; Jessica Ward; Jane M. Withka; Kay Ahn

Sirtuins catalyze NAD+-dependent protein deacetylation and are critical regulators of transcription, apoptosis, metabolism, and aging. There are seven human sirtuins (SIRT1–7), and SIRT1 has been implicated as a key mediator of the pathways downstream of calorie restriction that have been shown to delay the onset and reduce the incidence of age-related diseases such as type 2 diabetes. Increasing SIRT1 activity, either by transgenic overexpression of the Sirt1 gene in mice or by pharmacological activation by small molecule activators resveratrol and SRT1720, has shown beneficial effects in rodent models of type 2 diabetes, indicating that SIRT1 may represent an attractive therapeutic target. Herein, we have assessed purported SIRT1 activators by employing biochemical assays utilizing native substrates, including a p53-derived peptide substrate lacking a fluorophore as well as the purified native full-length protein substrates p53 and acetyl-CoA synthetase1. SRT1720, its structurally related compounds SRT2183 and SRT1460, and resveratrol do not lead to apparent activation of SIRT1 with native peptide or full-length protein substrates, whereas they do activate SIRT1 with peptide substrate containing a covalently attached fluorophore. Employing NMR, surface plasmon resonance, and isothermal calorimetry techniques, we provide evidence that these compounds directly interact with fluorophore-containing peptide substrates. Furthermore, we demonstrate that SRT1720 neither lowers plasma glucose nor improves mitochondrial capacity in mice fed a high fat diet. SRT1720, SRT2183, SRT1460, and resveratrol exhibit multiple off-target activities against receptors, enzymes, transporters, and ion channels. Taken together, we conclude that SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1.


Nature Structural & Molecular Biology | 2007

Structural and biophysical studies of PCSK9 and its mutants linked to familial hypercholesterolemia.

David Cunningham; Dennis E. Danley; Kieran F. Geoghegan; Matthew C. Griffor; Julie Hawkins; Timothy A. Subashi; Alison H. Varghese; Mark Ammirati; Jeffrey S. Culp; Lise R. Hoth; Mahmoud N. Mansour; Katherine M McGrath; Andrew P. Seddon; Shirish Shenolikar; Kim Jonelle Stutzman-Engwall; Laurie C. Warren; Donghui Xia; Xiayang Qiu

Proprotein convertase subtilisin kexin type 9 (PCSK9) lowers the abundance of surface low-density lipoprotein (LDL) receptor through an undefined mechanism. The structure of human PCSK9 shows the subtilisin-like catalytic site blocked by the prodomain in a noncovalent complex and inaccessible to exogenous ligands, and that the C-terminal domain has a novel fold. Biosensor studies show that PCSK9 binds the extracellular domain of LDL receptor with Kd = 170 nM at the neutral pH of plasma, but with a Kd as low as 1 nM at the acidic pH of endosomes. The D374Y gain-of-function mutant, associated with hypercholesterolemia and early-onset cardiovascular disease, binds the receptor 25 times more tightly than wild-type PCSK9 at neutral pH and remains exclusively in a high-affinity complex at the acidic pH. PCSK9 may diminish LDL receptors by a mechanism that requires direct binding but not necessarily receptor proteolysis.


Nature Structural & Molecular Biology | 2007

Crystal structure of cholesteryl ester transfer protein reveals a long tunnel and four bound lipid molecules

Xiayang Qiu; Anil Mistry; Mark Ammirati; Boris A. Chrunyk; Ronald W. Clark; Yang Cong; Jeffrey S. Culp; Dennis E. Danley; Thomas B. Freeman; Kieran F. Geoghegan; Matthew C. Griffor; Steven J. Hawrylik; Cheryl Myers Hayward; Preston Hensley; Lise R. Hoth; George A. Karam; Maruja E. Lira; David B. Lloyd; Katherine M McGrath; Kim Jonelle Stutzman-Engwall; Ann Subashi; Timothy A. Subashi; John F. Thompson; Ing-Kae Wang; Honglei Zhao; Andrew P. Seddon

Cholesteryl ester transfer protein (CETP) shuttles various lipids between lipoproteins, resulting in the net transfer of cholesteryl esters from atheroprotective, high-density lipoproteins (HDL) to atherogenic, lower-density species. Inhibition of CETP raises HDL cholesterol and may potentially be used to treat cardiovascular disease. Here we describe the structure of CETP at 2.2-Å resolution, revealing a 60-Å-long tunnel filled with two hydrophobic cholesteryl esters and plugged by an amphiphilic phosphatidylcholine at each end. The two tunnel openings are large enough to allow lipid access, which is aided by a flexible helix and possibly also by a mobile flap. The curvature of the concave surface of CETP matches the radius of curvature of HDL particles, and potential conformational changes may occur to accommodate larger lipoprotein particles. Point mutations blocking the middle of the tunnel abolish lipid-transfer activities, suggesting that neutral lipids pass through this continuous tunnel.


Antimicrobial Agents and Chemotherapy | 2002

Discovery of a Novel and Potent Class of FabI-Directed Antibacterial Agents

David J. Payne; William H. Miller; Valerie Berry; John Brosky; Walter J. Burgess; Emile Chen; Walter E. DeWolf; Andrew Fosberry; Rebecca Greenwood; Martha S. Head; Dirk A. Heerding; Cheryl A. Janson; Deborah Dee Jaworski; Paul M. Keller; Peter J. Manley; Terrance D. Moore; Kenneth A. Newlander; Stewart Pearson; Brian J. Polizzi; Xiayang Qiu; Stephen Rittenhouse; Courtney Slater-Radosti; Kevin L. Salyers; Mark A. Seefeld; Martin G. Smyth; Dennis T. Takata; Irene Nijole Uzinskas; Kalindi Vaidya; Nicola G. Wallis; Scott B. Winram

ABSTRACT Bacterial enoyl-acyl carrier protein (ACP) reductase (FabI) catalyzes the final step in each elongation cycle of bacterial fatty acid biosynthesis and is an attractive target for the development of new antibacterial agents. High-throughput screening of the Staphylococcus aureus FabI enzyme identified a novel, weak inhibitor with no detectable antibacterial activity against S. aureus. Iterative medicinal chemistry and X-ray crystal structure-based design led to the identification of compound 4 [(E)-N-methyl-N-(2-methyl-1H-indol-3-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)acrylamide], which is 350-fold more potent than the original lead compound obtained by high-throughput screening in the FabI inhibition assay. Compound 4 has exquisite antistaphylococci activity, achieving MICs at which 90% of isolates are inhibited more than 500 times lower than those of nine currently available antibiotics against a panel of multidrug-resistant strains of S. aureus and Staphylococcus epidermidis. Furthermore, compound 4 exhibits excellent in vivo efficacy in an S. aureus infection model in rats. Biochemical and genetic approaches have confirmed that the mode of antibacterial action of compound 4 and related compounds is via inhibition of FabI. Compound 4 also exhibits weak FabK inhibitory activity, which may explain its antibacterial activity against Streptococcus pneumoniae and Enterococcus faecalis, which depend on FabK and both FabK and FabI, respectively, for their enoyl-ACP reductase function. These results show that compound 4 is representative of a new, totally synthetic series of antibacterial agents that has the potential to provide novel alternatives for the treatment of S. aureus infections that are resistant to our present armory of antibiotics.


Journal of Biological Chemistry | 1999

Crystal structure of beta-ketoacyl-acyl carrier protein synthase III. A key condensing enzyme in bacterial fatty acid biosynthesis.

Xiayang Qiu; Cheryl A. Janson; Alex K. Konstantinidis; Silas Nwagwu; Carol Silverman; Ward W. Smith; Sanjay S. Khandekar; John T. Lonsdale; Sherin S. Abdel-Meguid

β-Ketoacyl-acyl carrier protein synthase III (FabH), the most divergent member of the family of condensing enzymes, is a key catalyst in bacterial fatty acid biosynthesis and a promising target for novel antibiotics. We report here the crystal structures of FabH determined in the presence and absence of acetyl-CoA. These structures display a fold that is common for condensing enzymes. The observed acetylation of Cys112 proves its catalytic role and clearly defines the primer binding pocket. Modeling based on a bound CoA molecule suggests catalytic roles for His244 and Asn274. The structures provide the molecular basis for FabH substrate specificity and reaction mechanism and are important for structure-based design of novel antibiotics.


Journal of Biological Chemistry | 2006

Crystal Structure of the Herpes Simplex Virus 1 DNA Polymerase

Shenping Liu; John D. Knafels; Jeanne S. Chang; Gregory A. Waszak; Eric T. Baldwin; Martin R. Deibel; Darrell R. Thomsen; Fred L. Homa; Peter A. Wells; Monica C. Tory; Roger A. Poorman; Hua Gao; Xiayang Qiu; Andrew P. Seddon

Herpesviruses are the second leading cause of human viral diseases. Herpes Simplex Virus types 1 and 2 and Varicella-zoster virus produce neurotropic infections such as cutaneous and genital herpes, chickenpox, and shingles. Infections of a lymphotropic nature are caused by cytomegalovirus, HSV-6, HSV-7, and Epstein-Barr virus producing lymphoma, carcinoma, and congenital abnormalities. Yet another series of serious health problems are posed by infections in immunocompromised individuals. Common therapies for herpes viral infections employ nucleoside analogs, such as Acyclovir, and target the viral DNA polymerase, essential for viral DNA replication. Although clinically useful, this class of drugs exhibits a narrow antiviral spectrum, and resistance to these agents is an emerging problem for disease management. A better understanding of herpes virus replication will help the development of new safe and effective broad spectrum anti-herpetic drugs that fill an unmet need. Here, we present the first crystal structure of a herpesvirus polymerase, the Herpes Simplex Virus type 1 DNA polymerase, at 2.7 Å resolution. The structural similarity of this polymerase to other α polymerases has allowed us to construct high confidence models of a replication complex of the polymerase and of Acyclovir as a DNA chain terminator. We propose a novel inhibition mechanism in which a representative of a series of non-nucleosidic viral polymerase inhibitors, the 4-oxo-dihydroquinolines, binds at the polymerase active site interacting non-covalently with both the polymerase and the DNA duplex.


Journal of Biological Chemistry | 2012

Crystal Structures of Cholesteryl Ester Transfer Protein in Complex with Inhibitors

Shenping Liu; Anil Mistry; Jennifer M. Reynolds; David B. Lloyd; Matthew C. Griffor; David Austen Perry; Roger Benjamin Ruggeri; Ronald W. Clark; Xiayang Qiu

Background: Human cholesteryl ester transfer protein (CETP) transfers cholesteryl esters from high-density to low-density lipoprotein particles. Results: Crystallographic, mutagenesis, and biochemical studies illuminated inhibition mechanisms of CETP by torcetrapib and a structurally distinct compound, ((2R)-3-{[4-(4-chloro-3-ethylphenoxy)pyrimidin-2-yl][3-(1,1,2,2-tetrafluoroethoxy)benzyl]amino}-1,1,1-trifluoropropan-2-ol. Conclusion: These small molecules inhibit CETP through blocking its lipid tunnel. Significance: Potential polar interactions at compound binding site may be utilized in design of inhibitors with improved physical properties. Human plasma cholesteryl ester transfer protein (CETP) transports cholesteryl ester from the antiatherogenic high-density lipoproteins (HDL) to the proatherogenic low-density and very low-density lipoproteins (LDL and VLDL). Inhibition of CETP has been shown to raise human plasma HDL cholesterol (HDL-C) levels and is potentially a novel approach for the prevention of cardiovascular diseases. Here, we report the crystal structures of CETP in complex with torcetrapib, a CETP inhibitor that has been tested in phase 3 clinical trials, and compound 2, an analog from a structurally distinct inhibitor series. In both crystal structures, the inhibitors are buried deeply within the protein, shifting the bound cholesteryl ester in the N-terminal pocket of the long hydrophobic tunnel and displacing the phospholipid from that pocket. The lipids in the C-terminal pocket of the hydrophobic tunnel remain unchanged. The inhibitors are positioned near the narrowing neck of the hydrophobic tunnel of CETP and thus block the connection between the N- and C-terminal pockets. These structures illuminate the unusual inhibition mechanism of these compounds and support the tunnel mechanism for neutral lipid transfer by CETP. These highly lipophilic inhibitors bind mainly through extensive hydrophobic interactions with the protein and the shifted cholesteryl ester molecule. However, polar residues, such as Ser-230 and His-232, are also found in the inhibitor binding site. An enhanced understanding of the inhibitor binding site may provide opportunities to design novel CETP inhibitors possessing more drug-like physical properties, distinct modes of action, or alternative pharmacological profiles.


MedChemComm | 2011

Designing glucokinase activators with reduced hypoglycemia risk: discovery of N,N-dimethyl-5-(2-methyl-6-((5-methylpyrazin-2-yl)-carbamoyl)benzofuran-4-yloxy)pyrimidine-2-carboxamide as a clinical candidate for the treatment of type 2 diabetes mellitus

Jeffrey A. Pfefferkorn; Angel Guzman-Perez; Peter J. Oates; John Litchfield; Gary E. Aspnes; Arindrajit Basak; John William Benbow; Martin A. Berliner; Jianwei Bian; Chulho Choi; Kevin Daniel Freeman-Cook; Jeffrey W. Corbett; Mary Theresa Didiuk; Joshua R. Dunetz; Kevin J. Filipski; William M. Hungerford; Christopher S. Jones; Kapil Karki; Anthony Lai Ling; Jian-Cheng Li; Leena Patel; Christian Perreault; Hud Risley; James Saenz; Wei Song; Meihua Tu; Robert J. Aiello; Karen Atkinson; Nicole Barucci; David A. Beebe

Glucokinase is a key regulator of glucose homeostasis and small molecule activators of this enzyme represent a promising opportunity for the treatment of Type 2 diabetes. Several glucokinase activators have advanced to clinical studies and demonstrated promising efficacy; however, many of these early candidates also revealed hypoglycemia as a key risk. In an effort to mitigate this hypoglycemia risk while maintaining the promising efficacy of this mechanism, we have investigated a series of substituted 2-methylbenzofurans as “partial activators” of the glucokinase enzyme leading to the identification of N,N-dimethyl-5-(2-methyl-6-((5-methylpyrazin-2-yl)-carbamoyl)benzofuran-4-yloxy)pyrimidine-2-carboxamide as an early development candidate.


Journal of Biological Chemistry | 2005

Cholesteryl ester transfer protein variants have differential stability but uniform inhibition by torcetrapib

David B. Lloyd; Maruja E. Lira; Linda S. Wood; L. Kathryn Durham; Thomas B. Freeman; Gregory M. Preston; Xiayang Qiu; Eliot Sugarman; Peter C. Bonnette; Anthony J. Lanzetti; Patrice M. Milos; John F. Thompson

Cholesteryl ester transfer protein (CETP) is an important modulator of high density lipoprotein cholesterol in humans and thus considered to be a therapeutic target for preventing cardiovascular disease. The gene encoding CETP has been shown to be highly variable, with multiple single nucleotide polymorphisms responsible for altering both its transcription and sequence. Examining nine missense variants of CETP, we found some had significant associations with CETP mass and high density lipoprotein cholesterol levels. Two variants, Pro-373 and Gln-451, appear to be more stable in vivo, an observation mirrored by partial proteolysis studies performed in vitro. Because these naturally occurring variant proteins are potentially present in clinical populations that will be treated with CETP inhibitors, all commonly occurring haplotypes were tested to determine whether the proteins they encode could be inhibited by torcetrapib, a compound currently in clinical trials in combination with atorvastatin. Torcetrapib behaved similarly with all variants, with no significant differences in inhibition.


Journal of Biological Chemistry | 2012

Insights into Mechanism of Glucokinase Activation: OBSERVATION OF MULTIPLE DISTINCT PROTEIN CONFORMATIONS.

Shenping Liu; Mark Ammirati; Xi Song; John D. Knafels; Jeff Zhang; Samantha E. Greasley; Jeffrey A. Pfefferkorn; Xiayang Qiu

Background: Human glucokinase (GK) is a principal regulator of glucose homeostasis. Results: The structure of the catalytic complex of GK has been determined, and multiple conformations have been directly observed in solution. Conclusion: In solution, glucose dose-dependently converts GK from the apo conformation to an active open conformation. Significance: Understanding the conformational changes of GK during the reaction is crucial for designing better glucokinase activators with safer profiles. Human glucokinase (GK) is a principal regulating sensor of plasma glucose levels. Mutations that inactivate GK are linked to diabetes, and mutations that activate it are associated with hypoglycemia. Unique kinetic properties equip GK for its regulatory role: although it has weak basal affinity for glucose, positive cooperativity in its binding of glucose causes a rapid increase in catalytic activity when plasma glucose concentrations rise above euglycemic levels. In clinical trials, small molecule GK activators (GKAs) have been efficacious in lowering plasma glucose and enhancing glucose-stimulated insulin secretion, but they carry a risk of overly activating GK and causing hypoglycemia. The theoretical models proposed to date attribute the positive cooperativity of GK to the existence of distinct protein conformations that interconvert slowly and exhibit different affinities for glucose. Here we report the respective crystal structures of the catalytic complex of GK and of a GK-glucose complex in a wide open conformation. To assess conformations of GK in solution, we also carried out small angle x-ray scattering experiments. The results showed that glucose dose-dependently converts GK from an apo conformation to an active open conformation. Compared with wild type GK, activating mutants required notably lower concentrations of glucose to be converted to the active open conformation. GKAs decreased the level of glucose required for GK activation, and different compounds demonstrated distinct activation profiles. These results lead us to propose a modified mnemonic model to explain cooperativity in GK. Our findings may offer new approaches for designing GKAs with reduced hypoglycemic risk.

Collaboration


Dive into the Xiayang Qiu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge