Network


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

Hotspot


Dive into the research topics where James B Herrington is active.

Publication


Featured researches published by James B Herrington.


Journal of Pharmacology and Experimental Therapeutics | 2010

Analgesic Effects of a Substituted N-Triazole Oxindole (TROX-1), a State-Dependent, Voltage-Gated Calcium Channel 2 Blocker

Catherine Abbadie; Owen B. McManus; Shu-Yu Sun; Randal M. Bugianesi; Ge Dai; Rodolfo J. Haedo; James B Herrington; Gregory J. Kaczorowski; McHardy M. Smith; Andrew M. Swensen; Vivien A. Warren; Brande S. Williams; Stephen P. Arneric; Cyrus Eduljee; Terrance P. Snutch; Elizabeth W. Tringham; Nina Jochnowitz; Annie Liang; D. Euan MacIntyre; Erin McGowan; Shruti Mistry; Valerie V. White; Scott B. Hoyt; Clare London; Kathryn A. Lyons; Patricia B. Bunting; Sylvia Volksdorf; Joseph L. Duffy

Voltage-gated calcium channel (Cav)2.2 (N-type calcium channels) are key components in nociceptive transmission pathways. Ziconotide, a state-independent peptide inhibitor of Cav2.2 channels, is efficacious in treating refractory pain but exhibits a narrow therapeutic window and must be administered intrathecally. We have discovered an N-triazole oxindole, (3R)-5-(3-chloro-4-fluorophenyl)-3-methyl-3-(pyrimidin-5-ylmethyl)-1-(1H-1,2,4-triazol-3-yl)-1,3-dihydro-2H-indol-2-one (TROX-1), as a small-molecule, state-dependent blocker of Cav2 channels, and we investigated the therapeutic advantages of this compound for analgesia. TROX-1 preferentially inhibited potassium-triggered calcium influx through recombinant Cav2.2 channels under depolarized conditions (IC50 = 0.27 μM) compared with hyperpolarized conditions (IC50 > 20 μM). In rat dorsal root ganglion (DRG) neurons, TROX-1 inhibited ω-conotoxin GVIA-sensitive calcium currents (Cav2.2 channel currents), with greater potency under depolarized conditions (IC50 = 0.4 μM) than under hyperpolarized conditions (IC50 = 2.6 μM), indicating state-dependent Cav2.2 channel block of native as well as recombinant channels. TROX-1 fully blocked calcium influx mediated by a mixture of Cav2 channels in calcium imaging experiments in rat DRG neurons, indicating additional block of all Cav2 family channels. TROX-1 reversed inflammatory-induced hyperalgesia with maximal effects equivalent to nonsteroidal anti-inflammatory drugs, and it reversed nerve injury-induced allodynia to the same extent as pregabalin and duloxetine. In contrast, no significant reversal of hyperalgesia was observed in Cav2.2 gene-deleted mice. Mild impairment of motor function in the Rotarod test and cardiovascular functions were observed at 20- to 40-fold higher plasma concentrations than required for analgesic activities. TROX-1 demonstrates that an orally available state-dependent Cav2 channel blocker may achieve a therapeutic window suitable for the treatment of chronic pain.


Assay and Drug Development Technologies | 2008

A high-throughput assay for evaluating state dependence and subtype selectivity of Cav2 calcium channel inhibitors.

Ge Dai; Rodolfo J. Haedo; Vivien A. Warren; Kevin S. Ratliff; Randal M. Bugianesi; Alison Rush; Mark E. Williams; James B Herrington; McHardy M. Smith; Owen B. McManus; Andrew M. Swensen

Cav2.2 channels play a critical role in pain signaling by controlling synaptic transmission between dorsal root ganglion neurons and dorsal horn neurons. The Cav2.2-selective peptide blocker ziconotide (Prialt, Elan Pharmaceuticals, Dublin, Ireland) has proven efficacious in pain relief, but has a poor therapeutic index and requires intrathecal administration. This has provided impetus for finding an orally active, state-dependent Cav2.2 inhibitor with an improved safety profile. Members of the Cav2 subfamily of calcium channels are the main contributors to central and peripheral synaptic transmission, but the pharmacological effects of blocking each subtype is not yet defined. Here we describe a high-throughput fluorescent assay using a fluorometric imaging plate reader (FLIPR [Molecular Devices, Sunnyvale, CA]) designed to quickly evaluate the state dependence and selectivity of inhibitors across the Cav2 subfamily. Stable cell lines expressing functional Cav2 channels (Ca(V)alpha, beta(3), and alpha(2)delta subunits) were co-transfected with an inward rectifier (Kir2.3) so that membrane potential, and therefore channel state, could be controlled by external potassium concentration. Following cell incubation in drug with varying concentrations of potassium, a high potassium trigger was added to elicit calcium influx through available, unblocked channels. State-dependent inhibitors that preferentially bind to channels in the open or inactivated state can be identified by their increased potency at higher potassium concentrations, where cells are depolarized and channels are biased towards these states. Although the Cav2 channel subtypes differ in their voltage dependence of inactivation, by adjusting pre-trigger potassium concentrations, the degree of steady-state inactivation can be more closely matched across Cav2 subtypes to assess molecular selectivity.


Molecular Pharmacology | 2012

Characterization of the Substituted N-Triazole Oxindole TROX-1, a Small-Molecule, State-Dependent Inhibitor of Cav2 Calcium Channels

Andrew M. Swensen; James B Herrington; Randal M. Bugianesi; Ge Dai; Rodolfo J. Haedo; Kevin S. Ratliff; McHardy M. Smith; Vivien A. Warren; Stephen P. Arneric; Cyrus Eduljee; David Parker; Terrance P. Snutch; Scott B. Hoyt; Clare London; Joseph L. Duffy; Gregory J. Kaczorowski; Owen B. McManus

Biological, genetic, and clinical evidence provide validation for N-type calcium channels (CaV2.2) as therapeutic targets for chronic pain. A state-dependent CaV2.2 inhibitor may provide an improved therapeutic window over ziconotide, the peptidyl CaV2.2 inhibitor used clinically. Supporting this notion, we recently reported that in preclinical models, the state-dependent CaV2 inhibitor (3R)-5-(3-chloro-4-fluorophenyl)-3-methyl-3-(pyrimidin-5-ylmethyl)-1-(1H-1,2,4-triazol-3-yl)-1,3-dihydro-2H-indol-2-one (TROX-1) has an improved therapeutic window compared with ziconotide. Here we characterize TROX-1 inhibition of Cav2.2 channels in more detail. When channels are biased toward open/inactivated states by depolarizing the membrane potential under voltage-clamp electrophysiology, TROX-1 inhibits CaV2.2 channels with an IC50 of 0.11 μM. The voltage dependence of CaV2.2 inhibition was examined using automated electrophysiology. TROX-1 IC50 values were 4.2, 0.90, and 0.36 μM at −110, −90, and −70 mV, respectively. TROX-1 displayed use-dependent inhibition of CaV2.2 with a 10-fold IC50 separation between first (27 μM) and last (2.7 μM) pulses in a train. In a fluorescence-based calcium influx assay, TROX-1 inhibited CaV2.2 channels with an IC50 of 9.5 μM under hyperpolarized conditions and 0.69 μM under depolarized conditions. Finally, TROX-1 potency was examined across the CaV2 subfamily. Depolarized IC50 values were 0.29, 0.19, and 0.28 μM by manual electrophysiology using matched conditions and 1.8, 0.69, and 1.1 μM by calcium influx for CaV2.1, CaV2.2, and CaV2.3, respectively. Together, these in vitro data support the idea that a state-dependent, non–subtype-selective CaV2 channel inhibitor can achieve an improved therapeutic window over the relatively state-independent CaV2.2-selective inhibitor ziconotide in preclinical models of chronic pain.


The Journal of Physiology | 2005

Biophysical and pharmacological properties of the voltage-gated potassium current of human pancreatic β-cells

James B Herrington; Manuel Calderon De La Barca Sanchez; Denize Wunderler; Lizhen Yan; Randal M. Bugianesi; Ivy E. Dick; Sam A. Clark; Richard M. Brochu; Birgit T. Priest; Martin Kohler; Owen B. McManus

Voltage‐gated potassium (Kv) currents of human pancreatic islet cells were studied by whole‐cell patch clamp recording. On average, 75% of the cells tested were identified as β‐cells by single cell, post‐recording RT‐PCR for insulin mRNA. In most cells, the dominant Kv current was a delayed rectifier. The delayed rectifier activated at potentials above −20 mV and had a V½ for activation of −5.3 mV. Onset of inactivation was slow for a major component (τ= 3.2 s at +20 mV) observed in all cells; a smaller component (τ= 0.30 s) with an amplitude of ∼25% was seen in some cells. Recovery from inactivation had a τ of 2.5 s at −80 mV and steady‐state inactivation had a V½ of −39 mV. In 12% of cells (21/182) a low‐threshold, transient Kv current (A‐current) was present. The A‐current activated at membrane potentials above −40 mV, inactivated with a time constant of 18.5 ms at −20 mV, and had a V½ for steady‐state inactivation of −52 mV. TEA inhibited total Kv current with an IC50= 0.54 mm and PAC, a disubstituted cyclohexyl Kv channel inhibitor, inhibited with an IC50= 0.57 μm. The total Kv current was insensitive to margatoxin (100 nm), agitoxin‐2 (50 nm), kaliotoxin (50 nm) and ShK (50 nm). Hanatoxin (100 nm) inhibited total Kv current by 65% at +20 mV. Taken together, these data provide evidence of at least two distinct types of Kv channels in human pancreatic β‐cells and suggest that more than one type of Kv channel may be involved in the regulation of glucose‐dependent insulin secretion.


Journal of Medicinal Chemistry | 2016

Discovery of GluN2A-Selective NMDA Receptor Positive Allosteric Modulators (PAMs): Tuning Deactivation Kinetics via Structure-Based Design.

Matthew Volgraf; Benjamin D. Sellers; Yu Jiang; Guosheng Wu; Cuong Ly; Elisia Villemure; Richard Pastor; Po-wai Yuen; Aijun Lu; Xifeng Luo; Mingcui Liu; Shun Zhang; Liang Sun; Yuhong Fu; Patrick J. Lupardus; Heidi J.A. Wallweber; Bianca M. Liederer; Gauri Deshmukh; Emile Plise; Suzanne Tay; Paul Reynen; James B Herrington; Amy Gustafson; Yichin Liu; Akim Dirksen; Matthias G. A. Dietz; Yanzhou Liu; Tzu-Ming Wang; Jesse E. Hanson; David H. Hackos

The N-methyl-D-aspartate receptor (NMDAR) is a Na(+) and Ca(2+) permeable ionotropic glutamate receptor that is activated by the coagonists glycine and glutamate. NMDARs are critical to synaptic signaling and plasticity, and their dysfunction has been implicated in a number of neurological disorders, including schizophrenia, depression, and Alzheimers disease. Herein we describe the discovery of potent GluN2A-selective NMDAR positive allosteric modulators (PAMs) starting from a high-throughput screening hit. Using structure-based design, we sought to increase potency at the GluN2A subtype, while improving selectivity against related α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs). The structure-activity relationship of channel deactivation kinetics was studied using a combination of electrophysiology and protein crystallography. Effective incorporation of these strategies resulted in the discovery of GNE-0723 (46), a highly potent and brain penetrant GluN2A-selective NMDAR PAM suitable for in vivo characterization.


Bioorganic & Medicinal Chemistry Letters | 2011

A potent and selective indole N-type calcium channel (Cav2.2) blocker for the treatment of pain

Sriram Tyagarajan; Prasun K. Chakravarty; Min Park; Bishan Zhou; James B Herrington; Kevin S. Ratliff; Randall M. Bugianesi; Brande S. Williams; Rodolfo J. Haedo; Andrew M. Swensen; Vivien A. Warren; McHardy M. Smith; Maria L. Garcia; Gregory J. Kaczorowski; Owen B. McManus; Kathryn A. Lyons; Xiaohua Li; Maria Madeira; Bindhu V. Karanam; Mitchell D. Green; Michael J. Forrest; Catherine Abbadie; Erin McGowan; Shruti Mistry; Nina Jochnowitz; Joseph L. Duffy

N-type calcium channels (Ca(v)2.2) have been shown to play a critical role in pain. A series of low molecular weight 2-aryl indoles were identified as potent Ca(v)2.2 blockers with good in vitro and in vivo potency.


Journal of Medicinal Chemistry | 2012

Aminopiperidine sulfonamide Cav2.2 channel inhibitors for the treatment of chronic pain.

Pengcheng P. Shao; Feng Ye; Prasun K. Chakravarty; Deepu J. Varughese; James B Herrington; Ge Dai; Randal M. Bugianesi; Rodolfo J. Haedo; Andrew M. Swensen; Vivien A. Warren; McHardy M. Smith; Maria L. Garcia; Owen B. McManus; Kathryn A. Lyons; Xiaohua Li; Mitchell D. Green; Nina Jochnowitz; Erin McGowan; Shruti Mistry; Shu-Yu Sun; Catherine Abbadie; Gregory J. Kaczorowski; Joseph L. Duffy

The voltage-gated calcium channel Ca(v)2.2 (N-type calcium channel) is a critical regulator of synaptic transmission and has emerged as an attractive target for the treatment of chronic pain. We report here the discovery of sulfonamide-derived, state-dependent inhibitors of Ca(v)2.2. In particular, 19 is an inhibitor of Ca(v)2.2 that is selective over cardiac ion channels, with a good preclinical PK and biodistribution profile. This compound exhibits dose-dependent efficacy in preclinical models of inflammatory hyperalgesia and neuropathic allodynia and is devoid of ancillary cardiovascular or CNS pharmacology at the doses tested. Importantly, 19 exhibited no efficacy in Ca(v)2.2 gene-deleted mice. The discovery of metabolite 26 confounds further development of members of this aminopiperidine sulfonamide series. This discovery also suggests specific structural liabilities of this class of compounds that must be addressed.


Molecular Pharmacology | 2011

Identification of novel and selective Kv2 channel inhibitors.

James B Herrington; Kelli Solly; Kevin S. Ratliff; Nina Li; Yun-Ping Zhou; Andrew W. Howard; L. L. Kiss; Maria L. Garcia; Owen B. McManus; Qiaolin Deng; Ranjit Desai; Yusheng Xiong; Gregory J. Kaczorowski

Identification of selective ion channel inhibitors represents a critical step for understanding the physiological role that these proteins play in native systems. In particular, voltage-gated potassium (KV2) channels are widely expressed in tissues such as central nervous system, pancreas, and smooth muscle, but their particular contributions to cell function are not well understood. Although potent and selective peptide inhibitors of KV2 channels have been characterized, selective small molecule KV2 inhibitors have not been reported. For this purpose, high-throughput automated electrophysiology (IonWorks Quattro; Molecular Devices, Sunnyvale, CA) was used to screen a 200,000-compound mixture (10 compounds per sample) library for inhibitors of KV2.1 channels. After deconvolution of 190 active samples, two compounds (A1 and B1) were identified that potently inhibit KV2.1 and the other member of the KV2 family, KV2.2 (IC50, 0.1–0.2 μM), and that possess good selectivity over KV1.2 (IC50 >10 μM). Modeling studies suggest that these compounds possess a similar three-dimensional conformation. Compounds A1 and B1 are >10-fold selective over NaV channels and other KV channels and display weak activity (5–9 μM) on CaV channels. The biological activity of compound A1 on native KV2 channels was confirmed in electrophysiological recordings of rat insulinoma cells, which are known to express KV2 channels. Medicinal chemistry efforts revealed a defined structure-activity relationship and led to the identification of two compounds (RY785 and RY796) without significant CaV channel activity. Taken together, these newly identified channel inhibitors represent important tools for the study of KV2 channels in biological systems.


ACS Medicinal Chemistry Letters | 2013

Improved Cav2.2 Channel Inhibitors through a gem-Dimethylsulfone Bioisostere Replacement of a Labile Sulfonamide

Pengcheng P. Shao; Feng Ye; Prasun K. Chakravarty; James B Herrington; Ge Dai; Randal M. Bugianesi; Rodolfo J. Haedo; Andrew M. Swensen; Vivien A. Warren; McHardy M. Smith; Maria L. Garcia; Owen B. McManus; Kathryn A. Lyons; Xiaohua Li; Mitchell D. Green; Nina Jochnowitz; Erin McGowan; Shruti Mistry; Shu-Yu Sun; Catherine Abbadie; Gregory J. Kaczorowski; Joseph L. Duffy

We report the investigation of sulfonamide-derived Cav2.2 inhibitors to address drug-metabolism liabilities with this lead class of analgesics. Modification of the benzamide substituent provided improvements in both potency and selectivity. However, we discovered that formation of the persistent 3-(trifluoromethyl)benzenesulfonamide metabolite was an endemic problem in the sulfonamide series and that the replacement of the center aminopiperidine scaffold failed to prevent this metabolic pathway. This issue was eventually addressed by application of a bioisostere strategy. The new gem-dimethyl sulfone series retained Cav2.2 potency without the liability of the circulating sulfonamide metabolite.


Assay and Drug Development Technologies | 2008

An Automated Electrophysiology Serum Shift Assay for KV Channels

Kevin S. Ratliff; Aleksandr Petrov; George J. Eiermann; Qiaolin Deng; Mitchell D. Green; Gregory J. Kaczorowski; Owen B. McManus; James B Herrington

The presence of serum in biological samples often negatively impacts the quality of in vitro assays. However, assays tolerant of serum are useful for assessing the in vivo availability of a small molecule for its target. Electrophysiology assays of ion channels are notoriously sensitive to serum because of their reliance on the interaction of the plasma membrane with a recording electrode. Here we investigate the tolerance of an automated electrophysiology assay for a voltage-gated potassium (K(V)) channel to serum and purified plasma proteins. The delayed rectifier channel, K(V)2.1, stably expressed in Chinese hamster ovary cells produces large, stable currents on the IonWorks Quattro platform (MDS Analytical Technologies, Sunnyvale, CA), making it an ideal test case. K(V)2.1 currents recorded on this platform are highly resistant to serum, allowing recordings in as high as 33% serum. Using a set of compounds related to the K(V) channel blocker, 4-phenyl-4-[3-(2-methoxyphenyl)-3-oxo-2-azaprop-1-yl]cyclohexanone, we show that shifts in compound potency with whole serum or isolated serum proteins can be reliably measured with this assay. Importantly, this assay is also relatively insensitive to plasma, allowing the creation of a bioassay for inhibitors of K(V)2.1 channel activity. Here we show that such a bioassay can quantify the levels of the gating modifier, guangxitoxin-1E, in plasma samples from mice dosed with the peptide. This study demonstrates the utility of using an automated electrophysiology platform for measuring serum shifts and for bioassays of ion channel modulators.

Collaboration


Dive into the James B Herrington's collaboration.

Researchain Logo
Decentralizing Knowledge