Network


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

Hotspot


Dive into the research topics where Ge Dai is active.

Publication


Featured researches published by Ge Dai.


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.


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 | 2012

Selective, direct activation of high-conductance, calcium-activated potassium channels causes smooth muscle relaxation.

Cristiano G. Ponte; Owen B. McManus; William A. Schmalhofer; Dong-Ming Shen; Ge Dai; Andra Stevenson; Sylvie Sur; Tarak Shah; L. L. Kiss; Min Shu; James B. Doherty; Ravi P. Nargund; Gregory J. Kaczorowski; Guilherme Suarez-Kurtz; Maria L. Garcia

High-conductance calcium-activated potassium (Maxi-K) channels are present in smooth muscle where they regulate tone. Activation of Maxi-K channels causes smooth muscle hyperpolarization and shortening of action-potential duration, which would limit calcium entry through voltage-dependent calcium channels leading to relaxation. Although Maxi-K channels appear to indirectly mediate the relaxant effects of a number of agents, activators that bind directly to the channel with appropriate potency and pharmacological properties useful for proof-of-concept studies are not available. Most agents identified to date display significant polypharmacy that severely compromises interpretation of experimental data. In the present study, a high-throughput, functional, cell-based assay for identifying Maxi-K channel agonists was established and used to screen a large sample collection (>1.6 million compounds). On the basis of potency and selectivity, a family of tetrahydroquinolines was further characterized. Medicinal chemistry efforts afforded identification of compound X, from which its two enantiomers, Y and Z, were resolved. In in vitro assays, Z is more potent than Y as a channel activator. The same profile is observed in tissues where the ability of either agent to relax precontracted smooth muscles, via a potassium channel-dependent mechanism, is demonstrated. These data, taken together, suggest that direct activation of Maxi-K channels represents a mechanism to be explored for the potential treatment of a number of diseases associated with smooth muscle hyperexcitability.


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.


JCI insight | 2018

A glucose-responsive insulin therapy protects animals against hypoglycemia

Ruojing Yang; Margaret Wu; Songnian Lin; Ravi P. Nargund; Xinghai Li; Theresa M. Kelly; Lin Yan; Ge Dai; Ying Qian; Qing Dallas-Yang; Paul Fischer; Yan Cui; Xiaolan Shen; Pei Huo; Danqing Dennis Feng; Mark D. Erion; David E. Kelley; James Mu

Hypoglycemia is commonly associated with insulin therapy, limiting both its safety and efficacy. The concept of modifying insulin to render its glucose-responsive release from an injection depot (of an insulin complexed exogenously with a recombinant lectin) was proposed approximately 4 decades ago but has been challenging to achieve. Data presented here demonstrate that mannosylated insulin analogs can undergo an additional route of clearance as result of their interaction with endogenous mannose receptor (MR), and this can occur in a glucose-dependent fashion, with increased binding to MR at low glucose. Yet, these analogs retain capacity for binding to the insulin receptor (IR). When the blood glucose level is elevated, as in individuals with diabetes mellitus, MR binding diminishes due to glucose competition, leading to reduced MR-mediated clearance and increased partitioning for IR binding and consequent glucose lowering. These studies demonstrate that a glucose-dependent locus of insulin clearance and, hence, insulin action can be achieved by targeting MR and IR concurrently.


Diabetes | 2018

Erratum. Engineering Glucose Responsiveness Into Insulin. Diabetes 2018;67:299–308

Niels C. Kaarsholm; Songnian Lin; Lin Yan; Theresa M. Kelly; Margaret van Heek; James Mu; Margaret Wu; Ge Dai; Yan Cui; Yonghua Zhu; Ester Carballo-Jane; Vijay Bhasker G. Reddy; Peter Zafian; Pei Huo; Shuai Shi; Valentyn Antochshuk; Aimie M. Ogawa; Franklin Liu; Sandra C. Souza; Wolfgang Seghezzi; Joseph L. Duffy; Mark D. Erion; Ravi P. Nargund; David E. Kelley

In the above-mentioned article, several scientists were erroneously omitted from the acknowledgments section. The authors wish to acknowledge Hsuan-shen Chen, Huaibing He, Tina …


Biochemistry | 2002

Two tarantula peptides inhibit activation of multiple sodium channels.

Richard E. Middleton; Vivien A. Warren; Richard L. Kraus; Jeremy Hwang; Chou J. Liu; Ge Dai; Richard M. Brochu; Martin Kohler; Ying-Duo Gao; Victor M. Garsky; Michael J. Bogusky; John T. Mehl; Charles J. Cohen; McHardy M. Smith


Biochemistry | 2005

Role of the C-terminus of the high-conductance calcium-activated potassium channel in channel structure and function.

William A. Schmalhofer; Manuel Sanchez; Ge Dai; Ashvin Dewan; Lorena Secades; Markus Hanner; Hans-Guenther Knaus; Owen B. McManus; Martin Köhler; Gregory J. Kaczorowski; Maria L. Garcia

Researchain Logo
Decentralizing Knowledge