Network


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

Hotspot


Dive into the research topics where Herbert H. Seltzman is active.

Publication


Featured researches published by Herbert H. Seltzman.


Annals of the New York Academy of Sciences | 2012

Designer drugs: a medicinal chemistry perspective

F. Ivy Carroll; Anita H. Lewin; S. Wayne Mascarella; Herbert H. Seltzman; P. Anantha Reddy

There are numerous medicinal chemistry reports in the literature describing the pharmacological properties of thousands of narcotics, stimulants, hallucinogens, sedative‐hypnotic drugs, cannabinoids, and other psychoactive substances as well as synthetic methods for their preparations. This information, while essential for the advancement of science, has been used by clandestine chemists to manufacture and market an endless variety of analogs of so‐called designer drugs. In this review, we describe how clandestine chemists used the principles of medicinal chemistry to design molecules, referred to as designer drugs, that elicit the effects of opioids, amphetamine and analogs, cannabinoids, and phencyclidine analogs while circumventing the law.


Molecular Pharmacology | 2010

Targeting of the Orphan Receptor GPR35 by Pamoic Acid: A Potent Activator of Extracellular Signal-Regulated Kinase and β-Arrestin2 with Antinociceptive Activity

Pingwei Zhao; Haleli Sharir; Ankur Kapur; Alan Cowan; Ellen B. Geller; Martin W. Adler; Herbert H. Seltzman; Patricia H. Reggio; Susanne Heynen-Genel; Michelle Sauer; Thomas Dy Chung; Yushi Bai; Wei Chen; Marc G. Caron; Larry S. Barak; Mary E. Abood

Known agonists of the orphan receptor GPR35 are kynurenic acid, zaprinast, 5-nitro-2-(3-phenylproplyamino) benzoic acid, and lysophosphatidic acids. Their relatively low affinities for GPR35 and prominent off-target effects at other pathways, however, diminish their utility for understanding GPR35 signaling and for identifying potential therapeutic uses of GPR35. In a screen of the Prestwick Library of drugs and drug-like compounds, we have found that pamoic acid is a potent GPR35 agonist. Pamoic acid is considered by the Food and Drug Administration as an inactive compound that enables long-acting formulations of numerous drugs, such as the antihelminthics oxantel pamoate and pyrantel pamoate; the psychoactive compounds hydroxyzine pamoate (Vistaril) and imipramine pamoate (Tofranil-PM); and the peptide hormones triptorelin pamoate (Trelstar) and octreotide pamoate (OncoLar). We have found that pamoic acid induces a Gi/o-linked, GPR35-mediated increase in the phosphorylation of extracellular signal-regulated kinase 1/2, recruitment of β-arrestin2 to GPR35, and internalization of GPR35. In mice, it attenuates visceral pain perception, indicating an antinociceptive effect, possibly through GPR35 receptors. We have also identified in collaboration with the Sanford-Burnham Institute Molecular Libraries Probe Production Center new classes of GPR35 antagonist compounds, including the nanomolar potency antagonist methyl-5-[(tert-butylcarbamothioylhydrazinylidene)methyl]-1-(2,4-difluorophenyl)pyrazole-4-carboxylate (CID2745687). Pamoic acid and potent antagonists such as CID2745687 present novel opportunities for expanding the chemical space of GPR35, elucidating GPR35 pharmacology, and stimulating GPR35-associated drug development. Our results indicate that the unexpected biological functions of pamoic acid may yield potential new uses for a common drug constituent.


Biochemistry | 2011

Identification of the GPR55 agonist binding site using a novel set of high-potency GPR55 selective ligands.

Evangelia Kotsikorou; Karla E. Madrigal; Dow P. Hurst; Haleli Sharir; Diane L. Lynch; Susanne Heynen-Genel; Loribelle Milan; Thomas Dy Chung; Herbert H. Seltzman; Yushi Bai; Marc G. Caron; Larry S. Barak; Mary E. Abood; Patricia H. Reggio

GPR55 is a class A G protein-coupled receptor (GPCR) that has been implicated in inflammatory pain, neuropathic pain, metabolic disorder, bone development, and cancer. Initially deorphanized as a cannabinoid receptor, GPR55 has been shown to be activated by non-cannabinoid ligands such as l-α-lysophosphatidylinositol (LPI). While there is a growing body of evidence of physiological and pathophysiological roles for GPR55, the paucity of specific antagonists has limited its study. In collaboration with the Molecular Libraries Probe Production Centers Network initiative, we identified a series of GPR55 antagonists using a β-arrestin, high-throughput, high-content screen of ~300000 compounds. This screen yielded novel, GPR55 antagonist chemotypes with IC50 values in the range of 0.16-2.72 μM [Heynen-Genel, S., et al. (2010) Screening for Selective Ligands for GPR55: Antagonists (ML191, ML192, ML193) (Bookshelf ID NBK66153; PMID entry 22091481)]. Importantly, many of the GPR55 antagonists were completely selective, with no agonism or antagonism against GPR35, CB1, or CB2 up to 20 μM. Using a model of the GPR55 inactive state, we studied the binding of an antagonist series that emerged from this screen. These studies suggest that GPR55 antagonists possess a head region that occupies a horizontal binding pocket extending into the extracellular loop region, a central ligand portion that fits vertically in the receptor binding pocket and terminates with a pendant aromatic or heterocyclic ring that juts out. Both the region that extends extracellularly and the pendant ring are features associated with antagonism. Taken together, our results provide a set of design rules for the development of second-generation GPR55 selective antagonists.


Journal of Medicinal Chemistry | 2010

Synthesis and Biological Evaluation of Bivalent Ligands for the Cannabinoid 1 Receptor

Yanan Zhang; Anne F. Gilliam; Rangan Maitra; M. Imad Damaj; Julianne Tajuba; Herbert H. Seltzman; Brian F. Thomas

Dimerization or oligomerization of many G-protein-coupled receptors (GPCRs), including the cannabinoid 1 (CB1) receptor, is now widely accepted and may have significant implications for medications development targeting these receptor complexes. A library of bivalent ligands composed of two identical CB1 antagonist pharmacophores derived from SR141716 linked by spacers of various lengths were developed. The affinities of these bivalent ligands at CB1 and CB2 receptors were determined using radiolabeled binding assays. Their functional activities were measured using GTP-γ-S accumulation and intracellular calcium mobilization assays. The results suggest that the nature of the linker and its length are crucial factors for optimum interactions of these ligands at CB1 receptor binding sites. Finally, selected bivalent ligands (5d and 7b) were able to attenuate the antinociceptive effects of the cannabinoid agonist CP55,940 (21) in a rodent tail-flick assay. These novel compounds may serve as probes that will enable further characterization of CB1 receptor dimerization and oligomerization and its functional significance and may prove useful in the development of new therapeutic approaches to G-protein-coupled receptor mediated disorders.


Journal of Biological Chemistry | 2014

Allosteric Modulation of a Cannabinoid G Protein-coupled Receptor BINDING SITE ELUCIDATION AND RELATIONSHIP TO G PROTEIN SIGNALING

Derek M. Shore; Gemma L. Baillie; Dow H. Hurst; Frank Navas; Herbert H. Seltzman; Jahan Marcu; Mary E. Abood; Ruth A. Ross; Patricia H. Reggio

Background: Cannabinoid-1 (CB1) receptor allosteric modulator ORG27569 increases CP55,940 binding, yet antagonizes G protein signaling. Results: ORG27569 binding sterically blocks movement in CB1 extracellular loops and transmembrane helix 6 (TMH6). Conclusion: ORG27569 increases CP55,940 binding by promoting an intermediate receptor conformation where changes important for signaling are blocked. Significance: This information may lead to the rational design of new allosteric modulators. The cannabinoid 1 (CB1) allosteric modulator, 5-chloro-3-ethyl-1H-indole-2-carboxylic acid [2-(4-piperidin-1-yl-phenyl)-ethyl]-amide) (ORG27569), has the paradoxical effect of increasing the equilibrium binding of [3H](−)-3-[2-hydroxyl-4-(1,1-dimethylheptyl)phenyl]-4-[3-hydroxylpropyl]cyclohexan-1-ol (CP55,940, an orthosteric agonist) while at the same time decreasing its efficacy (in G protein-mediated signaling). ORG27569 also decreases basal signaling, acting as an inverse agonist for the G protein-mediated signaling pathway. In ligand displacement assays, ORG27569 can displace the CB1 antagonist/inverse agonist, N-(piperidiny-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide(SR141716A). The goal of this work was to identify the binding site of ORG27569 at CB1. To this end, we used computation, synthesis, mutation, and functional studies to identify the ORG27569-binding site in the CB1 TMH3-6-7 region. This site is consistent with the results of K3.28192A, F3.36200A, W5.43279A, W6.48356A, and F3.25189A mutation studies, which revealed the ORG27569-binding site overlaps with our previously determined binding site of SR141716A but extends extracellularly. Additionally, we identified a key electrostatic interaction between the ORG27569 piperidine ring nitrogen and K3.28192 that is important for ORG27569 to act as an inverse agonist. At this allosteric site, ORG27569 promotes an intermediate conformation of the CB1 receptor, explaining ORG27569s ability to increase equilibrium binding of CP55,940. This site also explains ORG27569s ability to antagonize the efficacy of CP55,940 in three complementary ways. 1) ORG27569 sterically blocks movements of the second extracellular loop that have been linked to receptor activation. 2) ORG27569 sterically blocks a key electrostatic interaction between the third extracellular loop residue Lys-373 and D2.63176. 3) ORG27569 packs against TMH6, sterically hindering movements of this helix that have been shown to be important for receptor activation.


Journal of Medicinal Chemistry | 2012

Design and synthesis of cannabinoid receptor 1 antagonists for peripheral selectivity.

Alan Fulp; Katherine Bortoff; Herbert H. Seltzman; Yanan Zhang; James M. Mathews; Rodney W. Snyder; Timothy R. Fennell; Rangan Maitra

Antagonists of cannabinoid receptor 1 (CB1) have potential for the treatment of several diseases such as obesity, liver disease, and diabetes. Recently, development of several CB1 antagonists was halted because of adverse central nervous system (CNS) related side effects observed with rimonabant, the first clinically approved CB1 inverse agonist. However, recent studies indicate that regulation of peripherally expressed CB1 with CNS-sparing compounds is a viable strategy to treat several important disorders. Our efforts aimed at rationally designing peripherally restricted CB1 antagonists have resulted in compounds that have limited blood-brain barrier (BBB) permeability and CNS exposure in preclinical in vitro and in vivo models. Typically, compounds with high topological polar surface areas (TPSAs) do not cross the BBB passively. Compounds with TPSAs higher than that for rimonabant (rimonabant TPSA = 50) and excellent functional activity with limited CNS penetration were identified. These compounds will serve as templates for further optimization.


Cancer Letters | 2009

Synthesis and evaluation of Apogossypol Atropisomers as Potential Bcl-xL Antagonists

Jun Wei; Michele F. Rega; Shinichi Kitada; Hongbin Yuan; Dayong Zhai; Prabhakar Risbood; Herbert H. Seltzman; Charles Twine; John C. Reed; Maurizio Pellecchia

Anti-apoptotic Bcl-2 family proteins such as Bcl-2 and Bcl-X(L) have been recently validated as targets for the discovery of novel anti-cancer agents. We previously reported that racemic (+/-) Apogossypol, a semi-synthetic compound derived from the natural product Gossypol, binds and inhibits Bcl-2 and Bcl-X(L)in vitro and in cell. Given that (+) and (-) Gossypol display different proapoptotic activities, here we report on the synthesis of (+) and (-) Apogossypol and the evaluation of their in vitro and cellular activity.


Nuclear Medicine and Biology | 1996

Synthesis and in vivo studies of a selective ligand for the dopamine transporter: 3β-(4-[125I]iodophenyl) tropan-2β-carboxylic acid isopropyl ester ([125I]RTM-21)

John R. Lever; Ursula Scheffel; Marigo Stathis; Herbert H. Seltzman; Christopher D. Wyrick; Philip Abraham; Karol Parham; Brian F. Thomas; John W. Boja; Michael J. Kuhar; F. Ivy Carroll

Abstract A selective ligand for the dopamine transporter 3β-(4-iodophenyl)tropan-2β-carboxylic acid isopropyl ester (RTI-121) has been labeled with iodine-125 by electrophilic radioiododestannylation. The [ 125 I]RTI-121 was obtained in good yield (86 ± 7%, n = 3) with high radiochemical purity (>99%) and specific radioactivity (1210–1950 mCi/μmol). After i.v. administration of [ 125 I]RTI-121 to mice, the rank order of regional brain tissue radioactivity (striatum > olfactory tubercles ⪢> cortex, hippocampus, thalamus, hypothalamus, cerebellum) was consistent with dopamine transporter labeling. Specific in vivo binding in striatum and olfactory tubercles was saturable, and was blocked by the dopamine transporter ligands GBR 12,909 and (±)-nomifensine. By contrast, binding was not reduced by paroxetine, a serotonin transporter inhibitor, or desipramine, a norepinephrine transporter inhibitor. A variety of additional drugs having high affinities for recognition sites other than the neuronal dopamine transporter also had no effect. The [ 125 I]RTI-121 binding in striatum and olfactory tubercles was inhibited by d -amphetamine in dose-dependent fashion. Nonmetabolized radioligand represents 85% of the signal observed in extracts of whole mouse brain. Thus, [ 125 I]RTI-121 is readily prepared, and is a useful tracer for dopamine transporter studies in vivo .


Tetrahedron Letters | 1993

Nickel boride reduction of aryl nitro compounds

Herbert H. Seltzman; Bertold Berrang

Abstract Nickel boride smoothly reduces aryl nitro compounds to the corresponding anilines in the presence of iodo and ortho carboalkoxy groups in contrast to the problematic reductions by other methods.


Bioorganic & Medicinal Chemistry Letters | 2011

Towards rational design of cannabinoid receptor 1 (CB1) antagonists for peripheral selectivity.

Alan Fulp; Katherine Bortoff; Yanan Zhang; Herbert H. Seltzman; Rodney W. Snyder; Rangan Maitra

CB1 receptor antagonists that are peripherally restricted were targeted. Compounds with permanent charge as well as compounds that have increased polar surface area were made and tested against CB1 for binding and activity. Sulfonamide and sulfamide with high polar surface area and good activity at CB1 were rationally designed and pharmacologically tested. Further optimization of these compounds and testing could lead to the development of a new class of therapeutics to treat disorders where the CB1 receptor system has been implicated.

Collaboration


Dive into the Herbert H. Seltzman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patricia H. Reggio

University of North Carolina at Greensboro

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yanan Zhang

Research Triangle Park

View shared research outputs
Top Co-Authors

Avatar

Alan Fulp

Research Triangle Park

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge