Network


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

Hotspot


Dive into the research topics where Larry C. James is active.

Publication


Featured researches published by Larry C. James.


Brain Research | 2003

Immunohistochemical localization of PDE10A in the rat brain

Thomas Francis Seeger; Brenda Bartlett; Timothy M. Coskran; Jeffrey S. Culp; Larry C. James; David L Krull; Jerry Lanfear; Anne M. Ryan; Christopher J. Schmidt; Christine A. Strick; Alison H. Varghese; Robert Williams; Patricia G Wylie; Frank S. Menniti

PDE10A is a newly identified cAMP/cGMP phosphodiesterase for which mRNA is highly expressed in the mammalian striatum. In the present study, PDE10A protein and mRNA expression throughout the rat brain were determined, using a monoclonal antibody (24F3.F11) for Western blot and immunohistochemical analyses and an antisense riboprobe for in situ hybridization. High levels of mRNA are observed in most of the neuronal cell bodies of striatal complex (caudate n, n. accumbens and olfactory tubercle), indicating that PDE10A is expressed by the striatal medium spiny neurons. PDE10A-like immunoreactivity is dense throughout the striatal neuropil, as well as in the internal capsule, globus pallidus, and substantia nigra. These latter regions lack significant expression of PDE10A mRNA. Thus, PDE10A is transported throughout the dendritic tree and down the axons to the terminals of the medium spiny neurons. These data suggest a role for PDE10A in regulating activity within both the striatonigral and striatopallidal pathways. In addition, PDE10A immunoreactivity and mRNA are found at lower levels in the hippocampal pyramidal cell layer, dentate granule cell layer and throughout the cortex and cerebellar granule cell layer. Immunoreactivity is detected only in cell bodies in these latter regions. This more restricted subcellular localization of PDE10A outside the striatum suggests a second, distinct function for the enzyme in these regions.


Neuropharmacology | 2006

Genetic deletion of the striatum-enriched phosphodiesterase PDE10A : Evidence for altered striatal function

Judith A. Siuciak; Sheryl A. McCarthy; Douglas S. Chapin; Remie A. Fujiwara; Larry C. James; Robert Williams; Jeffrey L. Stock; John D. McNeish; Christine A. Strick; Frank S. Menniti; Christopher J. Schmidt

PDE10A is a newly identified phosphodiesterase that is highly expressed by the medium spiny projection neurons of the striatum. In order to investigate the physiological role of PDE10A in the central nervous system, PDE10A knockout mice (PDE10A(-/-)) were characterized both behaviorally and neurochemically. PDE10A(-/-) mice showed decreased exploratory activity and a significant delay in the acquisition of conditioned avoidance behavior when compared to wild-type (PDE10A(+/+)) mice. However, in a variety of other well-characterized behavioral tasks, including the elevated plus maze (anxiety), forced swim test (depression), hot plate (nociception) and two memory models (passive avoidance and Morris water maze), PDE10A(-/-) mice performed similarly to wild-type mice. When challenged with PCP or MK-801, PDE10A(-/-) mice showed a blunted locomotor response in comparison to PDE10A(+/+) mice. In contrast, PDE10A(-/-) and PDE10A(+/+) mice responded similarly to the locomotor stimulating effects of amphetamine and methamphetamine. Our findings suggest that PDE10A is involved in regulating striatal output, possibly by reducing the sensitivity of medium spiny neurons to glutamatergic excitation. These results are discussed in relationship to the hypothesis that PDE10A inhibition presents a novel treatment for psychosis.


Journal of Medicinal Chemistry | 2009

Discovery, SAR, and Pharmacokinetics of a Novel 3-Hydroxyquinolin-2(1H)-one Series of Potent d-Amino Acid Oxidase (DAAO) Inhibitors†

Allen J. Duplantier; Stacey L. Becker; Michael John Bohanon; Kris A. Borzilleri; Boris A. Chrunyk; James T. Downs; Lain-Yen Hu; Ayman El-Kattan; Larry C. James; Shenping Liu; Jiemin Lu; Noha Maklad; Mahmoud N. Mansour; Scot Mente; Mary Piotrowski; Subas M. Sakya; Susan Sheehan; Stefanus J. Steyn; Christine A. Strick; Victoria A. Williams; Lei Zhang

3-Hydroxyquinolin-2(1H)-one (2) was discovered by high throughput screening in a functional assay to be a potent inhibitor of human DAAO, and its binding affinity was confirmed in a Biacore assay. Cocrystallization of 2 with the human DAAO enzyme defined the binding site and guided the design of new analogues. The SAR, pharmacokinetics, brain exposure, and effects on cerebellum D-serine are described. Subsequent evaluation against the rat DAAO enzyme revealed a divergent SAR versus the human enzyme and may explain the high exposures of drug necessary to achieve significant changes in rat or mouse cerebellum D-serine.


Neuropharmacology | 2010

Alterations in gene regulation following inhibition of the striatum-enriched phosphodiesterase, PDE10A

Christine A. Strick; Larry C. James; Carol B. Fox; Thomas Francis Seeger; Frank S. Menniti; Christopher J. Schmidt

PDE10A is a member of the phosphodiesterase superfamily highly enriched within medium spiny neurons (MSN) in mammalian striatum. We have used inhibitors of PDE10A and quantitative measures of mRNA to demonstrate that PDE10A controls striatal gene expression by regulating MSN cyclic nucleotide signaling pathways. Acute treatment with PDE10A inhibitors produces rapid and transient transcription of the immediate early gene cfos in rat striatum. Although inhibition of PDE10A causes accumulation of both cAMP and cGMP, the increase in striatal cfos expression appears to depend on changes in cAMP, since the increase is present in mice deficient in nNOS which fail to increase cGMP in response to PDE10A inhibition. Consistent with its expression in a majority of striatal MSN, PDE10A inhibition significantly induces expression of both substance P and enkephalin, neuropeptide markers for the direct and indirect striatal output pathways, respectively. These findings support the hypothesis that PDE10A modulates signal transduction in both striatal output pathways and suggest that PDE10A inhibitors may offer a unique approach to the treatment of schizophrenia.


Neuropharmacology | 2011

Modulation of NMDA receptor function by inhibition of d-amino acid oxidase in rodent brain

Christine A. Strick; Cheryl Li; Liam Scott; Brian Harvey; Mihály Hajós; Stefanus J. Steyn; Mary Piotrowski; Larry C. James; James T. Downs; Brian Rago; Stacey L. Becker; Ayman El-Kattan; Youfen Xu; Alan H. Ganong; F. David Tingley; Andres D. Ramirez; Patricia A. Seymour; Victor Guanowsky; Mark J. Majchrzak; Carol B. Fox; Christopher J. Schmidt; Allen J. Duplantier

Observations that N-Methyl-D-Aspartate (NMDA) antagonists produce symptoms in humans that are similar to those seen in schizophrenia have led to the current hypothesis that schizophrenia might result from NMDA receptor hypofunction. Inhibition of D-amino acid oxidase (DAAO), the enzyme responsible for degradation of D-serine, should lead to increased levels of this co-agonist at the NMDA receptor, and thereby provide a therapeutic approach to schizophrenia. We have profiled some of the preclinical biochemical, electrophysiological, and behavioral consequences of administering potent and selective inhibitors of DAAO to rodents to begin to test this hypothesis. Inhibition of DAAO activity resulted in a significant dose and time dependent increase in D-serine only in the cerebellum, although a time delay was observed between peak plasma or brain drug concentration and cerebellum D-serine response. Pharmacokinetic/pharmacodynamic (PK/PD) modeling employing a mechanism-based indirect response model was used to characterize the correlation between free brain drug concentration and D-serine accumulation. DAAO inhibitors had little or no activity in rodent models considered predictive for antipsychotic activity. The inhibitors did, however, affect cortical activity in the Mescaline-Induced Scratching model, produced a modest but significant increase in NMDA receptor-mediated synaptic currents in primary neuronal cultures from rat hippocampus, and resulted in a significant increase in evoked hippocampal theta rhythm, an in vivo electrophysiological model of hippocampal activity. These findings demonstrate that although DAAO inhibition did not cause a measurable increase in D-serine in forebrain, it did affect hippocampal and cortical activity, possibly through augmentation of NMDA receptor-mediated currents.


American Journal of Human Genetics | 2016

Biallelic Mutations in PDE10A Lead to Loss of Striatal PDE10A and a Hyperkinetic Movement Disorder with Onset in Infancy

Christine P. Diggle; Stacey J. Sukoff Rizzo; Michael Popiolek; Reetta Hinttala; Jan-Philip Schülke; Manju A. Kurian; Ian M. Carr; Alexander F. Markham; David T. Bonthron; Christopher M. Watson; Saghira Malik Sharif; Veronica Reinhart; Larry C. James; Michelle Vanase-Frawley; Erik Charych; Melanie Allen; John F. Harms; Christopher J. Schmidt; Joanne Ng; Karen Pysden; Christine A. Strick; Päivi Vieira; Katariina Mankinen; Hannaleena Kokkonen; Matti Kallioinen; Raija Sormunen; Juha O. Rinne; Jarkko Johansson; Kati Alakurtti; Laura Huilaja

Deficits in the basal ganglia pathways modulating cortical motor activity underlie both Parkinson disease (PD) and Huntington disease (HD). Phosphodiesterase 10A (PDE10A) is enriched in the striatum, and animal data suggest that it is a key regulator of this circuitry. Here, we report on germline PDE10A mutations in eight individuals from two families affected by a hyperkinetic movement disorder due to homozygous mutations c.320A>G (p.Tyr107Cys) and c.346G>C (p.Ala116Pro). Both mutations lead to a reduction in PDE10A levels in recombinant cellular systems, and critically, positron-emission-tomography (PET) studies with a specific PDE10A ligand confirmed that the p.Tyr107Cys variant also reduced striatal PDE10A levels in one of the affected individuals. A knock-in mouse model carrying the homologous p.Tyr97Cys variant had decreased striatal PDE10A and also displayed motor abnormalities. Striatal preparations from this animal had an impaired capacity to degrade cyclic adenosine monophosphate (cAMP) and a blunted pharmacological response to PDE10A inhibitors. These observations highlight the critical role of PDE10A in motor control across species.


ACS Medicinal Chemistry Letters | 2013

Structure-Based Design of Irreversible Human KAT II Inhibitors: Discovery of New Potency-Enhancing Interactions

Jamison B. Tuttle; Marie Anderson; Bruce M. Bechle; Brian M. Campbell; Cheng Chang; Amy B. Dounay; Edelweiss Evrard; Kari R. Fonseca; Xinmin Gan; Somraj Ghosh; Weldon Horner; Larry C. James; Ji-Young Kim; Laura A. McAllister; Jayvardhan Pandit; Vinod D. Parikh; Brian Rago; Michelle A. Salafia; Christine A. Strick; Laura E. Zawadzke; Patrick Robert Verhoest

A series of aryl hydroxamates recently have been disclosed as irreversible inhibitors of kynurenine amino transferase II (KAT II), an enzyme that may play a role in schizophrenia and other psychiatric and neurological disorders. The utilization of structure-activity relationships (SAR) in conjunction with X-ray crystallography led to the discovery of hydroxamate 4, a disubstituted analogue that has a significant potency enhancement due to a novel interaction with KAT II. The use of k inact/K i to assess potency was critical for understanding the SAR in this series and for identifying compounds with improved pharmacodynamic profiles.


FEBS Letters | 1991

Identification and characterization of a C-terminally extended form of recombinant murine IL-6

Dennis E. Danley; Christine A. Strick; Larry C. James; Anthony J. Lanzetti; Ivan G. Otterness; Hernan E. Grenett; Gerald M. Fuller

Murine interleukin‐6 (mIL‐6) was expressed in Escherichia coli in the insoluble fraction of cell lysates. Approximately equal amounts of two polypeptide species, reactive with anti‐IL‐6 antibodies, were produced. The two forms of mIL‐6 were isolated and found to have identical N‐terminal sequences initiated by Met‐Phe‐Pro‐Thr‐Ser‐Gin‐. Peptide mapping after endoproteinase glu‐C digestion led to isolation led to isolation and characterization of the C‐terminal peptides from each of the two forms and allowed the source of the heterogeneity to be identified as a C‐terminal addition of three amino acids, Gin‐Lys‐Leu, to authentic mIL‐6. Inspection of the nucleotide sequence of the plasmid containing the mIL‐6 gene and expression of the plasmid in other strains suggested that the addition of three amino acids was caused by a readthrough of the termination codon arising from an unexpected suppressor mutation in the original host strain. Although the C‐terminus of IL‐6 is critical for the activity of this cytokine, the IL‐6 variant with extended C‐terminus was fully active in two separate bioassays. This suggests that the additional amino acids do not disrupt the structure or function of this important region of the molecule.


Journal of Medicinal Chemistry | 2011

Discovery of Two Clinical Histamine H3 Receptor Antagonists: trans-N-Ethyl-3-fluoro-3-[3-fluoro-4-(pyrrolidinylmethyl)phenyl]cyclobutanecarboxamide (PF-03654746) and trans-3-Fluoro-3-[3-fluoro-4-(pyrrolidin-1-ylmethyl)phenyl]-N-(2-methylpropyl)cyclobutanecarboxamide (PF-03654764)

Travis T. Wager; Betty Pettersen; Anne W. Schmidt; Douglas K. Spracklin; Scot Mente; Todd William Butler; Harry Ralph Howard; Daniel J. Lettiere; David M. Rubitski; Diane F. Wong; Frank M. Nedza; Frederick R. Nelson; Hans Rollema; Jeffrey W. Raggon; Jody Freeman; John M. Marcek; Julie Cianfrogna; Karen W. Cook; Larry C. James; Linda A. Chatman; Philip A. Iredale; Michael J. Banker; Michael L. Homiski; Jennifer B. Munzner; Rama Y. Chandrasekaran


Archive | 1994

Multiple integrative vectors and yarrowia lipolytica transformants

Larry C. James; Christine A. Strick

Researchain Logo
Decentralizing Knowledge