Network


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

Hotspot


Dive into the research topics where A. Richard Chamberlin is active.

Publication


Featured researches published by A. Richard Chamberlin.


Bioorganic & Medicinal Chemistry | 1997

Inhibition of the Ser-Thr phosphatases PP1 and PP2A by naturally occurring toxins.

James E. Sheppeck; Carla-Maria Gauss; A. Richard Chamberlin

The okadaic acid class of naturally occurring toxins is a structurally diverse group of molecules that inhibit the protein phosphatases PP1 and PP2A. Studies providing information about the mode of binding between the toxins and the phosphatases contribute to an overall understanding of the signal transduction pathways in which the phosphatases are involved.


Journal of Pharmacology and Experimental Therapeutics | 2008

Synthesis and Pharmacological in Vitro and in Vivo Profile of 3-Oxo-1,1-diphenyl-tetrahydro-oxazolo[3,4-a]pyrazine-7-carboxylic Acid 4-Fluoro-benzylamide (SHA 68), a Selective Antagonist of the Neuropeptide S Receptor

Naoe Okamura; Stephen A. Habay; Joanne Zeng; A. Richard Chamberlin; Rainer K. Reinscheid

Neuropeptide S (NPS) has been shown to modulate arousal, sleep wakefulness, anxiety-like behavior, and feeding after central administration of the peptide agonist to mice or rats. We report here the chemical synthesis and pharmacological characterization of SHA 66 (3-oxo-1,1-diphenyl-tetrahydro-oxazolo[3,4-a]pyrazine-7-carboxylic acid benzylamide) and SHA 68 (3-oxo-1,1-diphenyl-tetrahydro-oxazolo[3,4-a]pyrazine-7-carboxylic acid 4-fluoro-benzylamide), two closely related bicyclic piperazines with antagonistic properties at the NPS receptor (NPSR). The compounds block NPS-induced Ca2+ mobilization, and SHA 68 shows displaceable binding to NPSR in the nanomolar range. The antagonistic activity of SHA 68 seems to be specific because it does not affect signaling at 14 unrelated G protein-coupled receptors. Analysis of pharmacokinetic parameters of SHA 68 demonstrates that the compound reaches pharmacologically relevant levels in plasma and brain after i.p. administration. Furthermore, peripheral administration of SHA 68 in mice (50 mg/kg i.p.) is able to antagonize NPS-induced horizontal and vertical activity as well as stereotypic behavior. Therefore, SHA 68 could be a useful tool to characterize physiological functions and pharmacological parameters of the NPS system in vitro and in vivo.


Journal of Neurochemistry | 2002

Differing effects of substrate and non-substrate transport inhibitors on glutamate uptake reversal

Christopher M. Anderson; Richard J. Bridges; A. Richard Chamberlin; Keiko Shimamoto; Yoshimi Yasuda-Kamatani; Raymond A. Swanson

Na+‐dependent excitatory amino acid transporters (EAATs) normally function to remove extracellular glutamate from brain extracellular space, but EAATs can also increase extracellular glutamate by reversal of uptake. Effects of inhibitors on EAATs can be complex, depending on cell type, whether conditions favor glutamate uptake or uptake reversal and whether the inhibitor itself is a substrate for the transporters. The present study assessed EAAT inhibitors for their ability to inhibit glutamate uptake, act as transporter substrates and block uptake reversal in astrocyte and neuron cultures. lthreo‐β‐hydroxyaspartate (l‐TBHA), dlthreo‐β‐benzyloxyaspartate (dl‐TBOA), ltrans‐pyrrolidine‐2,4‐dicarboxylic acid (ltrans‐2,4‐PDC) (+/–)‐cis‐4‐methy‐trans‐pyrrolidine‐2,4‐dicarboxylic acid (cis‐4‐methy‐trans‐2,4‐PDC) and lantiendo‐3,4‐methanopyrrolidine‐2,4‐dicarboxylic acid (lantiendo‐3,4‐MPDC) inhibited l‐[14C]glutamate uptake in astrocytes with equilibrium binding constants ranging from 17 µm (dl‐TBOA and l‐TBHA) – 43 µm (cis‐4‐methy‐trans‐2,4‐PDC). Transportability of inhibitors was assessed in astrocytes and neurons. While l‐TBHA, ltrans‐2,4‐PDC, cis‐4‐methy‐trans‐2,4‐PDC and lantiendo‐3,4‐MPDC displayed significant transporter substrate activities in neurons and astrocytes, dl‐TBOA was a substrate only in astrocytes. This effect of dl‐TBOA was concentration‐dependent, leading to complex effects on glutamate uptake reversal. At concentrations low enough to produce minimal dl‐TBOA uptake velocity (≤ 10 µm), dl‐TBOA blocked uptake reversal in ATP‐depleted astrocytes; this blockade was negated at concentrations that drove substantial dl‐TBOA uptake (> 10 µm). These findings indicate that the net effects of EAAT inhibitors can vary with cell type and exposure conditions.


Brain Research | 1983

Synthesis of [3H]2-Amino-4-phosphonobutyric acid and characterization of its binding to rat brain membranes: a selective ligand for the chloride/calcium-dependent class ofl-glutamate binding sites

Daniel T. Monaghan; M.C. McMills; A. Richard Chamberlin; Carl W. Cotman

[3H]2-amino-4-phosphonobutyric acid was synthesized by the conjugate addition of 1-lithio-2-trimethylsilyethyne to diethyl ethynylphosphate followed by catalytic tritiation and hydrolysis. Radiolabelled 2-amino-4-phosphonobutyric acid binds to a distinct class of L-glutamate binding sites and does not exhibit appreciable binding to sites not displaced by L-glutamate. The binding affinity (Kd = 5.1 +/- 0.4 microM) and pharmacological profile correspond to those values obtained from physiological studies of 2-amino-4-phosphonobutyric acid inhibition of synaptic transmission, and to those values obtained in [3H]L-glutamate binding assays. [3H]2-amino-4-phosphonobutyric acid does not exhibit significant binding to the Cl-/Ca2+-independent L-glutamate binding site(s), nor to the Na+-dependent L-glutamate binding site (up to 50 mM Na+). These data provide further evidence that the physiological action of 2-amino-4-phosphonobutyric acid is mediated by the previously described Cl-/Ca2+-dependent L-glutamate binding sites, and provides an assay system which is optimal for the study of these sites.


ChemBioChem | 2006

A central strategy for converting natural products into fluorescent probes

Matthew D. Alexander; Michael D. Burkart; Michael S. Leonard; Padma Portonovo; Bo Liang; Xiaobin Ding; Madeleine M. Joullié; Brian M. Gulledge; James Aggen; A. Richard Chamberlin; Joel S. Sandler; William Fenical; Jian Cui; Santosh J. Gharpure; Alexei Polosukhin; Hai Ren Zhang; P. Andrew Evans; Adam D. Richardson; Mary Kay Harper; Chris M. Ireland; Binh G. Vong; Thomas P. Brady; Emmanuel A. Theodorakis; James J. La Clair

A Central Strategy for Converting Natural Products into Fluorescent Probes Matthew D. Alexander, Michael D. Burkart, Michael S. Leonard, Padma Portonovo, Bo Liang, Xiaobin Ding, Madeleine M. Joulli!, Brian M. Gulledge, James B. Aggen, A. Richard Chamberlin, Joel Sandler, William Fenical, Jian Cui, Santosh J. Gharpure, Alexei Polosukhin, Hai-Ren Zhang, P. Andrew Evans, Adam D. Richardson, Mary Kay Harper, Chris M. Ireland, Binh G. Vong, Thomas P. Brady, Emmanuel A. Theodorakis, and James J. La Clair*


Journal of Medicinal Chemistry | 2009

Synthesis and biological evaluation of a series of novel inhibitor of Nek2/Hec1 analogues.

Xiao-Long Qiu; Guideng Li; Guikai Wu; Jiewen Zhu; Longen Zhou; Phang Lang Chen; A. Richard Chamberlin; Wen-Hwa Lee

High expression in cancer 1 (Hec1) is an oncogene overly expressed in many human cancers. Small molecule inhibitor of Nek2/Hec1 (INH) targeting the Hec1 and its regulator, Nek2, in the mitotic pathway, was identified to inactivate Hec1/Nek2 function mediated by protein degradation that subsequently leads to chromosome mis-segregation and cell death. To further improve the efficacy of INH, a series of INH analogues were designed, synthesized, and evaluated. Among these 33 newly synthesized analogues, three of them, 6, 13, and 21, have 6-8 fold more potent cell killing activity than the previous lead compound INH1. Compounds 6 and 21 were chosen for analyzing the underlying action mechanism. They target directly the Hec1/Nek2 pathway and cause chromosome mis-alignment as well as cell death, a mechanism similar to that of INH1. This initial exploration of structural/functional relationship of INH may advance the progress for developing clinically applicable INH analogue.


Tetrahedron | 1991

Site-specific incorporation of non-natural residues into peptides: Effect of residue structure on suppression and translation efficiencies

J. D. Bain; Dean A. Wacker; Eric E. Kuo; A. Richard Chamberlin

Abstract A systematic survey of the structural requirements for biosynthetic incorporation of non-natural residues into a polypeptide is presented. Relative translation efficiencies for a series of 12 semi-synthetic acylated suppressor tRNAs ranged from 0 to 91% depending on the structure of the residue incorporated. A systematic survey of the structural requirements for biosynthetic incorporation of non-natural residues into a polypeptide is presented. Relative translation efficiencies for a series of 12 semi-synthetic acylated suppressor tRNAs ranged from 0 to 91% depending on the structure of the residue incorporated.


Neuropharmacology | 2005

The substituted aspartate analogue L-β-threo-benzyl-aspartate preferentially inhibits the neuronal excitatory amino acid transporter EAAT3

C. Sean Esslinger; Shailesh Agarwal; John M. Gerdes; Paul A. Wilson; Erin S. Davis; Alicia N. Awes; Erin O'brien; Teri Mavencamp; Hans P. Koch; David J. Poulsen; Joseph F. Rhoderick; A. Richard Chamberlin; Michael P. Kavanaugh; Richard J. Bridges

The excitatory amino acid transporters (EAATs) play key roles in the regulation of CNS L-glutamate, especially related to synthesis, signal termination, synaptic spillover, and excitotoxic protection. Inhibitors available to delineate EAAT pharmacology and function are essentially limited to those that non-selectively block all EAATs or those that exhibit a substantial preference for EAAT2. Thus, it is difficult to selectively study the other subtypes, particularly EAAT1 and EAAT3. Structure activity studies on a series of beta-substituted aspartate analogues identify L-beta-benzyl-aspartate (L-beta-BA) as among the first blockers that potently and preferentially inhibits the neuronal EAAT3 subtype. Kinetic analysis of D-[(3)H]aspartate uptake into C17.2 cells expressing the hEAATs demonstrate that L-beta-threo-BA is the more potent diastereomer, acts competitively, and exhibits a 10-fold preference for EAAT3 compared to EAAT1 and EAAT2. Electrophysiological recordings of EAAT-mediated currents in Xenopus oocytes identify L-beta-BA as a non-substrate inhibitor. Analyzing L-beta-threo-BA within the context of a novel EAAT2 pharmacophore model suggests: (1) a highly conserved positioning of the electrostatic carboxyl and amino groups; (2) nearby regions that accommodate select structural modifications (cyclopropyl rings, methyl groups, oxygen atoms); and (3) a unique region L-beta-threo-BA occupied by the benzyl moieties of L-TBOA, L-beta-threo-BA and related analogues. It is plausible that the preference of L-beta-threo-BA and L-TBOA for EAAT3 and EAAT2, respectively, could reside in the latter two pharmacophore regions.


Biophysical Journal | 2011

Acyl-Chain Methyl Distributions of Liquid-Ordered and -Disordered Membranes

Mihaela Mihailescu; Rishi G. Vaswani; Eduardo Jardón-Valadez; Francisco Castro-Roman; J. Alfredo Freites; David L. Worcester; A. Richard Chamberlin; Douglas J. Tobias; Stephen H. White

A central feature of the lipid raft concept is the formation of cholesterol-rich lipid domains. The introduction of relatively rigid cholesterol molecules into fluid liquid-disordered (L(d)) phospholipid bilayers can produce liquid-ordered (L(o)) mixtures in which the rigidity of cholesterol causes partial ordering of the flexible hydrocarbon acyl chains of the phospholipids. Several lines of evidence support this concept, but direct structural information about L(o) membranes is lacking. Here we present the structure of L(o) membranes formed from cholesterol and dioleoylphosphatidylcholine (DOPC). Specific deuteration of the DOPC acyl-chain methyl groups and neutron diffraction measurements reveal an extraordinary disorder of the acyl chains of neat L(d) DOPC bilayers. The disorder is so great that >20% of the methyl groups are in intimate contact with water in the bilayer interface. The ordering of the DOPC acyl chains by cholesterol leads to retraction of the methyl groups away from the interface. Molecular dynamics simulations based on experimental systems reveal asymmetric transbilayer distributions of the methyl groups associated with each bilayer leaflet.


Bioorganic & Medicinal Chemistry Letters | 1993

Conformationally restricted inhibitors of the high affinity L-glutamate transporter

Richard J. Bridges; Frank Lovering; John M. Humphrey; Mark S. Stanley; Tracy N. Blakely; Michael F. Cristofaro; A. Richard Chamberlin

Abstract A series of acidic amino acids has been prepared and evaluated in an effort to identify the structural features required for binding to and inhibiting the high affinity uptake system that clears L-glutamate from the synaptic cleft during excitatory amino acid-mediated neurotransmission in the mammalian CNS.

Collaboration


Dive into the A. Richard Chamberlin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James Aggen

University of California

View shared research outputs
Top Co-Authors

Avatar

Jiewen Zhu

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

F. Thomas Bond

University of California

View shared research outputs
Top Co-Authors

Avatar

Milana Dezube

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge