Network


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

Hotspot


Dive into the research topics where Wenkang Huang is active.

Publication


Featured researches published by Wenkang Huang.


Bioinformatics | 2013

Allosite: a method for predicting allosteric sites

Wenkang Huang; Shaoyong Lu; Zhimin Huang; Xinyi Liu; Linkai Mou; Yu Luo; Yanlong Zhao; Yaqin Liu; Zhongjie Chen; Tingjun Hou; Jian Zhang

MOTIVATION The use of allosteric modulators as preferred therapeutic agents against classic orthosteric ligands has colossal advantages, including higher specificity, fewer side effects and lower toxicity. Therefore, the computational prediction of allosteric sites in proteins is receiving increased attention in the field of drug discovery. Allosite is a newly developed automatic tool for the prediction of allosteric sites in proteins of interest and is now available through a web server. AVAILABILITY The Allosite server and tutorials are freely available at http://mdl.shsmu.edu.cn/AST CONTACT: [email protected] SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.


Drug Discovery Today | 2014

Recent computational advances in the identification of allosteric sites in proteins.

Shaoyong Lu; Wenkang Huang; Jian Zhang

Allosteric modulators have the potential to fine-tune protein functional activity. Therefore, the targeting of allosteric sites, as a strategy in drug design, is gaining increasing attention. Currently, it is not trivial to find and characterize new allosteric sites by experimental approaches. Alternatively, computational approaches are useful in helping researchers analyze and select potential allosteric sites for drug discovery. Here, we review state-of-the-art computational approaches directed at predicting putative allosteric sites in proteins, along with examples of successes in identifying allosteric sites utilizing these methods. We also discuss the challenges in developing reliable methods for predicting allosteric sites and tactics to resolve demanding tasks.


Nucleic Acids Research | 2016

ASD v3.0: unraveling allosteric regulation with structural mechanisms and biological networks

Qiancheng Shen; Guanqiao Wang; Shuai Li; Xinyi Liu; Shaoyong Lu; Zhongjie Chen; Kun Song; Junhao Yan; Lv Geng; Zhimin Huang; Wenkang Huang; Guo-Qiang Chen; Jian Zhang

Allosteric regulation, the most direct and efficient way of regulating protein function, is induced by the binding of a ligand at one site that is topographically distinct from an orthosteric site. Allosteric Database (ASD, available online at http://mdl.shsmu.edu.cn/ASD) has been developed to provide comprehensive information featuring allosteric regulation. With increasing data, fundamental questions pertaining to allostery are currently receiving more attention from the mechanism of allosteric changes in an individual protein to the entire effect of the changes in the interconnected network in the cell. Thus, the following novel features were added to this updated version: (i) structural mechanisms of more than 1600 allosteric actions were elucidated by a comparison of site structures before and after the binding of an modulator; (ii) 261 allosteric networks were identified to unveil how the allosteric action in a single protein would propagate to affect downstream proteins; (iii) two of the largest human allosteromes, protein kinases and GPCRs, were thoroughly constructed; and (iv) web interface and data organization were completely redesigned for efficient access. In addition, allosteric data have largely expanded in this update. These updates are useful for facilitating the investigation of allosteric mechanisms, dynamic networks and drug discoveries.


PLOS Computational Biology | 2014

The structural basis of ATP as an allosteric modulator.

Shaoyong Lu; Wenkang Huang; Qi Wang; Qiancheng Shen; Shuai Li; Ruth Nussinov; Jian Zhang

Adenosine-5’-triphosphate (ATP) is generally regarded as a substrate for energy currency and protein modification. Recent findings uncovered the allosteric function of ATP in cellular signal transduction but little is understood about this critical behavior of ATP. Through extensive analysis of ATP in solution and proteins, we found that the free ATP can exist in the compact and extended conformations in solution, and the two different conformational characteristics may be responsible for ATP to exert distinct biological functions: ATP molecules adopt both compact and extended conformations in the allosteric binding sites but conserve extended conformations in the substrate binding sites. Nudged elastic band simulations unveiled the distinct dynamic processes of ATP binding to the corresponding allosteric and substrate binding sites of uridine monophosphate kinase, and suggested that in solution ATP preferentially binds to the substrate binding sites of proteins. When the ATP molecules occupy the allosteric binding sites, the allosteric trigger from ATP to fuel allosteric communication between allosteric and functional sites is stemmed mainly from the triphosphate part of ATP, with a small number from the adenine part of ATP. Taken together, our results provide overall understanding of ATP allosteric functions responsible for regulation in biological systems.


International Immunopharmacology | 2012

Total glucosides of paeony attenuated functional maturation of dendritic cells via blocking TLR4/5 signaling in vivo.

Zhou Zhou; Jinpiao Lin; Rongfen Huo; Wenkang Huang; Jian Zhang; Li Wang; Yue Sun; Baihua Shen; Ningli Li

It is well known that dendritic cells (DCs) play a critical role in the initiation and development of an immune response. Inhibitory effect on DC maturation alters immune-mediated inflammatory reaction in vivo. Total glucosides of paeony (TGP) are active compounds extracted from the roots of Paeonia lactiflora and have been widely used to ameliorate inflammation in therapy for autoimmune diseases. However, whether TGP act on DC maturation remains unknown. In this study, we investigated the effect of TGP on DC maturation in ovalbumin (OVA) immunized mice. Ear inflammation was inhibited by TGP (150 mgkg(-1), i.p.×11 days) obviously. The antigen presenting capacity of DC derived from TGP-treated mice was arrested. Meanwhile, OVA specific T cell proliferation was inhibited. In addition, we found that maturation of DCs was decreased by TGP treatment. Furthermore, OVA specific T cell proliferation was rescued by the adoptive transfer of mature DCs (mDCs) into TGP treated OVA-challenged mice. The research on the mechanism showed that TGP significantly inhibited activation of TLR4/5 singling. All these results demonstrated that TGP inhibited DC maturation and function by selectively blocking TLR4/5 activation in vivo, which in turn leads to reduce immune-mediated inflammation in vivo, adding a novel mechanism and therapeutic target of TGP for inflammatory and autoimmune disease treatment.


Structure | 2015

The Mechanism of ATP-Dependent Allosteric Protection of Akt Kinase Phosphorylation

Shaoyong Lu; Rong Deng; Haiming Jiang; Huili Song; Shuai Li; Qiancheng Shen; Wenkang Huang; Ruth Nussinov; Jianxiu Yu; Jian Zhang

Kinases use ATP to phosphorylate substrates; recent findings underscore the additional regulatory roles of ATP. Here, we propose a mechanism for allosteric regulation of Akt1 kinase phosphorylation by ATP. Our 4.7-μs molecular dynamics simulations of Akt1 and its mutants in the ATP/ADP bound/unbound states revealed that ATP occupancy of the ATP-binding site stabilizes the closed conformation, allosterically protecting pT308 by restraining phosphatase access and key interconnected residues on the ATP→pT308 allosteric pathway. Following ATP→ADP hydrolysis, pT308 is exposed and readily dephosphorylated. Site-directed mutagenesis validated these predictions and indicated that the mutations do not impair PDK1 and PP2A phosphatase recruitment. We further probed the function of residues around pT308 at the atomic level, and predicted and experimentally confirmed that Akt1(H194R/R273H) double mutant rescues pathology-related Akt1(R273H). Analysis of classical Akt homologs suggests that this mechanism can provide a general model of allosteric kinase regulation by ATP; as such, it offers a potential avenue for allosteric drug discovery.


Journal of Chemical Information and Modeling | 2012

Insights into the role of magnesium triad in myo-inositol monophosphatase: metal mechanism, substrate binding, and lithium therapy.

Shaoyong Lu; Wenkang Huang; Xiaobai Li; Zhimin Huang; Xinyi Liu; Yingyi Chen; Ting Shi; Jian Zhang

myo-Inositol monophosphatase (IMPase) plays a pivotal role in the intracellular phosphatidylinositol cell signaling pathway. It has attracted considerable attention as a putative therapeutic target for lithium therapy in the treatment of bipolar disorder. A trio of activated cofactor Mg²⁺ ions is required for inositol monophosphate hydrolysis by IMPase. In the present study, computational studies, including two-layered ONIOM-based quantum mechanics/mechanical mechanics (QM/MM) calculations, molecular modeling, and molecular dynamics (MD) simulations, were performed to ascertain the role of the Mg²⁺ triad in the IMPase active site. The QM/MM calculations show that the structural identity of the nucleophilic water molecule W1 shared by Mg²⁺-1 and Mg²⁺-3, activated by Thr95/Asp47 dyad, is a hydroxide ion. Moreover, Mg²⁺-3 needs to be conjugated with Mg²⁺-1 in the binding site to create the activated nucleophilic hydroxide ion in accordance with the three-metal ion catalytic mechanism. The MD simulation of the IMPase-substrate-Mg²⁺ complex shows that the three Mg²⁺ ions promote substrate binding and help fix the phosphate moiety of the substrate for nucleophilic attack by the hydroxide ion. When Mg²⁺-2 is displaced with Li⁺, the MD simulations of the postreaction complex indicate that the conformation of the catalytic loop (residues 33 to 44) is disrupted and water molecule W2 does not coordinate with Li⁺. This disruption traps the inorganic phosphate and inositolate in the active site, which lead to IMPase inhibition. By contrast, in the native Mg²⁺ system, the W2 ligated by Mg²⁺-2 and Asp200 aids in protonation of the leaving inositolate moiety.


Proteins | 2013

How calcium inhibits the magnesium‐dependent kinase gsk3β: A molecular simulation study

Shaoyong Lu; Zhimin Huang; Wenkang Huang; Xinyi Liu; Yingyi Chen; Ting Shi; Jian Zhang

Glycogen synthase kinase 3β (GSK3β) is a ubiquitous serine/threonine kinase that plays a pivotal role in many biological processes. GSK3β catalyzes the transfer of γ‐phosphate of ATP to the unique substrate Ser/Thr residues with the assistance of two natural activating cofactors Mg2+. Interestingly, the biological observation reveals that a non‐native Ca2+ ion can inhibit the GSK3β catalytic activity. Here, the inhibitory mechanism of GSK3β by the displacement of native Mg2+ at site 1 by Ca2+ was investigated by means of 80 ns comparative molecular dynamics (MD) simulations of the GSK3β···Mg2+‐2/ATP/ Mg2+‐1 and GSK3β···Mg2+‐2/ATP/Ca2+‐1 systems. MD simulation results revealed that using the AMBER point charge model force field for Mg2+ was more appropriate in the reproduction of the active site architectural characteristics of GSK3β than using the magnesium‐cationic dummy atom model force field. Compared with the native Mg2+ bound system, the misalignment of the critical triphosphate moiety of ATP, the erroneous coordination environments around the Mg2+ ion at site 2, and the rupture of the key hydrogen bond between the invariant Lys85 and the ATP Oβ2 atom in the Ca2+ substituted system were observed in the MD simulation due to the Ca2+ ion in active site in order to achieve its preferred sevenfold coordination geometry, which adequately abolish the enzymatic activity. The obtained results are valuable in understanding the possible mechanism by why Ca2+ inhibits the GSK3β activity and also provide insights into the mechanism of Ca2+ inhibition in other structurally related protein kinases. Proteins 2013.


PLOS ONE | 2014

The Mechanism of Allosteric Inhibition of Protein Tyrosine Phosphatase 1B

Shuai Li; Jingmiao Zhang; Shaoyong Lu; Wenkang Huang; Lv Geng; Qiancheng Shen; Jian Zhang

As the prototypical member of the PTP family, protein tyrosine phosphatase 1B (PTP1B) is an attractive target for therapeutic interventions in type 2 diabetes. The extremely conserved catalytic site of PTP1B renders the design of selective PTP1B inhibitors intractable. Although discovered allosteric inhibitors containing a benzofuran sulfonamide scaffold offer fascinating opportunities to overcome selectivity issues, the allosteric inhibitory mechanism of PTP1B has remained elusive. Here, molecular dynamics (MD) simulations, coupled with a dynamic weighted community analysis, were performed to unveil the potential allosteric signal propagation pathway from the allosteric site to the catalytic site in PTP1B. This result revealed that the allosteric inhibitor compound-3 induces a conformational rearrangement in helix α7, disrupting the triangular interaction among helix α7, helix α3, and loop11. Helix α7 then produces a force, pulling helix α3 outward, and promotes Ser190 to interact with Tyr176. As a result, the deviation of Tyr176 abrogates the hydrophobic interactions with Trp179 and leads to the downward movement of the WPD loop, which forms an H-bond between Asp181 and Glu115. The formation of this H-bond constrains the WPD loop to its open conformation and thus inactivates PTP1B. The discovery of this allosteric mechanism provides an overall view of the regulation of PTP1B, which is an important insight for the design of potent allosteric PTP1B inhibitors.


Chemical Biology & Drug Design | 2014

Identification of novel compounds for human bitter taste receptors.

Mingfei Ji; Xubo Su; Xiaohong Su; Yingyi Chen; Wenkang Huang; Jian Zhang; Zhaobin Gao; Chuangang Li; Xuefeng Lu

The finely tuned bitter taste sensing in humans is orchestrated by a group of 25 bitter taste receptors (TAS2Rs), which belong to the G‐protein‐coupled receptor superfamily. TAS2Rs are expressed in the specialized taste bud cells of the gustatory system and perceive a plethora of bitter substances with versatile structures. To date, more than one hundred bitter ligands have been matched with their cognate receptors, but the understanding of the molecular mechanisms of TAS2Rs remains limited. Additionally, the extraoral expression of TAS2R genes was found in the gastrointestinal tract and respiratory system, which suggests other important physiological functions for TAS2Rs. To gain insight into the physiological functions of TAS2Rs, we established a heterologous expression system and characterized the response of 24 TAS2Rs against a library of potential bitter compounds. Among these bitter compounds of interest, 18 bitter compounds activated 16 TAS2Rs, representing 42 tastant–receptor pairs. We then calculated 14 descriptor properties for the 18 positive compounds. By comparison with 102 previously annotated bitter compounds in the database, we discovered common descriptor properties that may contribute to the discovery of additional bitter ligands and further expand the known molecular receptive ranges of human TAS2Rs.

Collaboration


Dive into the Wenkang Huang's collaboration.

Top Co-Authors

Avatar

Jian Zhang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Shaoyong Lu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Qiancheng Shen

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Xinyi Liu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Zhimin Huang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Shuai Li

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Yingyi Chen

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Lv Geng

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Ruth Nussinov

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Qi Wang

Shanghai Jiao Tong University

View shared research outputs
Researchain Logo
Decentralizing Knowledge