Network


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

Hotspot


Dive into the research topics where John A. Butera is active.

Publication


Featured researches published by John A. Butera.


Journal of Pharmacology and Experimental Therapeutics | 2011

The Metabotropic Glutamate Receptor 7 Allosteric Modulator AMN082: A Monoaminergic Agent in Disguise?

Stacey J. Sukoff Rizzo; Sarah K. Leonard; Adam M. Gilbert; Paul Jeffrey Dollings; Deborah L. Smith; Mei-Yi Zhang; Li Di; Brian Platt; Sarah Neal; Jason M. Dwyer; Corey N. Bender; Jean Zhang; Tim Lock; Dianne Kowal; Angela Kramer; Andrew D. Randall; Christine Huselton; Karthick Vishwanathan; Susanna Y. Tse; John A. Butera; Robert H. Ring; Sharon Rosenzweig-Lipson; Zoë A. Hughes; John Dunlop

Metabotropic glutamate receptor 7 (mGluR7) remains the most elusive of the eight known mGluRs primarily because of the limited availability of tool compounds to interrogate its potential therapeutic utility. The discovery of N,N′-dibenzhydrylethane-1,2-diamine dihydrochloride (AMN082) as the first orally active, brain-penetrable, mGluR7-selective allosteric agonist by Mitsukawa and colleagues (Proc Natl Acad Sci USA 102:18712–18717, 2005) provides a means to investigate this receptor system directly. AMN082 demonstrates mGluR7 agonist activity in vitro and interestingly has a behavioral profile that supports utility across a broad spectrum of psychiatric disorders including anxiety and depression. The present studies were conducted to extend the in vitro and in vivo characterization of AMN082 by evaluating its pharmacokinetic and metabolite profile. Profiling of AMN082 in rat liver microsomes revealed rapid metabolism (t1/2 < 1 min) to a major metabolite, N-benzhydrylethane-1,2-diamine (Met-1). In vitro selectivity profiling of Met-1 demonstrated physiologically relevant transporter binding affinity at serotonin transporter (SERT), dopamine transporter (DAT), and norepinephrine transporter (NET) (323, 3020, and 3410 nM, respectively); whereas the parent compound AMN082 had appreciable affinity at NET (1385 nM). AMN082 produced antidepressant-like activity and receptor occupancy at SERT up to 4 h postdose, a time point at which AMN082 is significantly reduced in brain and plasma while the concentration of Met-1 continues to increase in brain. Acute Met-1 administration produced antidepressant-like activity as would be expected from its in vitro profile as a mixed SERT, NET, DAT inhibitor. Taken together, these data suggest that the reported in vivo actions of AMN082 should be interpreted with caution, because they may involve other mechanisms in addition to mGluR7.


European Journal of Pharmacology | 2009

Pharmacological comparison of muscarinic ligands: historical versus more recent muscarinic M1-preferring receptor agonists.

Julia N. Heinrich; John A. Butera; Tikva Carrick; Angela Kramer; Dianne Kowal; Tim Lock; Karen L. Marquis; Mark H. Pausch; Mike Popiolek; Shaiu-Ching Sun; Eugene Tseng; Albert J. Uveges; Scott Christian Mayer

In functional assay assessments using the five muscarinic receptor subtypes, a second generation of muscarinic M(1)-preferring receptor agonists [AC-42 (1), AC-260584 (2), 77-LH-28-1 (3) and LY-593039 (4)] was shown to have higher selectivity for muscarinic M(1) over M(3) receptor as compared to historical agonists [talsaclidine (8), sabcomeline (10), xanomeline (11), WAY-132983 (12), cevimeline (9) and NGX-267 (6)]. Another striking difference of these more recent compounds is their affinities for the dopamine D(2) and 5-HT(2B) receptors. Taken together, these results suggest that the newer compounds may have a greater clinical safety profile, especially with regard to muscarinic M(3) receptor-mediated events, than the historical agonists, but their affinities for other receptors may still compromise their use to validate the therapeutic potential of muscarinic M(1) receptor agonists.


Bioorganic & Medicinal Chemistry Letters | 2001

Synthesis and potassium channel opening activity of substituted 10H-benzo[4,5]furo[3,2-b]indole-and 5,10-dihydro-indeno[1,2-b]indole-1-carboxylic acids.

John A. Butera; Schuyler Adam Antane; Bradford H. Hirth; Joseph R. Lennox; Jeffrey H. Sheldon; N. Wesley Norton; Dawn Warga; Thomas M. Argentieri

Compounds in a structurally novel series of substituted 10H-benzo[4,5]furo[3,2-b]indole-1-carboxylic acids and related 5,10-dihydro-indeno[1,2-b]indole-1-carboxylic acids were prepared and shown to possess potent, bladder-selective smooth muscle relaxant properties and thus are potentially useful for the treatment of urge urinary incontinence. Electrophysiological studies using rat detrusor myocytes have demonstrated that prototype compound 7 produces a significant increase in hyperpolarizing current, which is iberiotoxin (IbTx)-reversed, thus consistent with activation of the large-conductance Ca(2+)-activated potassium channel (BK(Ca)).


Journal of Pharmacology and Experimental Therapeutics | 2009

The Gap Junction Modifier, GAP-134 [(2S,4R)-1-(2-Aminoacetyl)-4-benzamido-pyrrolidine-2-carboxylic Acid], Improves Conduction and Reduces Atrial Fibrillation/Flutter in the Canine Sterile Pericarditis Model

Eric I. Rossman; Kun Liu; Gwen A. Morgan; Robert E. Swillo; Julie A. Krueger; Stephen J. Gardell; John A. Butera; Matthew Gruver; Joel Kantrowitz; Hal S. Feldman; Jørgen Søberg Petersen; Ketil Haugan; James K. Hennan

Gap junction uncoupling can alter conduction pathways and promote cardiac re-entry mechanisms that potentiate many supraventricular arrhythmias, such as atrial fibrillation (AF) and atrial flutter (AFL). Our objective was to determine whether GAP-134 [(2S,4R)-1-(2-aminoacetyl)-4-benzamido-pyrrolidine-2-carboxylic acid], a small dipeptide gap junction modifier, can improve conduction and ultimately prevent AF/AFL. In rat atrial strips subjected to metabolic stress, GAP-134 prevented significantly conduction velocity slowing at 10 nM compared with vehicle (p < 0.01). In the canine sterile pericarditis model, conduction time (CT; n = 5), atrial effective refractory period (AERP; n = 3), and AF/AFL duration/inducibility (n = 16) were measured 2 to 3 days postoperatively in conscious dogs. CT was significantly faster after GAP-134 infusion (average plasma concentration, 250 nM) at cycle lengths of 300 ms (66.2 ± 1.0 versus 62.0 ± 1.0 ms; p < 0.001) and 200 ms (64.4 ± 0.9 versus 61.0 ± 1.3 ms; p < 0.001). No significant changes in AERP were noted after GAP-134 infusion. The mean number of AF/AFL inductions per animal was significantly decreased after GAP-134 infusion (2.7 ± 0.6 versus 1.6 ± 0.8; p < 0.01), with total AF/AFL burden being decreased from 12,280 to 6063 s. Western blot experiments showed no change in connexin 43 expression. At concentrations exceeding those described in the AF/AFL experiments, GAP-134 had no effect on heart rate, blood pressure, or any electrocardiogram parameters. In conclusion, GAP-134 shows consistent efficacy on measures of conduction and AF/AFL inducibility in the canine sterile pericarditis model. These findings, along with its oral bioavailability, underscore its potential antiarrhythmic efficacy.


Journal of Medicinal Chemistry | 2009

Discovery of (2S,4R)-1-(2-Aminoacetyl)-4-benzamidopyrrolidine-2-carboxylic Acid Hydrochloride (GAP-134)13, an Orally Active Small Molecule Gap-Junction Modifier for the Treatment of Atrial Fibrillation

John A. Butera; Bjarne Due Larsen; James K. Hennan; Edward H. Kerns; Li Di; Asaf Alimardanov; Robert E. Swillo; Gwen A. Morgan; Kun Liu; Qiang Wang; Eric I. Rossman; Rayomand J. Unwalla; Leonard A. McDonald; Christine Huselton; Jørgen Søberg Petersen

Rotigaptide (3) is an antiarrhythmic peptide that improves cardiac conduction by modifying gap-junction communication. Small molecule gap-junction modifiers with improved physical properties were identified from a Zealand Pharma peptide library using pharmaceutical profiling, established SAR around 3, and a putative pharmacophore model for rotigaptide. Activity of the compounds was confirmed in a mouse cardiac conduction block model of arrhythmia. Dipeptide 9f (GAP-134) was identified as a potent, orally active gap-junction modifier for clinical development.


Journal of Medicinal Chemistry | 2009

Prodrugs of Perzinfotel with Improved Oral Bioavailability

Reinhardt Bernhard Baudy; John A. Butera; Magid Abou-Gharbia; Hong Chen; Boyd L. Harrison; Uday Jain; Ronald L. Magolda; Jean Y. Sze; Michael R. Brandt; Terri Cummons; Diane Kowal; Menelas N. Pangalos; Bojana Zupan; Matthew Hoffmann; Michael May; Cheryl P. Mugford; Jeffrey D. Kennedy; Wayne E. Childers

Previous studies with perzinfotel (1), a potent, selective, competitive NMDA receptor antagonist, showed it to be efficacious in inflammatory and neuropathic pain models. To increase the low oral bioavailability of 1 (3-5%), prodrug derivatives (3a-h) were synthesized and evaluated. The oxymethylene-spaced diphenyl analogue 3a demonstrated good stability at acidic and neutral pH, as well as in simulated gastric fluid. In rat plasma, 3a was rapidly converted to 1 via 2a. Pharmacokinetic studies indicated that the amount of systemic exposure of 1 produced by a 10 mg/kg oral dose of 3a was 2.5-fold greater than that produced by a 30 mg/kg oral dose of 1. Consistent with these results, 3a was significantly more potent and had a longer duration of activity than 1 following oral administration in a rodent model of inflammatory pain. Taken together, these results demonstrate that an oxymethylene-spaced prodrug approach increased the bioavailability of 1.


Bioorganic & Medicinal Chemistry Letters | 2010

Identification of pyridazino[4,5-b]indolizines as selective PDE4B inhibitors.

Andrew Forrest Donnell; Paul Jeffrey Dollings; John A. Butera; Arlene Dietrich; Kerri Lipinski; Afshin Ghavami; Warren D. Hirst

Substituted pyridazino[4,5-b]indolizines were identified as potent and selective PDE4B inhibitors. We describe the structure-activity relationships generated around an HTS hit that led to a series of compounds with low nanomolar affinity for PDE4B and high selectivity over the PDE4D subtype.


Bioorganic & Medicinal Chemistry | 2009

Identification and SAR of squarate inhibitors of mitogen activated protein kinase-activated protein kinase 2 (MK-2)

Frank Lovering; Steve Kirincich; Weiheng Wang; Kerry Combs; Lynn Resnick; Joan Eileen Sabalski; John A. Butera; Julie Liu; Kevin D. Parris; Jean-Baptiste Telliez

A novel series of inhibitors for mitogen activated protein kinase-activated protein kinase 2 (MK-2) are reported. These squarate based inhibitors were identified via a high-throughput screen. An MK2 co-structure with the starting ligand was obtained and a structure based approach was followed to optimize potency and selectivity.


Journal of Medicinal Chemistry | 2013

Phenotypic screening as a strategic component of drug discovery programs targeting novel antiparasitic and antimycobacterial agents: an editorial.

John A. Butera

Drug development for neglected tropical diseases, defined as a collection of infectious diseases affecting over 4 billion people worldwide (especially endemic to poverty-stricken populations in underdeveloped regions of Africa, Asia, the Pacific Rim, and Latin America), has been underfunded and stagnant. A much needed resurgence of R&D activity in this area is currently developing. Target-directed screening and whole-cell phenotypic screening represent two complementary approaches to discover viable new starting point scaffolds for medicinal chemistry optimization. This editorial will provide introductory comments to a series of six miniperspectives that focus on the special challenges faced by scientists in discovering potential new chemical leads that could be optimized into promising clinical candidates for neglected tropical diseases.


The International Journal of Neuropsychopharmacology | 2010

Preclinical characterization of BRL 44408: antidepressant- and analgesic-like activity through selective α2A-adrenoceptor antagonism

Jason M. Dwyer; Brian Platt; Stacey J. Sukoff Rizzo; Claudine Pulicicchio; Caitlin Wantuch; Mei-Yi Zhang; Terri Cummons; Liza Leventhal; Corey N. Bender; Jean Zhang; Dianne Kowal; Shendi Lu; S. Johannes R. Rajarao; Deborah L. Smith; Adam D. Shilling; Jianyao Wang; John A. Butera; Lynn Resnick; Sharon Rosenzweig-Lipson; Lee E. Schechter; Chad E. Beyer

Biogenic amines such as norepinephrine, dopamine, and serotonin play a well-described role in the treatment of mood disorders and some types of pain. As alpha2A-adrenoceptors regulate the release of these neurotransmitters, we examined the therapeutic potential of BRL 44408, a potent (Ki=8.5 nM) and selective (>50-fold) alpha2A-adrenoceptor antagonist (K(B)=7.9 nM). In rats, BRL 44408 penetrated the central nervous system resulting in peak brain and plasma concentrations of 586 ng/g and 1124 ng/ml, respectively. In a pharmacodynamic assay, pretreatment with BRL 44408 to rats responding under a fixed-ratio 30 operant response paradigm resulted in a rightward shift of the clonidine dose-response curve, an effect indicative of alpha2-adrenoceptor antagonism in vivo. Consistent with presynaptic autoreceptor antagonism and tonic regulation of neurotransmitter release, acute administration of BRL 44408 elevated extracellular concentrations of norepinephrine and dopamine, but not serotonin, in the medial prefrontal cortex. Additionally, BRL 44408, probably by inhibiting alpha2A heteroceptors, produced a significant increase in cortical levels of acetylcholine. In the forced swim test and schedule-induced polydipsia assay, BRL 44408 produced an antidepressant-like response by dose-dependently decreasing immobility time and adjunctive water intake, respectively, while in a model of visceral pain, BRL 44408 exhibited analgesic activity by decreasing para-phenylquinone (PPQ)-induced abdominal stretching. Finally, BRL 44408 did not produce deficits in overall motor coordination nor alter general locomotor activity. This preclinical characterization of the neurochemical and behavioural profile of BRL 44408 suggests that selective antagonism of alpha2A-adrenoceptors may represent an effective treatment strategy for mood disorders and visceral pain.

Collaboration


Dive into the John A. Butera's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

James K. Hennan

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jørgen Søberg Petersen

University Medical Center New Orleans

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Edward H. Kerns

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge