Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
SLAS Discov ; 26(8): 1004-1013, 2021 09.
Article in English | MEDLINE | ID: mdl-33980065

ABSTRACT

The ability to detect and characterize drug binding to a target protein is of high priority in drug discovery research. However, there are inherent challenges when the target of interest is an integral membrane protein (IMP). Assuming successful purification of the IMP, traditional approaches for measuring binding such as surface plasmon resonance (SPR) and fluorescence resonance energy transfer (FRET) have been proven valuable. However, the mass dependence of SPR signals may preclude the detection of binding events when the ligand has a significantly smaller mass than the target protein. In FRET-based experiments, protein labeling through modification may inadvertently alter protein dynamics. Graphene Bio-Electronic Sensing Technology (GBEST) aims to overcome these challenges. Label-free characterization takes place in a microfluidic chamber wherein a fluid lipid membrane is reconstituted directly above the GBEST sensor surface. By leveraging the high conductivity, sensitivity, and electrical properties of monolayer graphene, minute changes in electrostatic charges arising from the binding and unbinding of a ligand to a native IMP target can be detected in real time and in a mass-independent manner. Using crude membrane fractions prepared from cells overexpressing monocarboxylate transporter 1 (MCT1), we demonstrate the ability to (1) form a fluid lipid bilayer enriched with MCT1 directly on top of the GBEST sensor and (2) obtain kinetic binding data for an anti-MCT1 antibody. Further development of this novel technology will enable characterization of target engagement by both low- and high-molecular-weight drug candidates to native IMP targets in a physiologically relevant membrane environment.


Subject(s)
Biosensing Techniques , Drug Discovery/methods , Electrochemical Techniques , Ligands , Membrane Transport Proteins/chemistry , Graphite , Humans , Kinetics , Membrane Transport Proteins/metabolism , Protein Binding , Surface Plasmon Resonance/methods
3.
SLAS Discov ; 25(5): 434-446, 2020 06.
Article in English | MEDLINE | ID: mdl-32292096

ABSTRACT

The voltage-gated sodium channel Nav1.7 is a genetically validated target for pain; pharmacological blockers are promising as a new class of nonaddictive therapeutics. The search for Nav1.7 subtype selective inhibitors requires a reliable, scalable, and sensitive assay. Previously, we developed an all-optical electrophysiology (Optopatch) Spiking HEK platform to study activity-dependent modulation of Nav1.7 in a format compatible with high-throughput screening. In this study, we benchmarked the Optopatch Spiking HEK assay with an existing validated automated electrophysiology assay on the IonWorks Barracuda (IWB) platform. In a pilot screen of 3520 compounds, which included compound plates from a random library as well as compound plates enriched for Nav1.7 inhibitors, the Optopatch Spiking HEK assay identified 174 hits, of which 143 were confirmed by IWB. The Optopatch Spiking HEK assay maintained the high reliability afforded by traditional fluorescent assays and further demonstrated comparable sensitivity to IWB measurements. We speculate that the Optopatch assay could provide an affordable high-throughput screening platform to identify novel Nav1.7 subtype selective inhibitors with diverse mechanisms of action, if coupled with a multiwell parallel optogenetic recording instrument.


Subject(s)
High-Throughput Screening Assays , NAV1.7 Voltage-Gated Sodium Channel/drug effects , Patch-Clamp Techniques , Voltage-Gated Sodium Channel Blockers/isolation & purification , Animals , CHO Cells , Cricetulus , Electrophysiological Phenomena , Electrophysiology , HEK293 Cells , Humans , NAV1.7 Voltage-Gated Sodium Channel/genetics
4.
SLAS Discov ; 25(2): 215-222, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31849250

ABSTRACT

The real-time quantification of target engagement (TE) by small-molecule ligands in living cells remains technically challenging. Systematic quantification of such interactions in a high-throughput setting holds promise for identification of target-specific, potent small molecules within a pathophysiological and biologically relevant cellular context. The salt-inducible kinases (SIKs) belong to a subfamily of the AMP-activated protein kinase (AMPK) family and are composed of three isoforms in humans (SIK1, SIK2, and SIK3). They modulate the production of pro- and anti-inflammatory cytokines in immune cells. Although pan-SIK inhibitors are sufficient to reverse SIK-dependent inflammatory responses, the apparent toxicity associated with SIK3 inhibition suggests that isoform-specific inhibition is required to realize therapeutic benefit with acceptable safety margins. Here, we used the NanoBRET TE intracellular kinase assay, a sensitive energy transfer technique, to directly measure molecular proximity and quantify TE in HEK293T cells overexpressing SIK2 or SIK3. Our 384-well high-throughput screening of 530 compounds demonstrates that the NanoBRET TE intracellular kinase assay was sensitive and robust enough to reveal differential engagement of candidate compounds with the two SIK isoforms and further highlights the feasibility of high-throughput implementation of NanoBRET TE intracellular kinase assays for target-driven small-molecule screening.


Subject(s)
Phosphotransferases/isolation & purification , Protein Isoforms/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fluorescence Resonance Energy Transfer , Gene Expression Regulation, Enzymologic/drug effects , HEK293 Cells , High-Throughput Screening Assays/methods , Humans , Phosphotransferases/genetics , Protein Isoforms/antagonists & inhibitors , Protein Kinases/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors
5.
Front Pharmacol ; 10: 51, 2019.
Article in English | MEDLINE | ID: mdl-30837866

ABSTRACT

There is an unmet need in severe asthma where approximately 40% of patients exhibit poor ß-agonist responsiveness, suffer daily symptoms and show frequent exacerbations. Antagonists of the Ca2+-activated Cl- channel, TMEM16A, offers a new mechanism to bronchodilate airways and block the multiple contractiles operating in severe disease. To identify TMEM16A antagonists we screened a library of ∼580,000 compounds. The anthelmintics niclosamide, nitazoxanide, and related compounds were identified as potent TMEM16A antagonists that blocked airway smooth muscle depolarization and contraction. To evaluate whether TMEM16A antagonists resist use- and inflammatory-desensitization pathways limiting ß-agonist action, we tested their efficacy under harsh conditions using maximally contracted airways or airways pretreated with a cytokine cocktail. Stunningly, TMEM16A antagonists fully bronchodilated airways, while the ß-agonist isoproterenol showed only partial effects. Thus, antagonists of TMEM16A and repositioning of niclosamide and nitazoxanide represent an important additional treatment for patients with severe asthma and COPD that is poorly controlled with existing therapies. It is of note that drug repurposing has also attracted wide interest in niclosamide and nitazoxanide as a new treatment for cancer and infectious disease. For the first time we identify TMEM16A as a molecular target for these drugs and thus provide fresh insights into their mechanism for the treatment of these disorders in addition to respiratory disease.

6.
J Biomol Screen ; 20(4): 433-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25805607

ABSTRACT

In this special issue of the Journal of biomolecular screening, we have assembled a series of articles that exemplify and discuss various aspects and challenges associated with the discovery, development, and manufacture of biologics with an emphasis on those topics that we feel will appeal to readers of this journal. We hope you enjoy them!


Subject(s)
Antibodies/therapeutic use , Biological Products/therapeutic use , Publishing
7.
Expert Opin Ther Targets ; 11(3): 265-71, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17298286

ABSTRACT

Many human diseases result from over- or underactivity in one or more critical physiologic systems. One of the foremost challenges in modern drug discovery is the identification and selection of cellular proteins that can be specifically targeted with therapeutic agents in order to normalize aberrant processes/systems. Suitable drug targets must be validated in the human disease state and ideally, the targeted protein will fulfill similar physiologic and pathologic functions in humans and at least one animal species so that in vivo efficacy and toxicology assays with some predictive clinical relevance may be developed. Nowadays, drug targets must also be amenable to high-throughput screening so that novel molecules, which are capable of modifying cellular protein function, can be identified in large libraries of compounds. Voltage-gated ion channels satisfy many of these requirements and, as a class, are viewed as promising drug targets. Nevertheless, despite their relevance to human disease, voltage-gated ion channels remain considerably underexploited. Therein lie some of the opportunities and advantages associated with voltage-gated ion channels as drug targets.


Subject(s)
Ion Channels/physiology , Animals , Drug Design , Drug Therapy , Humans , Ion Channel Gating
8.
Neuropsychiatr Dis Treat ; 3(1): 69-85, 2007 Feb.
Article in English | MEDLINE | ID: mdl-19300539

ABSTRACT

Ziconotide is a powerful analgesic drug that has a unique mechanism of action involving potent and selective block of N-type calcium channels, which control neurotransmission at many synapses. The analgesic efficacy of ziconotide likely results from its ability to interrupt pain signaling at the level of the spinal cord. Ziconotide is a peptidic drug and has been approved for the treatment of severe chronic pain in patients only when administered by the intrathecal route. Importantly, prolonged administration of ziconotide does not lead to the development of addiction or tolerance. The current review discusses the various studies that have addressed the in vitro biochemical and electrophysiological actions of ziconotide as well as the numerous pre-clinical studies that were conducted to elucidate its antinociceptive mechanism of action in animals. In addition, this review considers the pivotal Phase 3 (and other) clinical trials that were conducted in support of ziconotide's approval for the treatment of severe chronic pain and tries to offer some insights regarding the future discovery and development of newer analgesic drugs that would act by a similar mechanism to ziconotide but which might offer improved safety, tolerability and ease of use.

9.
CNS Neurol Disord Drug Targets ; 5(6): 587-603, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17168744

ABSTRACT

Voltage-gated calcium channels are found in the plasma membrane of many excitable and non-excitable cells. When open, they permit influx of calcium, which acts as a second messenger to initiate diverse physiological cellular processes. Ten unique alpha1 subunits, grouped in three families (CaV1, CaV2, and CaV3), encode biophysically and pharmacologically distinct low-voltage-activated T-type and high-voltage-activated L-type, N-type, P/Q-type, and R-type calcium channels. T-type calcium channels are found in neurons where they generate low-threshold calcium spikes and influence action potential firing patterns, in heart cells where they influence pacemaking and impulse conduction, in smooth muscle cells where they regulate myogenic tone and proliferation, in endocrine cells where they regulate hormone secretion, and in sperm where they regulate the acrosome reaction. Validation of T-type calcium channels in disease is based on an abundance of data pertaining to clinical efficacy of T-type calcium channel blockers in certain human conditions as well as information relating to the distribution, functional properties, and physiological roles of these channels. This review focuses on the cellular and molecular pharmacology of T-type calcium channels. It describes novel research approaches to discover potent and selective T-type calcium channel modulators as potential drugs for treating human disease and as tools for understanding better the physiological roles of T-type calcium channels.


Subject(s)
Calcium Channels, T-Type/drug effects , Animals , Anticonvulsants/pharmacology , Antihypertensive Agents/pharmacology , Antipsychotic Agents/pharmacology , Drug Design , Humans
10.
Drug Discov Today ; 11(5-6): 245-53, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16580601

ABSTRACT

Severe chronic pain afflicts a large number of people worldwide but satisfactory relief from such pain is difficult to achieve with drugs that are currently available, and so there is a great need for the development of new, efficacious and safe analgesics. Voltage-gated calcium-permeable ion channels are multi-subunit complexes that regulate neuronal excitability, action-potential firing patterns and neurotransmission in nociceptive pathways. Although multiple subtypes of voltage-gated calcium channels exist, pharmacological and ion-channel gene knockdown approaches in animals have revealed N-type and T-type calcium channels to be particularly attractive molecular targets for the discovery and development of new analgesic drugs. The recent approval of Prialt (Elan Pharmaceuticals) provides the ultimate target validation for N-type calcium channels, namely proof that they are key regulators of nociceptive signaling in humans.


Subject(s)
Analgesics/therapeutic use , Calcium Channels, N-Type/drug effects , Calcium Channels, T-Type/drug effects , Pain/drug therapy , Acute Disease , Analgesics/pharmacology , Animals , Chronic Disease , Humans , Pain/physiopathology , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/physiopathology , omega-Conotoxins/pharmacology , omega-Conotoxins/therapeutic use
11.
Curr Drug Targets CNS Neurol Disord ; 3(6): 457-78, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15578964

ABSTRACT

This review focuses on the importance of voltage-gated calcium channels in modulating and controlling the function of peripheral and central neurons involved in nociceptive processing. We describe the different families of voltage-gated calcium channels that are expressed in pain pathway neurons, how the expression levels of calcium channel currents change in chronic pain conditions, and the validation of N-type, T-type, and P-type calcium channels as targets for the treatment of pain. The molecular mechanism of action is reviewed for the most prominent calcium channel-targeted drugs including gabapentin and ziconotide as well as antiepileptics administered off-label for the treatment of pain. We discuss how the major genetic, functional, and pharmacological differences between subtypes of neuronal calcium channels can be leveraged to identify new molecular targets and to discover and develop new therapeutic agents for the treatment of chronic pain syndromes.


Subject(s)
Afferent Pathways/physiopathology , Calcium Channels/genetics , Neurons, Afferent/physiology , Nociceptors/physiology , Pain, Intractable/physiopathology , Afferent Pathways/drug effects , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channels/drug effects , Drug Design , Humans , Neurons, Afferent/drug effects , Nociceptors/drug effects , Pain, Intractable/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL