Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nucleic Acids Res ; 49(4): 2141-2160, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33524148

ABSTRACT

The tRNA modification N6-threonylcarbamoyladenosine (t6A) is universally conserved in all organisms. In bacteria, the biosynthesis of t6A requires four proteins (TsaBCDE) that catalyze the formation of t6A via the unstable intermediate l-threonylcarbamoyl-adenylate (TC-AMP). While the formation and stability of this intermediate has been studied in detail, the mechanism of its transfer to A37 in tRNA is poorly understood. To investigate this step, the structure of the TsaBD heterodimer from Escherichia coli has been solved bound to a stable phosphonate isosteric mimic of TC-AMP. The phosphonate inhibits t6A synthesis in vitro with an IC50 value of 1.3 µM in the presence of millimolar ATP and L-threonine. The inhibitor binds to TsaBD by coordination to the active site Zn atom via an oxygen atom from both the phosphonate and the carboxylate moieties. The bound conformation of the inhibitor suggests that the catalysis exploits a putative oxyanion hole created by a conserved active site loop of TsaD and that the metal essentially serves as a binding scaffold for the intermediate. The phosphonate bound crystal structure should be useful for the rational design of potent, drug-like small molecule inhibitors as mechanistic probes or potentially novel antibiotics.


Subject(s)
Adenosine/analogs & derivatives , Escherichia coli Proteins/chemistry , RNA, Transfer/metabolism , Adenosine/biosynthesis , Adenosine/chemistry , Catalytic Domain , Escherichia coli/enzymology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Organophosphonates/chemistry , Organophosphonates/pharmacology , Protein Multimerization , RNA, Transfer/chemistry
2.
Bioorg Med Chem Lett ; 37: 127838, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33556572

ABSTRACT

A high throughput screen (HTS) identified a novel, but weak (EC50 = 6.2 µM, 97% Glu Max) mGlu4 PAM chemotype based on a 1,4-thiazepane core, VU0544412. Reaction development and chemical optimization delivered a potent mGlu4 PAM VU6022296 (EC50 = 32.8 nM, 108% Glu Max) with good CNS penetration (Kp = 0.45, Kp,uu = 0.70) and enantiopreference. Finally, VU6022296 displayed robust, dose-dependent efficacy in reversing Haloperidol-Induced Catalepsy (HIC), a rodent preclinical Parkinson's disease model.


Subject(s)
Catalepsy/drug therapy , Disease Models, Animal , Drug Discovery , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Catalepsy/chemically induced , Dose-Response Relationship, Drug , Haloperidol , Mice , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Receptors, Metabotropic Glutamate/metabolism , Structure-Activity Relationship
3.
Bioorg Med Chem Lett ; 30(13): 127212, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32371100

ABSTRACT

This Letter details our ongoing efforts to develop selective positive allosteric modulators (PAMs) of the mGlu2/4 heterodimeric receptor that exists in the CNS and may represent a novel drug target to modulate the glutamatergic system. As multiple hit-to-lead campaigns from HTS hits failed to produce selective small molecule mGlu2/4 heterodimer PAMs, we were inspired by the work of Portoghese to synthesize and evaluate a set of nine bivalent tethered ligands (possessing an mGlu2 PAM at one terminus and an mGlu4 PAM at the other). Utilizing G protein-Inwardly Rectifying Potassium (GIRK) channel functional assays, we found that the tethered ligands displayed PAM activity in a cell line co-expressing both mGlu2 and mGlu4 but also in cells expressing mGlu2 or mGlu4 alone. In a CODA-RET assay, one of the tethered ligands potentiated mGlu2/4 heterodimers; however, another compound displayed 75-fold preference for the mGlu2/2 homodimer over heterodimeric mGlu2/4 or homomeric mGlu4/4. This work highlights the development of mGlu receptor PAMs with homodimer/heterodimer preference and expands the potential for PAMs as tethered ligands beyond the more classical antagonists and NAMs.


Subject(s)
Benzamides/pharmacology , Indans/pharmacology , Receptors, Metabotropic Glutamate/agonists , Allosteric Regulation/drug effects , Animals , Benzamides/chemical synthesis , HEK293 Cells , Humans , Indans/chemical synthesis , Ligands , Molecular Structure , Protein Structure, Quaternary , Rats , Receptors, Metabotropic Glutamate/chemistry , Structure-Activity Relationship
4.
J Org Chem ; 84(18): 12187-12191, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31436090

ABSTRACT

A concise formal total synthesis of pericoannosin A, by the synthesis of an advanced intermediate of pericoannosin A, was achieved in eight steps from commercially available isoprene in a 21.7% overall yield. Key transformations for this expedited route include an enantioselective organocatalytic Diels-Alder reaction to construct the C ring, a diastereoselective reduction (under Felkin-Ahn control), and a hydroboration/oxidation sequence for chain homologation. This work represents the second synthetic effort toward pericoannosin A, the only reported natural product based on a hexahydro-1H-isochromen-5-isobutylpyrrolidin-2-one core.

5.
J Pharmacol Exp Ther ; 370(3): 350-359, 2019 09.
Article in English | MEDLINE | ID: mdl-31201216

ABSTRACT

Glucose-stimulated insulin secretion from pancreatic ß-cells is controlled by ATP-regulated potassium (KATP) channels composed of Kir6.2 and sulfonylurea receptor 1 (SUR1) subunits. The KATP channel-opener diazoxide is FDA-approved for treating hyperinsulinism and hypoglycemia but suffers from off-target effects on vascular KATP channels and other ion channels. The development of more specific openers would provide critically needed tool compounds for probing the therapeutic potential of Kir6.2/SUR1 activation. Here, we characterize a novel scaffold activator of Kir6.2/SUR1 that our group recently discovered in a high-throughput screen. Optimization efforts with medicinal chemistry identified key structural elements that are essential for VU0071063-dependent opening of Kir6.2/SUR1. VU0071063 has no effects on heterologously expressed Kir6.1/SUR2B channels or ductus arteriole tone, indicating it does not open vascular KATP channels. VU0071063 induces hyperpolarization of ß-cell membrane potential and inhibits insulin secretion more potently than diazoxide. VU0071063 exhibits metabolic and pharmacokinetic properties that are favorable for an in vivo probe and is brain penetrant. Administration of VU0071063 inhibits glucose-stimulated insulin secretion and glucose-lowering in mice. Taken together, these studies indicate that VU0071063 is a more potent and specific opener of Kir6.2/SUR1 than diazoxide and should be useful as an in vitro and in vivo tool compound for investigating the therapeutic potential of Kir6.2/SUR1 expressed in the pancreas and brain.


Subject(s)
Ion Channel Gating/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , Sulfonylurea Receptors/metabolism , Xanthines/pharmacology , Xanthines/pharmacokinetics , Animals , Ductus Arteriosus/drug effects , Ductus Arteriosus/physiology , Glucose/pharmacology , HEK293 Cells , Humans , Insulin Secretion/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mice , Structure-Activity Relationship , Vasodilation/drug effects , Xanthines/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...