Network


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

Hotspot


Dive into the research topics where Earl J. Gubbins is active.

Publication


Featured researches published by Earl J. Gubbins.


The Journal of Neuroscience | 2007

Broad-Spectrum Efficacy across Cognitive Domains by α7 Nicotinic Acetylcholine Receptor Agonism Correlates with Activation of ERK1/2 and CREB Phosphorylation Pathways

Robert S. Bitner; William H. Bunnelle; David J. Anderson; Clark A. Briggs; Jerry J. Buccafusco; Peter Curzon; Michael W. Decker; Jennifer M. Frost; Jens Halvard Grønlien; Earl J. Gubbins; Jinhe Li; John Malysz; Stella Markosyan; Kennan C. Marsh; Michael D. Meyer; Arthur L. Nikkel; Richard J. Radek; Holly M. Robb; Daniel B. Timmermann; James P. Sullivan; Murali Gopalakrishnan

The α7 nicotinic acetylcholine receptor (nAChR) plays an important role in cognitive processes and may represent a drug target for treating cognitive deficits in neurodegenerative and psychiatric disorders. In the present study, we used a novel α7 nAChR-selective agonist, 2-methyl-5-(6-phenyl-pyridazin-3-yl)-octahydro-pyrrolo[3,4-c]pyrrole (A-582941) to interrogate cognitive efficacy, as well as examine potential cellular mechanisms of cognition. Exhibiting high affinity to native rat (K i = 10.8 nm) and human (K i = 16.7 nm) α7 nAChRs, A-582941 enhanced cognitive performance in behavioral assays including the monkey delayed matching-to-sample, rat social recognition, and mouse inhibitory avoidance models that capture domains of working memory, short-term recognition memory, and long-term memory consolidation, respectively. In addition, A-582941 normalized sensory gating deficits induced by the α7 nAChR antagonist methyllycaconitine in rats, and in DBA/2 mice that exhibit a natural sensory gating deficit. Examination of signaling pathways known to be involved in cognitive function revealed that α7 nAChR agonism increased extracellular-signal regulated kinase 1/2 (ERK1/2) phosphorylation in PC12 cells. Furthermore, increases in ERK1/2 and cAMP response element-binding protein (CREB) phosphorylation were observed in mouse cingulate cortex and/or hippocampus after acute A-582941 administration producing plasma concentrations in the range of α7 binding affinities and behavioral efficacious doses. The MEK inhibitor SL327 completely blocked α7 agonist-evoked ERK1/2 phosphorylation. Our results demonstrate that α7 nAChR agonism can lead to broad-spectrum efficacy in animal models at doses that enhance ERK1/2 and CREB phosphorylation/activation and may represent a mechanism that offers potential to improve cognitive deficits associated with neurodegenerative and psychiatric diseases, such as Alzheimers disease and schizophrenia.


Journal of Biological Chemistry | 2001

Potent Inhibition of NFAT Activation and T Cell Cytokine Production by Novel Low Molecular Weight Pyrazole Compounds

James M. Trevillyan; X. Grace Chiou; Yung-Wu Chen; Stephen J. Ballaron; Michael P. Sheets; Morey L. Smith; Paul E. Wiedeman; Usha Warrior; Julie Wilkins; Earl J. Gubbins; Gerard D. Gagne; Jane A. Fagerland; George W. Carter; Jay R. Luly; Karl W. Mollison; Stevan W. Djuric

NFAT (nuclearfactor of activated T cell) proteins are expressed in most immune system cells and regulate the transcription of cytokine genes critical for the immune response. The activity of NFAT proteins is tightly regulated by the Ca2+/calmodulin-dependent protein phosphatase 2B/calcineurin (CaN). Dephosphorylation of NFAT by CaN is required for NFAT nuclear localization. Current immunosuppressive drugs such as cyclosporin A and FK506 block CaN activity thus inhibiting nuclear translocation of NFAT and consequent cytokine gene transcription. The inhibition of CaN in cells outside of the immune system may contribute to the toxicities associated with cyclosporin A therapy. In a search for safer immunosuppressive drugs, we identified a series of 3,5-bistrifluoromethyl pyrazole (BTP) derivatives that block Th1 and Th2 cytokine gene transcription. The BTP compounds block the activation-dependent nuclear localization of NFAT as determined by electrophoretic mobility shift assays. Confocal microscopy of cells expressing fluorescent-tagged NFAT confirmed that the BTP compounds block calcium-induced movement of NFAT from the cytosol to the nucleus. Inhibition of NFAT was selective because the BTP compounds did not affect the activation of NF-κB and AP-1 transcription factors. Treatment of intact T cells with the BTP compounds prior to calcium ionophore-induced activation of CaN caused NFAT to remain in a highly phosphorylated state. However, the BTP compounds did not directly inhibit the dephosphorylation of NFAT by CaNin vitro, nor did the drugs block the dephosphorylation of other CaN substrates including the type II regulatory subunit of protein kinase A and the transcription factor Elk-1. The data suggest that the BTP compounds cause NFAT to be maintained in the cytosol in a phosphorylated state and block the nuclear import of NFAT and, hence, NFAT-dependent cytokine gene transcription by a mechanism other than direct inhibition of CaN phosphatase activity. The novel inhibitors described herein will be useful in better defining the cellular regulation of NFAT activation and may lead to identification of new therapeutic targets for the treatment of autoimmune disease and transplant rejection.


Molecular Pharmacology | 2008

Positive Allosteric Modulation of the α7 Nicotinic Acetylcholine Receptor : Ligand Interactions with Distinct Binding Sites and Evidence for a Prominent Role of the M2-M3 Segment

Daniel Bertrand; Sonia Bertrand; Steven Cassar; Earl J. Gubbins; Jinhe Li; Murali Gopalakrishnan

The α7 nicotinic acetylcholine receptor (nAChR), a homopentameric, rapidly activating and desensitizing ligand-gated ion channel with relatively high degree of calcium permeability, is expressed in the mammalian central nervous system, including regions associated with cognitive processing. Selective agonists targeting the α7 nAChR have shown efficacy in animal models of cognitive dysfunction. Use of positive allosteric modulators selective for the α7 receptor is another strategy that is envisaged in the design of active compounds aiming at improving attention and cognitive dysfunction. The recent discovery of novel positive allosteric modulators such as 1-(5-chloro-2-hydroxyphenyl)-3-(2-chloro-5-trifluoromethylphenyl)urea (NS-1738) and 1-(5-chloro-2,4-dimethoxyphenyl)-3-(5-methylisoxazol-3-yl)urea (PNU-120596) that are selective for the α7 nAChRs but display significant phenotypic differences in their profile of allosteric modulation, suggests that these molecules may act at different sites on the receptor. Taking advantage of the possibility to obtain functional receptors by the fusion of proteins domains from the α7 and the 5-HT3 receptor, we examined the structural determinants required for positive allosteric modulation. This strategy revealed that the extracellular N-terminal domain of α7 plays a critical role in allosteric modulation by NS-1738. In addition, α7-5HT3 chimeras harboring the M2-M3 segment showed that spontaneous activity in response to NS-1738, which confirmed the critical contribution of this small extracellular segment in the receptor gating. In contrast to NS-1738, positive allosteric modulation by PNU-120596 could not be restored in the α7-5HT3 chimeras but was selectively observed in the reverse 5HT3-α7 chimera. All together, these data illustrate the existence of distinct allosteric binding sites with specificity of different profiles of allosteric modulators and open new possibilities to investigate the α7 receptor function.


CNS Neuroscience & Therapeutics | 2008

Preclinical Characterization of A‐582941: A Novel α7 Neuronal Nicotinic Receptor Agonist with Broad Spectrum Cognition‐Enhancing Properties

Karin R. Tietje; David J. Anderson; R. Scott Bitner; Eric A.G. Blomme; Paul J. Brackemeyer; Clark A. Briggs; Kaitlin E. Browman; Dagmar Bury; Peter Curzon; Karla Drescher; Jennifer M. Frost; Ryan M. Fryer; Gerard B. Fox; Jens Halvard Grønlien; Monika Håkerud; Earl J. Gubbins; Sabine Halm; Richard R. Harris; Rosalind Helfrich; Kathy L. Kohlhaas; Devalina Law; John Malysz; Kennan C. Marsh; Ruth L. Martin; Michael D. Meyer; Angela L. Molesky; Arthur L. Nikkel; Stephani Otte; Liping Pan; Pamela S. Puttfarcken

Among the diverse sets of nicotinic acetylcholine receptors (nAChRs), the α7 subtype is highly expressed in the hippocampus and cortex and is thought to play important roles in a variety of cognitive processes. In this review, we describe the properties of a novel biaryl diamine α7 nAChR agonist, A‐582941. A‐582941 was found to exhibit high‐affinity binding and partial agonism at α7 nAChRs, with acceptable pharmacokinetic properties and excellent distribution to the central nervous system (CNS). In vitro and in vivo studies indicated that A‐582941 activates signaling pathways known to be involved in cognitive function such as ERK1/2 and CREB phosphorylation. A‐582941 enhanced cognitive performance in behavioral models that capture domains of working memory, short‐term recognition memory, memory consolidation, and sensory gating deficit. A‐582941 exhibited a benign secondary pharmacodynamic and tolerability profile as assessed in a battery of assays of cardiovascular, gastrointestinal, and CNS function. The studies summarized in this review collectively provide preclinical validation that α7 nAChR agonism offers a mechanism with potential to improve cognitive deficits associated with various neurodegenerative and psychiatric disorders.


Journal of Pharmacology and Experimental Therapeutics | 2010

In Vitro Pharmacological Characterization of a Novel Selective α7 Neuronal Nicotinic Acetylcholine Receptor Agonist ABT-107

John Malysz; David J. Anderson; Jens Halvard Grønlien; Jianguo Ji; William H. Bunnelle; Monika Håkerud; Kirten Thorin-Hagene; Hilde Ween; Rosalind Helfrich; Min Hu; Earl J. Gubbins; Sujatha M. Gopalakrishnan; Pamela S. Puttfarcken; Clark A. Briggs; Jinhe Li; Michael D. Meyer; Tino Dyhring; Philip K. Ahring; Elsebet Ø. Nielsen; Dan Peters; Daniel B. Timmermann; Murali Gopalakrishnan

Enhancement of α7 nicotinic acetylcholine receptor (nAChR) activity is considered a therapeutic approach for ameliorating cognitive deficits present in Alzheimers disease and schizophrenia. In this study, we describe the in vitro profile of a novel selective α7 nAChR agonist, 5-(6-[(3R)-1-azabicyclo[2,2,2]oct-3-yloxy]pyridazin-3-yl)-1H-indole (ABT-107). ABT-107 displayed high affinity binding to α7 nAChRs [rat or human cortex, [3H](1S,4S)-2,2-dimethyl-5-(6-phenylpyridazin-3-yl)-5-aza-2-azoniabicyclo[2.2.1]heptane (A-585539), Ki = 0.2–0.6 nM or [3H]methyllycaconitine (MLA), 7 nM] that was at least 100-fold selective versus non-α7 nAChRs and other receptors. Functionally, ABT-107 did not evoke detectible currents in Xenopus oocytes expressing human or nonhuman α3β4, chimeric (α6/α3)β4, or 5-HT3A receptors, and weak or negligible Ca2+ responses in human neuroblastoma IMR-32 cells (α3* function) and human α4β2 and α4β4 nAChRs expressed in human embryonic kidney 293 cells. ABT-107 potently evoked human and rat α7 nAChR current responses in oocytes (EC50, 50–90 nM total charge, ∼80% normalized to acetylcholine) that were enhanced by the positive allosteric modulator (PAM) 4-[5-(4-chloro-phenyl)-2-methyl-3-propionyl-pyrrol-1-yl]-benzenesulfonamide (A-867744). In rat hippocampus, ABT-107 alone evoked α7-like currents, which were inhibited by the α7 antagonist MLA. In dentate gyrus granule cells, ABT-107 enhanced spontaneous inhibitory postsynaptic current activity when coapplied with A-867744. In the presence of an α7 PAM [A-867744 or N-[(3R)-1-azabicyclo[2.2.2]oct-3-yl]-4-chlorobenzamide hydrochloride (PNU-120596)], the addition of ABT-107 elicited MLA-sensitive α7 nAChR-mediated Ca2+ signals in IMR-32 cells and rat cortical cultures and enhanced extracellular signal-regulated kinase phosphorylation in differentiated PC-12 cells. ABT-107 was also effective in protecting rat cortical cultures against glutamate-induced toxicity. In summary, ABT-107 is a selective high affinity α7 nAChR agonist suitable for characterizing the roles of this subtype in pharmacological studies.


Brain Research | 2010

α7 nAChR-mediated activation of MAP kinase pathways in PC12 cells

Earl J. Gubbins; Murali Gopalakrishnan; Jinhe Li

The alpha7 nicotinic acetylcholine receptor (alpha7 nAChR) plays a fundamental role in Ca(2+)-dependent activation of signaling pathways that can modulate intracellular events involved in learning and memory. Activation of extracellular signal-regulated kinase-1 and -2 (ERK1/2) are well documented Ca(2+) signaling events, but these have not been well characterized in response to alpha7 nAChR-selective ligands. The present study examined activation of ERK1/2 and explored pathways leading to CREB phosphorylation utilizing alpha7 nAChR-selective ligands in PC12 cells endogenously expressing alpha7 nAChRs. Robust concentration-dependent increase in ERK1/2 phosphorylation was triggered by structurally diverse alpha7 nAChR agonists such as nicotine, choline, GTS-21, SSR-180711A and PNU-282987 in the presence of the positive allosteric modulator (PAM) PNU-120596. This effect was attenuated by selective alpha7 nAChR antagonists or by chelation of extracellular Ca(2+). ERK1/2 phosphorylation was also attenuated by inhibitors of calmodulin-dependent protein kinase II (CaMKII), p38 MAP kinase and mitogen-activated protein kinase kinase1/2 (MEK1/2), indicating the involvement of these kinases upstream of ERK1/2. This was confirmed by direct measurement of p38 MAPK and MEK1/2 phosphorylation. These data suggest that alpha7 nAChR agonist-triggered Ca(2+) transient in PC12 cells induces activation of CaMKII, leading to sequential phosphorylation of p38 MAPK, MEK1/2, ERK1/2 and CREB. Such mechanisms may endow the alpha7 nAChRs with roles in modulating Ca(2+)-dependent intracellular second messenger events implicated in diverse aspects of cognition.


Molecular Pharmacology | 2006

Untranslated region-dependent exclusive expression of high-sensitivity subforms of α4β2 and α3β2 nicotinic acetylcholine receptors

Clark A. Briggs; Earl J. Gubbins; Michael J. Marks; C. Brent Putman; Rama Thimmapaya; Michael D. Meyer; Carol S. Surowy

α4β2 nicotinic acetylcholine receptors (nAChRs) are recognized as the principal nicotine binding site in brain. Recombinant α4β2 nAChR demonstrate biphasic concentration-response relationships with low- and high-EC50 components. This study shows that untranslated regions (UTR) can influence expression of high-sensitivity subforms of α4β2 and α3β2 nAChR. Oocytes injected with α4 and β2 RNA lacking UTR expressed biphasic concentration-response relationships for acetylcholine with high-sensitivity EC50 values of 0.5 to 2.5 μM (14–24% of the population) and low-sensitivity EC50 values of 110 to 180 μM (76–86%). In contrast, message with UTR expressed exclusively the high-sensitivity α4β2 nAChR subform with an acetylcholine EC50 value of 2.2 μM. Additional studies revealed pharmacological differences between high- and low-sensitivity α4β2 subforms. Whereas the antagonists dihydro-β-erythroidine (IC50 of 3–6 nM) and methyllycaconitine (IC50 of 40–135 nM) were not selective between high- and low-sensitivity α4β2, chlorisondamine, mecamylamine, and d-tubocurarine were, respectively, 100-, 8-, and 5-fold selective for the α4β2 subform with low sensitivity to acetylcholine. Conversely, agonists that selectively activated the high-sensitivity α4β2 subform with respect to efficacy as well as potency were identified. Furthermore, two of these agonists were shown to activate mouse brain α4β2 as well as the ferret high-sensitivity α4β2 expressed in Xenopus laevis oocytes. With the use of UTR-containing RNA, exclusive expression of a novel high-sensitivity α3β2 nAChR was also achieved. These studies 1) provide further evidence for the existence of multiple subforms of α4β2 nAChR, 2) extend that to α3β2 nAChR, 3) demonstrate UTR influence on β2-containing nAChR properties, and 4) reveal compounds that interact with α4β2 in a subform-selective manner.


British Journal of Pharmacology | 2009

α7 nicotinic acetylcholine receptor agonist properties of tilorone and related tricyclic analogues

Clark A. Briggs; Michael R. Schrimpf; David J. Anderson; Earl J. Gubbins; Jens Halvard Grønlien; Monika Håkerud; Hilde Ween; Kirsten Thorin-Hagene; John Malysz; Jing Li; William H. Bunnelle; Murali Gopalakrishnan; Michael D. Meyer

The α7 nicotinic acetylcholine receptor (nAChR) has attracted considerable interest as a target for cognitive enhancement in schizophrenia and Alzheimers Disease. However, most recently described α7 agonists are derived from the quinuclidine structural class. Alternatively, the present study identifies tilorone as a novel α7‐selective agonist and characterizes analogues developed from this lead.


Cellular Immunology | 2002

TH1 and TH2 cytokine inhibition by 3,5-bis(trifluoromethyl)pyrazoles, a novel class of immunomodulators.

Yung-wu Chen; Morey L. Smith; Grace X Chiou; Stephen J. Ballaron; Michael P. Sheets; Earl J. Gubbins; Usha Warrior; Julie Wilkins; Carol Surowy; Masaki Nakane; George W. Carter; James M. Trevillyan; Karl W. Mollison; Stevan W. Djuric

In order to discover novel immunomodulators for application in treating autoimmune diseases, a stable Jurkat transfectant was constructed in which luciferase reporter gene is driven by a full-length IL-2 promotor. A chemical library was screened to identify compounds that inhibited luciferase expression in Jurkat transfectants stimulated with PMA and ionomycin. A class of compounds (bis-trifluoromethyl pyrazole, BTPs) was identified from this screen. BTPs were shown to inhibit anti-CD3 and anti-CD28 antibody-induced IL-2 secretion, mixed lymphocyte reaction, and Con A-induced T cell proliferation in normal human peripheral blood T cells. In addition, mRNA levels of IL-4, IL-5, IL-9, IL-10, IL-13, IL-15, and IFN-gamma were markedly inhibited by BTPs in peripheral blood mononuclear cells stimulated by Con A as determined by multi-probe RNA protection assay. Furthermore, IL-2, IL-4, IL-5, and IFN-gamma secretion by Hut 78 cells or CD3(+) T cells stimulated with PMA plus ionomycin or anti-CD3 antibody plus PMA were inhibited in a concentration-dependent manner by BTPs. Therefore, BTPs inhibit a wide spectrum of cytokine production including TH1 and TH2 type cytokines. Taken together, these compounds may be useful for treating autoimmune diseases and organ transplant rejection.


Analytical Biochemistry | 2003

A cell-based microarrayed compound screening format for identifying agonists of G-protein-coupled receptors.

Sujatha M. Gopalakrishnan; Robert B. Moreland; James L. Kofron; Rosalind Helfrich; Earl J. Gubbins; Jennifer McGowen; Jeffrey N. Masters; Diana L. Donnelly-Roberts; Jorge D. Brioni; David J. Burns; Usha Warrior

The identification of agonist and antagonist leads for G-protein-coupled receptors (GPCRs) is of critical importance to the pharmaceutical and biotechnology industries. We report on the utilization of a novel, high-density, well-less screening platform known as microarrayed compound screening microARCS) that tests 8640 compounds in the footprint of a standard microtiter plate for the identification of novel agonists for a specific G-protein-coupled receptor. Although receptors coupled to the G alpha(q) protein can readily be assessed by fluorescence-based Ca(2+) release measurements, many GPCRs that are coupled to G alpha(s) or G alpha(i/o) proteins are not amenable to functional evaluation in such a high-throughput manner. In this study, the human dopamine D(4.4) receptor, which normally couples through the G alpha(i/o) protein to inhibit adenylate cyclase and to reduce levels of intracellular cAMP, was coupled to intracellular Ca(2+) release by stably coexpressing this receptor with a chimeric G(alpha qo5) protein in HEK-293 cells. In microARCS format, the cells expressing D(4.4) receptor and G alpha(qo5) protein were preloaded with fluo-4, cast into a 1% agarose gel, placed above the compound sheets, and imaged successively using a ViewLux charge-coupled device imaging system. Dopamine and other agonists evoked an increase in fluorescence response that appeared as bright spots in a time- and concentration-dependent manner. Utilizing this technology, a library of 260,000 compounds was rapidly screened and led to the identification of several novel agonists. These agonists were further characterized using a fluorometric imaging plate reader assay. Excellent confirmation rates coupled with enhanced efficiency and throughput enable microARCS to serve as an alternative platform for the screening and identification of novel GPCR agonists.

Collaboration


Dive into the Earl J. Gubbins's collaboration.

Top Co-Authors

Avatar

Murali Gopalakrishnan

State University of New York System

View shared research outputs
Top Co-Authors

Avatar

John Malysz

University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Clark A. Briggs

Beckman Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James M. Trevillyan

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jay R. Luly

Millennium Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge