Network


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

Hotspot


Dive into the research topics where Jonathan W. Dickerson is active.

Publication


Featured researches published by Jonathan W. Dickerson.


Journal of Pharmacology and Experimental Therapeutics | 2012

The Metabotropic Glutamate Receptor 4-Positive Allosteric Modulator VU0364770 Produces Efficacy Alone and in Combination with l-DOPA or an Adenosine 2A Antagonist in Preclinical Rodent Models of Parkinson's Disease

Carrie K. Jones; Michael Bubser; Analisa D. Thompson; Jonathan W. Dickerson; Nathalie Turle-Lorenzo; Marianne Amalric; Anna L. Blobaum; Thomas M. Bridges; Ryan D. Morrison; Satyawan Jadhav; Darren W. Engers; Kimberly Italiano; Jacob Bode; J. Scott Daniels; Craig W. Lindsley; Corey R. Hopkins; P. Jeffrey Conn; Colleen M. Niswender

Parkinsons disease (PD) is a debilitating neurodegenerative disorder associated with severe motor impairments caused by the loss of dopaminergic innervation of the striatum. Previous studies have demonstrated that positive allosteric modulators (PAMs) of metabotropic glutamate receptor 4 (mGlu4), including N-phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide, can produce antiparkinsonian-like effects in preclinical models of PD. However, these early mGlu4 PAMs exhibited unsuitable physiochemical properties for systemic dosing, requiring intracerebroventricular administration and limiting their broader utility as in vivo tools to further understand the role of mGlu4 in the modulation of basal ganglia function relevant to PD. In the present study, we describe the pharmacologic characterization of a systemically active mGlu4 PAM, N-(3-chlorophenyl)picolinamide (VU0364770), in several rodent PD models. VU0364770 showed efficacy alone or when administered in combination with l-DOPA or an adenosine 2A (A2A) receptor antagonist currently in clinical development (preladenant). When administered alone, VU0364770 exhibited efficacy in reversing haloperidol-induced catalepsy, forelimb asymmetry-induced by unilateral 6-hydroxydopamine (6-OHDA) lesions of the median forebrain bundle, and attentional deficits induced by bilateral 6-OHDA nigrostriatal lesions in rats. In addition, VU0364770 enhanced the efficacy of preladenant to reverse haloperidol-induced catalepsy when given in combination. The effects of VU0364770 to reverse forelimb asymmetry were also potentiated when the compound was coadministered with an inactive dose of l-DOPA, suggesting that mGlu4 PAMs may provide l-DOPA-sparing activity. The present findings provide exciting support for the potential role of selective mGlu4 PAMs as a novel approach for the symptomatic treatment of PD and a possible augmentation strategy with either l-DOPA or A2A antagonists.


Neuropsychopharmacology | 2016

Potentiation of M1 Muscarinic Receptor Reverses Plasticity Deficits and Negative and Cognitive Symptoms in a Schizophrenia Mouse Model.

Ayan Ghoshal; Jerri M. Rook; Jonathan W. Dickerson; G N Roop; Ryan D. Morrison; Nidhi Jalan-Sakrikar; Atin Lamsal; Meredith J. Noetzel; Mike Poslusney; Michael R. Wood; Bruce J. Melancon; Shaun R. Stauffer; Zixiu Xiang; Daniels Js; Colleen M. Niswender; Carrie K. Jones; Craig W. Lindsley; P.J. Conn

Schizophrenia patients exhibit deficits in signaling of the M1 subtype of muscarinic acetylcholine receptor (mAChR) in the prefrontal cortex (PFC) and also display impaired cortical long-term depression (LTD). We report that selective activation of the M1 mAChR subtype induces LTD in PFC and that this response is completely lost after repeated administration of phencyclidine (PCP), a mouse model of schizophrenia. Furthermore, discovery of a novel, systemically active M1 positive allosteric modulator (PAM), VU0453595, allowed us to evaluate the impact of selective potentiation of M1 on induction of LTD and behavioral deficits in PCP-treated mice. Interestingly, VU0453595 fully restored impaired LTD as well as deficits in cognitive function and social interaction in these mice. These results provide critical new insights into synaptic changes that may contribute to behavioral deficits in this mouse model and support a role for selective M1 PAMs as a novel approach for the treatment of schizophrenia.


Neuropharmacology | 2017

M1 muscarinic activation induces long-lasting increase in intrinsic excitability of striatal projection neurons

Xiaohui Lv; Jonathan W. Dickerson; Jerri M. Rook; Craig W. Lindsley; P. Jeffrey Conn; Zixiu Xiang

ABSTRACT The dorsolateral striatum is critically involved in movement control and motor learning. Striatal function is regulated by a variety of neuromodulators including acetylcholine. Previous studies have shown that cholinergic activation excites striatal principal projection neurons, medium spiny neurons (MSNs), and this action is mediated by muscarinic acetylcholine subtype 1 receptors (M1) through modulating multiple potassium channels. In the present study, we used electrophysiology techniques in conjunction with optogenetic and pharmacological tools to determine the long‐term effects of striatal cholinergic activation on MSN intrinsic excitability. A transient increase in acetylcholine release in the striatum by optogenetic stimulation resulted in a long‐lasting increase in excitability of MSNs, which was associated with hyperpolarizing shift of action potential threshold and decrease in afterhyperpolarization (AHP) amplitude, leading to an increase in probability of EPSP‐action potential coupling. The M1 selective antagonist VU0255035 prevented, while the M1 selective positive allosteric modulator (PAM) VU0453595 potentiated the cholinergic activation‐induced persistent increase in MSN intrinsic excitability, suggesting that M1 receptors are critically involved in the induction of this long‐lasting response. This M1 receptor‐dependent long‐lasting change in MSN intrinsic excitability could have significant impact on striatal processing and might provide a novel mechanism underlying cholinergic regulation of the striatum‐dependent motor learning and cognitive function. Consistent with this, behavioral studies indicate that potentiation of M1 receptor signaling by VU0453595 enhanced performance of mice in cue‐dependent water‐based T‐maze, a dorsolateral striatum‐dependent learning task. HIGHLIGHTSCholinergic activation induces persistent increase in excitability of dorsolateral striatal MSNs.It is associated with negative shift of AP threshold, decrease in AHP amplitude and increase in EPSP‐AP coupling.M1 mAChR antagonist VU0255035 prevents the persistent increase in MSN excitability.M1 PAM VU0453595 potentiates the persistent increase in MSN excitability.VU0453595 enhances performance of mice in cue‐dependent water‐based T‐maze.


ACS Chemical Neuroscience | 2017

Diverse Effects on M1 Signaling and Adverse Effect Liability within a Series of M1 Ago-PAMs

Jerri M. Rook; Masahito Abe; Hyekyung P. Cho; Kellie D. Nance; Vincent B. Luscombe; Jeffrey J. Adams; Jonathan W. Dickerson; Daniel H. Remke; Pedro M. Garcia-Barrantes; Darren W. Engers; Julie L. Engers; Sichen Chang; Jarrett J. Foster; Anna L. Blobaum; Colleen M. Niswender; Carrie K. Jones; P. Jeffrey Conn; Craig W. Lindsley

Both historical clinical and recent preclinical data suggest that the M1 muscarinic acetylcholine receptor is an exciting target for the treatment of Alzheimers disease and the cognitive and negative symptom clusters in schizophrenia; however, early drug discovery efforts targeting the orthosteric binding site have failed to afford selective M1 activation. Efforts then shifted to focus on selective activation of M1 via either allosteric agonists or positive allosteric modulators (PAMs). While M1 PAMs have robust efficacy in rodent models, some chemotypes can induce cholinergic adverse effects (AEs) that could limit their clinical utility. Here, we report studies aimed at understanding the subtle structural and pharmacological nuances that differentiate efficacy from adverse effect liability within an indole-based series of M1 ago-PAMs. Our data demonstrate that closely related M1 PAMs can display striking differences in their in vivo activities, especially their propensities to induce adverse effects. We report the discovery of a novel PAM in this series that is devoid of observable adverse effect liability. Interestingly, the molecular pharmacology profile of this novel PAM is similar to that of a representative M1 PAM that induces severe AEs. For instance, both compounds are potent ago-PAMs that demonstrate significant interaction with the orthosteric site (either bitopic or negative cooperativity). However, there are subtle differences in efficacies of the compounds at potentiating M1 responses, agonist potencies, and abilities to induce receptor internalization. While these differences may contribute to the differential in vivo profiles of these compounds, the in vitro differences are relatively subtle and highlight the complexities of allosteric modulators and the need to focus on in vivo phenotypic screening to identify safe and effective M1 PAMs.


Bioorganic & Medicinal Chemistry Letters | 2016

Optimization of the choline transporter (CHT) inhibitor ML352: Development of VU6001221, an improved in vivo tool compound

Jeanette L. Bertron; Elizabeth A. Ennis; Christopher Tarr; Jane Wright; Jonathan W. Dickerson; Charles W. Locuson; Anna L. Blobaum; Jerri M. Rook; Randy D. Blakely; Craig W. Lindsley

This Letter describes the further lead optimization of the CHT inhibitor probe, ML352 (VU0476201), and the development of VU6001221, an improved in vivo tool. A multi-dimensional optimization effort encountered steep SAR, and ultimately, subtle tuning of the electronics of the central phenyl core provided VU6001221, a CHT inhibitor with comparable potency for choline uptake inhibition as ML352, yet improved PK and CNS penetration. Moreover, VU6001221 enabled evaluation, for the first time, of a CHT inhibitor in a standard preclinical rodent cognition model, novel object recognition (NOR). We observed VU6001221 to elicit a dose-responsive increase in NOR, raising the possibility of agonism of synaptic α7 nicotinic ACh receptors by elevated extracellular choline, that if confirmed would represent a novel molecular strategy to enhance cognition.


Neuropsychopharmacology | 2017

Co-Activation of Metabotropic Glutamate Receptor 3 and Beta-Adrenergic Receptors Modulates Cyclic-AMP and Long-Term Potentiation, and Disrupts Memory Reconsolidation

Adam G. Walker; Douglas J. Sheffler; Andrew S Lewis; Jonathan W. Dickerson; Daniel J. Foster; Rebecca K. Senter; Mark S. Moehle; Xiaohui Lv; Branden J. Stansley; Zixiu Xiang; Jerri M. Rook; Kyle A. Emmitte; Craig W. Lindsley; P. Jeffrey Conn

Activation of β-adrenergic receptors (βARs) enhances both the induction of long-term potentiation (LTP) in hippocampal CA1 pyramidal cells and hippocampal-dependent cognitive function. Interestingly, previous studies reveal that coincident activation of group II metabotropic glutamate (mGlu) receptors with βARs in the hippocampal astrocytes induces a large increase in cyclic-AMP (cAMP) accumulation and release of adenosine. Adenosine then acts on A1 adenosine receptors at neighboring excitatory Schaffer collateral terminals, which could counteract effects of activation of neuronal βARs on excitatory transmission. On the basis of this, we postulated that activation of the specific mGlu receptor subtype that mediates this response could inhibit βAR-mediated effects on hippocampal synaptic plasticity and cognitive function. Using novel mGlu receptor subtype-selective allosteric modulators along with knockout mice we now report that the effects of mGlu2/3 agonists on βAR-mediated increases in cAMP accumulation are exclusively mediated by mGlu3. Furthermore, mGlu3 activation inhibits the ability of the βAR agonist isoproterenol to enhance hippocampal LTP, and this effect is absent in slices treated with either a glial toxin or an adenosine A1 receptor antagonist. Finally, systemic administration of the mGlu2/3 agonist LY379268 disrupted contextual fear memory in a manner similar to the effect of the βAR antagonist propranolol, and this effect was reversed by the mGlu3-negative allosteric modulator VU0650786. Taken together, these data suggest that mGlu3 can influence astrocytic signaling and modulate βAR-mediated effects on hippocampal synaptic plasticity and cognitive function.


Neuropsychopharmacology | 2018

M 1 -positive allosteric modulators lacking agonist activity provide the optimal profile for enhancing cognition

Sean P. Moran; Jonathan W. Dickerson; Hyekyung P. Cho; Zixiu Xiang; James Maksymetz; Daniel H. Remke; Xiaohui Lv; Catherine A. Doyle; Deepa H. Rajan; Colleen M. Niswender; Darren W. Engers; Craig W. Lindsley; Jerri M. Rook; P. Jeffrey Conn

Highly selective positive allosteric modulators (PAMs) of the M1 subtype of muscarinic acetylcholine receptor have emerged as an exciting new approach for improving cognitive function in patients suffering from Alzheimer’s disease and schizophrenia. However, excessive activation of M1 is known to induce seizure activity and have actions in the prefrontal cortex (PFC) that could impair cognitive function. We now report a series of pharmacological, electrophysiological, and behavioral studies in which we find that recently reported M1 PAMs, PF-06764427 and MK-7622, have robust agonist activity in cell lines and agonist effects in the mouse PFC, and have the potential to overactivate the M1 receptor and disrupt PFC function. In contrast, structurally distinct M1 PAMs (VU0453595 and VU0550164) are devoid of agonist activity in cell lines and maintain activity dependence of M1 activation in the PFC. Consistent with the previously reported effect of PF-06764427, the ago-PAM MK-7622 induces severe behavioral convulsions in mice. In contrast, VU0453595 does not induce behavioral convulsions at doses well above those required for maximal efficacy in enhancing cognitive function. Furthermore, in contrast to the robust efficacy of VU0453595, the ago-PAM MK-7622 failed to improve novel object recognition, a rodent assay of cognitive function. These findings suggest that in vivo cognition-enhancing efficacy of M1 PAMs can be observed with PAMs lacking intrinsic agonist activity and that intrinsic agonist activity of M1 PAMs may contribute to adverse effects and reduced efficacy in improving cognitive function.


ACS Medicinal Chemistry Letters | 2018

VU6007477, a novel M1 PAM based on a pyrrolo[2,3-b]pyridine carboxamide core devoid of cholinergic adverse events

Julie L. Engers; Elizabeth S Childress; Madeline F Long; Rory A Capstick; Vincent B. Luscombe; Hyekyung P. Cho; Jonathan W. Dickerson; Jerri M. Rook; Anna L. Blobaum; Colleen M. Niswender; Darren W. Engers; P. Jeffrey Conn; Craig W. Lindsley

Herein, we report the chemical optimization of a new series of M1 positive allosteric modulators (PAMs) based on a novel pyrrolo[2,3-b]pyridine core, developed via scaffold hopping and iterative parallel synthesis. The vast majority of analogs in this series proved to display robust cholinergic seizure activity. However, by removal of the secondary hydroxyl group, VU6007477 resulted with good rat M1 PAM potency (EC50 = 230 nM, 93% ACh max), minimal M1 agonist activity (agonist EC50 > 10 μM), good CNS penetration (rat brain/plasma K p = 0.28, K p,uu = 0.32; mouse K p = 0.16, K p,uu = 0.18), and no cholinergic adverse events (AEs, e.g., seizures). This work demonstrates that within a chemical series prone to robust M1 ago-PAM activity, SAR can result, which affords pure M1 PAMs, devoid of cholinergic toxicity/seizure liability.


ACS Chemical Neuroscience | 2018

A Novel M1 PAM VU0486846 Exerts Efficacy in Cognition Models without Displaying Agonist Activity or Cholinergic Toxicity

Jerri M. Rook; Jeanette L. Bertron; Hyekyung P. Cho; Pedro M. Garcia-Barrantes; Sean P. Moran; James Maksymetz; Kellie D. Nance; Jonathan W. Dickerson; Daniel H. Remke; Sichen Chang; Joel M. Harp; Anna L. Blobaum; Colleen M. Niswender; Carrie K. Jones; Shaun R. Stauffer; P. Jeffrey Conn; Craig W. Lindsley

Selective activation of the M1 subtype of muscarinic acetylcholine receptor, via positive allosteric modulation (PAM), is an exciting strategy to improve cognition in schizophrenia and Alzheimers disease patients. However, highly potent M1 ago-PAMs, such as MK-7622, PF-06764427, and PF-06827443, can engender excessive activation of M1, leading to agonist actions in the prefrontal cortex (PFC) that impair cognitive function, induce behavioral convulsions, and result in other classic cholinergic adverse events (AEs). Here, we report a fundamentally new and highly selective M1 PAM, VU0486846. VU0486846 possesses only weak agonist activity in M1-expressing cell lines with high receptor reserve and is devoid of agonist actions in the PFC, unlike previously reported ago-PAMs MK-7622, PF-06764427, and PF-06827443. Moreover, VU0486846 shows no interaction with antagonist binding at the orthosteric acetylcholine (ACh) site (e.g., neither bitopic nor displaying negative cooperativity with [3H]-NMS binding at the orthosteric site), no seizure liability at high brain exposures, and no cholinergic AEs. However, as opposed to ago-PAMs, VU0486846 produces robust efficacy in the novel object recognition model of cognitive function. Importantly, we show for the first time that an M1 PAM can reverse the cognitive deficits induced by atypical antipsychotics, such as risperidone. These findings further strengthen the argument that compounds with modest in vitro M1 PAM activity (EC50 > 100 nM) and pure-PAM activity in native tissues display robust procognitive efficacy without AEs mediated by excessive activation of M1. Overall, the combination of compound assessment with recombinant in vitro assays (mindful of receptor reserve), native tissue systems (PFC), and phenotypic screens (behavioral convulsions) is essential to fully understand and evaluate lead compounds and enhance success in clinical development.


Neuron | 2015

Biased mGlu5-Positive Allosteric Modulators Provide In Vivo Efficacy without Potentiating mGlu5 Modulation of NMDAR Currents.

Jerri M. Rook; Zixiu Xiang; Xiaohui Lv; Ayan Ghoshal; Jonathan W. Dickerson; Thomas M. Bridges; Kari A. Johnson; Daniel J. Foster; Karen J. Gregory; Paige N. Vinson; Analisa D. Thompson; Nellie Byun; Rebekah L. Collier; Michael Bubser; Michael T. Nedelcovych; Robert W. Gould; Shaun R. Stauffer; J. Scott Daniels; Colleen M. Niswender; Hilde Lavreysen; Claire Mackie; Susana Conde-Ceide; Jesús Alcázar; José Manuel Bartolomé-Nebreda; Gregor James Macdonald; John Talpos; Thomas Steckler; Carrie K. Jones; Craig W. Lindsley; P. Jeffrey Conn

Collaboration


Dive into the Jonathan W. Dickerson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge