Network


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

Hotspot


Dive into the research topics where Beiyan Zou is active.

Publication


Featured researches published by Beiyan Zou.


Cell | 2015

Advancing Biological Understanding and Therapeutics Discovery with Small-Molecule Probes

Stuart L. Schreiber; Joanne Kotz; Min Li; Jeffrey Aubé; Christopher P. Austin; John C. Reed; Hugh Rosen; E. Lucile White; Larry A. Sklar; Craig W. Lindsley; Benjamin Alexander; Joshua Bittker; Paul A. Clemons; Andrea de Souza; Michael Foley; Michelle Palmer; Alykhan F. Shamji; Mathias J. Wawer; Owen B. McManus; Meng Wu; Beiyan Zou; Haibo Yu; Jennifer E. Golden; Frank J. Schoenen; Anton Simeonov; Ajit Jadhav; Michael R. Jackson; Anthony B. Pinkerton; Thomas Dy Chung; Patrick R. Griffin

Small-molecule probes can illuminate biological processes and aid in the assessment of emerging therapeutic targets by perturbing biological systems in a manner distinct from other experimental approaches. Despite the tremendous promise of chemical tools for investigating biology and disease, small-molecule probes were unavailable for most targets and pathways as recently as a decade ago. In 2005, the NIH launched the decade-long Molecular Libraries Program with the intent of innovating in and broadening access to small-molecule science. This Perspective describes how novel small-molecule probes identified through the program are enabling the exploration of biological pathways and therapeutic hypotheses not otherwise testable. These experiences illustrate how small-molecule probes can help bridge the chasm between biological research and the development of medicines but also highlight the need to innovate the science of therapeutic discovery.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Modulation of hERG potassium channel gating normalizes action potential duration prolonged by dysfunctional KCNQ1 potassium channel

Hongkang Zhang; Beiyan Zou; Haibo Yu; Alessandra Moretti; Xiaoying Wang; Wei Yan; Joseph J. Babcock; Milena Bellin; Owen B. McManus; Gordon F. Tomaselli; Fajun Nan; Karl-Ludwig Laugwitz; Min Li

Long QT syndrome (LQTS) is a genetic disease characterized by a prolonged QT interval in an electrocardiogram (ECG), leading to higher risk of sudden cardiac death. Among the 12 identified genes causal to heritable LQTS, ∼90% of affected individuals harbor mutations in either KCNQ1 or human ether-a-go-go related genes (hERG), which encode two repolarizing potassium currents known as IKs and IKr. The ability to quantitatively assess contributions of different current components is therefore important for investigating disease phenotypes and testing effectiveness of pharmacological modulation. Here we report a quantitative analysis by simulating cardiac action potentials of cultured human cardiomyocytes to match the experimental waveforms of both healthy control and LQT syndrome type 1 (LQT1) action potentials. The quantitative evaluation suggests that elevation of IKr by reducing voltage sensitivity of inactivation, not via slowing of deactivation, could more effectively restore normal QT duration if IKs is reduced. Using a unique specific chemical activator for IKr that has a primary effect of causing a right shift of V1/2 for inactivation, we then examined the duration changes of autonomous action potentials from differentiated human cardiomyocytes. Indeed, this activator causes dose-dependent shortening of the action potential durations and is able to normalize action potentials of cells of patients with LQT1. In contrast, an IKr chemical activator of primary effects in slowing channel deactivation was not effective in modulating action potential durations. Our studies provide both the theoretical basis and experimental support for compensatory normalization of action potential duration by a pharmacological agent.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Dynamic subunit stoichiometry confers a progressive continuum of pharmacological sensitivity by KCNQ potassium channels

Haibo Yu; Zhihong Lin; Margrith E. Mattmann; Beiyan Zou; Cecile Terrenoire; Hongkang Zhang; Meng Wu; Owen B. McManus; Robert S. Kass; Craig W. Lindsley; Corey R. Hopkins; Min Li

Voltage-gated KCNQ1 (Kv7.1) potassium channels are expressed abundantly in heart but they are also found in multiple other tissues. Differential coassembly with single transmembrane KCNE beta subunits in different cell types gives rise to a variety of biophysical properties, hence endowing distinct physiological roles for KCNQ1–KCNEx complexes. Mutations in either KCNQ1 or KCNE1 genes result in diseases in brain, heart, and the respiratory system. In addition to complexities arising from existence of five KCNE subunits, KCNE1 to KCNE5, recent studies in heterologous systems suggest unorthodox stoichiometric dynamics in subunit assembly is dependent on KCNE expression levels. The resultant KCNQ1–KCNE channel complexes may have a range of zero to two or even up to four KCNE subunits coassembling per KCNQ1 tetramer. These findings underscore the need to assess the selectivity of small-molecule KCNQ1 modulators on these different assemblies. Here we report a unique small-molecule gating modulator, ML277, that potentiates both homomultimeric KCNQ1 channels and unsaturated heteromultimeric (KCNQ1)4(KCNE1)n (n < 4) channels. Progressive increase of KCNE1 or KCNE3 expression reduces efficacy of ML277 and eventually abolishes ML277-mediated augmentation. In cardiomyocytes, the slowly activating delayed rectifier potassium current, or IKs, is believed to be a heteromultimeric combination of KCNQ1 and KCNE1, but it is not entirely clear whether IKs is mediated by KCNE-saturated KCNQ1 channels or by channels with intermediate stoichiometries. We found ML277 effectively augments IKs current of cultured human cardiomyocytes and shortens action potential duration. These data indicate that unsaturated heteromultimeric (KCNQ1)4(KCNE1)n channels are present as components of IKs and are pharmacologically distinct from KCNE-saturated KCNQ1–KCNE1 channels.


Assay and Drug Development Technologies | 2011

HERGCentral: A large database to store, retrieve, and analyze compound-human ether-à-go-go related gene channel interactions to facilitate cardiotoxicity assessment in drug development

Fang Du; Haibo Yu; Beiyan Zou; Joseph J. Babcock; Shunyou Long; Min Li

The unintended and often promiscous inhibition of the cardiac human Ether-à-go-go related gene (hERG) potassium channel is a common cause for either delay or removal of therapeutic compounds from development and withdrawal of marketed drugs. The clinical manifestion is prolongation of the duration between QRS complex and T-wave measured by surface electrocardiogram (ECG)-hence Long QT Syndrome. There are several useful online resources documenting hERG inhibition by known drugs and bioactives. However, their utilities remain somewhat limited because they are biased toward well-studied compounds and their number of data points tends to be much smaller than many commercial compound libraries. The hERGCentral ( www.hergcentral.org ) is mainly based on experimental data obtained from a primary screen by electrophysiology against more than 300,000 structurally diverse compounds. The system is aimed to display and combine three resources: primary electrophysiological data, literature, as well as online reports and chemical library collections. Currently, hERGCentral has annotated datasets of more than 300,000 compounds including structures and chemophysiological properties of compounds, raw traces, and biophysical properties. The system enables a variety of query formats, including searches for hERG effects according to either chemical structure or properties, and alternatively according to the specific biophysical properties of current changes caused by a compound. Therefore, the hERGCentral, as a unique and evolving resource, will facilitate investigation of chemically induced hERG inhibition and therefore drug development.


Assay and Drug Development Technologies | 2010

Profiling diverse compounds by flux- and electrophysiology-based primary screens for inhibition of human Ether-a-go-go related gene potassium channels.

Beiyan Zou; Haibo Yu; Joseph J. Babcock; Pritam Chanda; Joel S. Bader; Owen B. McManus; Min Li

Compound effects on cloned human Ether-à-go-go related gene (hERG) potassium channels have been used to assess the potential cardiac safety liabilities of drug development candidate compounds. In addition to radioactive ligand displacement tests, two other common approaches are surrogate ion-based flux assays and electrophysiological recordings. The former has much higher throughput, whereas the latter measures directly the effects on ionic currents. Careful characterization in earlier reports has been performed to compare the relative effectiveness of these approaches for known hERG blockers, which often yielded good overall correlation. However, cases were reported showing significant and reproducible differences in potency and/or sensitivity by the two methods. This raises a question concerning the rationale and criteria on which an assay should be selected for evaluating unknown compounds. To provide a general basis for considering assays to profile large compound libraries for hERG activity, we have conducted parallel flux and electrophysiological analyses of 2,000 diverse compounds, representative of the 300,000 compound collection of NIH Molecular Library Small Molecular Repository (MLSMR). Our results indicate that at the conventional testing concentration 1.0 μM, the overlap between the two assays ranges from 32% to 50% depending on the hit selection criteria. There was a noticeable rate of false negatives by the thallium-based assay relative to electrophysiological recording, which may be greatly reduced under modified comparative conditions. As these statistical results identify a preferred method for cardiac safety profiling of unknown compounds, they suggest an efficient method combining flux and electrophysiological assays to rapidly profile hERG liabilities of large collection of naive compounds.


Bioorganic & Medicinal Chemistry Letters | 2014

Potent and Selective Inhibitors of the TASK-1 Potassium Channel Through Chemical Optimization of a Bis-amide Scaffold

Daniel P. Flaherty; Denise S. Simpson; Melissa Miller; Brooks E. Maki; Beiyan Zou; Jie Shi; Meng Wu; Owen B. McManus; Jeffrey Aubé; Min Li; Jennifer E. Golden

TASK-1 is a two-pore domain potassium channel that is important to modulating cell excitability, most notably in the context of neuronal pathways. In order to leverage TASK-1 for therapeutic benefit, its physiological role needs better characterization; however, designing selective inhibitors that avoid the closely related TASK-3 channel has been challenging. In this study, a series of bis-amide derived compounds were found to demonstrate improved TASK-1 selectivity over TASK-3 compared to reported inhibitors. Optimization of a marginally selective hit led to analog 35 which displays a TASK-1 IC50=16 nM with 62-fold selectivity over TASK-3 in an orthogonal electrophysiology assay.


European Journal of Pharmacology | 2014

Identification of novel small molecule modulators of K2P18.1 two-pore potassium channel

J. Kyle Bruner; Beiyan Zou; Hongkang Zhang; Yixin Zhang; Katharina Schmidt; Min Li

Two-pore domain potassium (K2P) channels are responsible for background potassium (K+) current, which is crucial for the maintenance of resting membrane potential. K2P18.1, also called TWIK-related spinal cord K+ channel (TRESK) or KCNK18, is thought to be a major contributor to background K+ currents, particularly in sensory neurons where it is abundantly expressed. Despite its critical role and potential therapeutic implication, pharmacological tools for probing K2P18.1 activity remain unavailable. Here, we report a high-throughput screen against a collection of bioactive compounds that yielded 26 inhibitors and 8 activators of K2P18.1 channel activity with more than 10-fold selectivity over the homologous channel K2P9.1. Among these modulators, the antihistamine loratadine inhibited K2P18.1 activity with IC50 of 0.49±0.23 µM and is considerably more potent than existing K2P18.1 inhibitors. Importantly, the inhibition by loratadine remains equally efficacious upon potentiation of K2P18.1 by calcium signaling. Furthermore, the loratadine effect is dependent on transmembrane residues F145 and F352, providing orthogonal evidence that the inhibition is caused by a direct compound-channel interaction. This study reveals new pharmacological modulators of K2P18.1 activity useful in dissecting native K2P18.1 function.


Journal of Medicinal Chemistry | 2012

Discovery of a Series of 2-phenyl-N-(2-(pyrrolidin-1-yl)phenyl)acetamides as Novel Molecular Switches that Modulate Modes of Kv7.2 (KCNQ2) Channel Pharmacology: Identification of (S)-2-phenyl-N-(2-(pyrrolidin-1-yl)phenyl)butanamide (ML252) as a Potent, Brain Penetrant Kv7.2 Channel Inhibitor

Yiu Yin Cheung; Haibo Yu; Kaiping Xu; Beiyan Zou; Meng Wu; Owen B. McManus; Min Li; Craig W. Lindsley; Corey R. Hopkins

A potent and selective inhibitor of KCNQ2, (S)-5 (ML252, IC(50) = 69 nM), was discovered after a high-throughput screen of the MLPCN library was performed. SAR studies revealed a small structural change (ethyl group to hydrogen) caused a functional shift from antagonist to agonist activity (37, EC(50) = 170 nM), suggesting an interaction at a critical site for controlling gating of KCNQ2 channels.


Journal of Biological Chemistry | 2012

Identification and characterization of a compound that protects cardiac tissue from human Ether-à-go-go-related gene (hERG)-related drug-induced arrhythmias

Franck Potet; Amanda N. Lorinc; Sebastien Chaigne; Corey R. Hopkins; Raghav Venkataraman; Svetlana Z. Stepanovic; L. Michelle Lewis; Emily Days; Veniamin Y. Sidorov; Darren W. Engers; Beiyan Zou; David Afshartous; Alfred L. George; Courtney M. Campbell; Jeffrey R. Balser; Min Li; Franz J. Baudenbacher; Craig W. Lindsley; C. David Weaver; Sabina Kupershmidt

Background: Inhibition of the cardiac hERG channel by essential pharmaceuticals is unpredictable and leads to fatal arrhythmias. Results: Pretreatment with a newly identified compound, VU0405601, reduces sensitivity of hERG to inhibition by multiple blockers and prevents arrhythmias. Conclusion: hERG-related arrhythmias are amenable to preventive therapy. Significance: A novel approach at ion channel modulation that impacts drug discovery and safety concerns is outlined. The human Ether-à-go-go-related gene (hERG)-encoded K+ current, IKr is essential for cardiac repolarization but is also a source of cardiotoxicity because unintended hERG inhibition by diverse pharmaceuticals can cause arrhythmias and sudden cardiac death. We hypothesized that a small molecule that diminishes IKr block by a known hERG antagonist would constitute a first step toward preventing hERG-related arrhythmias and facilitating drug discovery. Using a high-throughput assay, we screened a library of compounds for agents that increase the IC70 of dofetilide, a well characterized hERG blocker. One compound, VU0405601, with the desired activity was further characterized. In isolated, Langendorff-perfused rabbit hearts, optical mapping revealed that dofetilide-induced arrhythmias were reduced after pretreatment with VU0405601. Patch clamp analysis in stable hERG-HEK cells showed effects on current amplitude, inactivation, and deactivation. VU0405601 increased the IC50 of dofetilide from 38.7 to 76.3 nm. VU0405601 mitigates the effects of hERG blockers from the extracellular aspect primarily by reducing inactivation, whereas most clinically relevant hERG inhibitors act at an inner pore site. Structure-activity relationships surrounding VU0405601 identified a 3-pyridiyl and a naphthyridine ring system as key structural components important for preventing hERG inhibition by multiple inhibitors. These findings indicate that small molecules can be designed to reduce the sensitivity of hERG to inhibitors.


Neuropharmacology | 2017

Loperamide inhibits sodium channels to alleviate inflammatory hyperalgesia

Ying Wu; Beiyan Zou; Lingli Liang; Min Li; Yuan Xiang Tao; Haibo Yu; Xiaoliang Wang

&NA; Previous studies demonstrated that Loperamide, originally known as an anti‐diarrheal drug, is a promising analgesic agent primarily targeting mu‐opioid receptors. However some evidences suggested that non‐opioid mechanisms may be contributing to its analgesic effect. In the present study, Loperamide was identified as a Nav1.7 blocker in a pilot screen. In HEK293 cells expressing Nav1.7 sodium channels, Loperamide blocked the resting state of Nav1.7 channels (IC50 = 1.86 ± 0.11 &mgr;M) dose‐dependently and reversibly. Loperamide produced a 10.4 mV of hyperpolarizing shift for the steady‐state inactivation of Nav1.7 channels without apparent effect on the voltage‐dependent activation. The drug displayed a mild use‐ and state‐dependent inhibition on Nav1.7 channels, which was removed by the local anesthetic‐insensitive construct Nav1.7‐F1737A. Inhibition of Nav1.7 at resting state was not altered significantly by the F1737A mutation. Compared to its effects on Nav1.7, Loperamide exhibited higher potency on recombinant Nav1.8 channels in ND7/23 cells (IC50 = 0.60 ± 0.10 &mgr;M) and weaker potency on Nav1.9 channels (3.48 ± 0.33 &mgr;M). Notably more pronounced inhibition was observed in the native Nav1.8 channels (0.11 ± 0.08 &mgr;M) in DRG neurons. Once mu‐opioid receptor was antagonized by Naloxone in DRG neurons, potency of Loperamide on Nav1.8 was identical to that of recombinant Nav1.8 channels. The inhibition on Nav channels may be the main mechanism of Loperamide for pain relief beyond mu‐opioid receptor. In the meanwhile, the opioid receptor pathway may also influence the blocking effect of Loperamide on sodium channels, implying a cross‐talk between sodium channels and opioid receptors in pain processing. HighlightsA non‐opioid mechanism for Loperamide to relieve pain is proposed.The inhibition on Nav channels might be the possible mechanism of Loperamide for pain relief beyond mu‐opioid receptor.Crosstalk may exist between opioid receptor and sodium channels.

Collaboration


Dive into the Beiyan Zou's collaboration.

Top Co-Authors

Avatar

Min Li

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Haibo Yu

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jie Shi

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Melissa R. Miller

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge